1
|
Pillaiyar T, Laufer S. A patent review of CXCR7 modulators (2019-present). Expert Opin Ther Pat 2025:1-27. [PMID: 40122070 DOI: 10.1080/13543776.2025.2477475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/13/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Atypical chemokine receptor 3 (ACKR3) (formerly CXCR7) regulates various biological processes through its ligands and is closely associated with numerous diseases, including inflammation, cancer, cardiovascular diseases (CVDs), pain, and neurological disorders. Therefore, ACKR3 has emerged as a potential target for disease treatment. AREAS COVERED This review summarizes the ACKR3 modulators published in patents from 2019 to 2024 using data from Google Patents, the European Patent Office, and the World Intellectual Property Organization's online databases. This includes information on their chemical structures, syntheses, activities, and developmental stages. EXPERT OPINION ACKR3 agonists gained traction as a treatment for cardiovascular and pain conditions. WW-12, which was derived from the chemical modifications of conolidine, became a novel small-molecule pain modulator by activating ACKR3, which in turn boosted endogenous opioid peptides for the classical opioid receptors.ACKR3 antagonist ACT-1004-1239 from Idorsia Pharmaceuticals Ltd. has demonstrated the ability to treat cancer, acute lung injury/ARDS, and autoimmune diseases, including multiple sclerosis. The outcomes of these clinical trials will direct the development and indications of future ACKR3 modulators.
Collapse
Affiliation(s)
- Thanigaimalai Pillaiyar
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Tübingen, Germany
- Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Eberhard Karls University Tübingen, Tübingen, Germany
| | - Stefan Laufer
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Tübingen, Germany
- Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Eberhard Karls University Tübingen, Tübingen, Germany
- iFIT Cluster of Excellence (EXC 2180) "Image-guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, Tübingen, Germany
| |
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
|
Melgrati S, Radice E, Ameti R, Hub E, Thelen S, Pelczar P, Jarrossay D, Rot A, Thelen M. Atlas of the anatomical localization of atypical chemokine receptors in healthy mice. PLoS Biol 2023; 21:e3002111. [PMID: 37159457 PMCID: PMC10198502 DOI: 10.1371/journal.pbio.3002111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/19/2023] [Accepted: 04/05/2023] [Indexed: 05/11/2023] Open
Abstract
Atypical chemokine receptors (ACKRs) scavenge chemokines and can contribute to gradient formation by binding, internalizing, and delivering chemokines for lysosomal degradation. ACKRs do not couple to G-proteins and fail to induce typical signaling induced by chemokine receptors. ACKR3, which binds and scavenges CXCL12 and CXCL11, is known to be expressed in vascular endothelium, where it has immediate access to circulating chemokines. ACKR4, which binds and scavenges CCL19, CCL20, CCL21, CCL22, and CCL25, has also been detected in lymphatic and blood vessels of secondary lymphoid organs, where it clears chemokines to facilitate cell migration. Recently, GPR182, a novel ACKR-like scavenger receptor, has been identified and partially deorphanized. Multiple studies point towards the potential coexpression of these 3 ACKRs, which all interact with homeostatic chemokines, in defined cellular microenvironments of several organs. However, an extensive map of ACKR3, ACKR4, and GPR182 expression in mice has been missing. In order to reliably detect ACKR expression and coexpression, in the absence of specific anti-ACKR antibodies, we generated fluorescent reporter mice, ACKR3GFP/+, ACKR4GFP/+, GPR182mCherry/+, and engineered fluorescently labeled ACKR-selective chimeric chemokines for in vivo uptake. Our study on young healthy mice revealed unique and common expression patterns of ACKRs in primary and secondary lymphoid organs, small intestine, colon, liver, and kidney. Furthermore, using chimeric chemokines, we were able to detect distinct zonal expression and activity of ACKR4 and GPR182 in the liver, which suggests their cooperative relationship. This study provides a broad comparative view and a solid stepping stone for future functional explorations of ACKRs based on the microanatomical localization and distinct and cooperative roles of these powerful chemokine scavengers.
Collapse
Affiliation(s)
- Serena Melgrati
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Egle Radice
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Rafet Ameti
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Elin Hub
- Centre for Microvascular Research, The William Harvey Research Institute, Queen Mary University London, London, United Kingdom
| | - Sylvia Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Pawel Pelczar
- University of Basel, Center for Transgenic Models, Basel, Switzerland
| | - David Jarrossay
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Antal Rot
- Centre for Microvascular Research, The William Harvey Research Institute, Queen Mary University London, London, United Kingdom
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University London, London, United Kingdom
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University, Munich, Germany
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| |
Collapse
|
4
|
Radice E, Ameti R, Melgrati S, Foglierini M, Antonello P, Stahl RAK, Thelen S, Jarrossay D, Thelen M. Marginal Zone Formation Requires ACKR3 Expression on B Cells. Cell Rep 2021; 32:107951. [PMID: 32755592 DOI: 10.1016/j.celrep.2020.107951] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 05/12/2020] [Accepted: 07/02/2020] [Indexed: 12/27/2022] Open
Abstract
The marginal zone (MZ) contributes to the highly organized spleen microarchitecture. We show that expression of atypical chemokine receptor 3 (ACKR3) defines two equal-sized populations of mouse MZ B cells (MZBs). ACKR3 is required for development of a functional MZ and for positioning of MZBs. Deletion of ACKR3 on B cells distorts the MZ, and MZBs fail to deliver antigens to follicles, reducing humoral responses. Reconstitution of MZ-deficient CD19ko mice shows that ACKR3- MZBs can differentiate into ACKR3+ MZBs, but not vice versa. The lack of a MZ is rescued by adoptive transfer of ACKR3-sufficient, and less by ACKR3-deficient, follicular B cells (FoBs); hence, ACKR3 expression is crucial for establishment of the MZ. The inability of CD19ko mice to respond to T-independent antigen is rescued when ACKR3-proficient, but not ACKR3-deficient, FoBs are transferred. Accordingly, ACKR3-deficient FoBs are able to reconstitute the MZ if the niche is pre-established by ACKR3-proficient MZBs.
Collapse
Affiliation(s)
- Egle Radice
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, 6500 Bellinzona, Switzerland; Graduate School of Cellular and Molecular Sciences, University of Bern, 3012 Bern, Switzerland
| | - Rafet Ameti
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, 6500 Bellinzona, Switzerland; Graduate School of Cellular and Molecular Sciences, University of Bern, 3012 Bern, Switzerland
| | - Serena Melgrati
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, 6500 Bellinzona, Switzerland; Graduate School of Cellular and Molecular Sciences, University of Bern, 3012 Bern, Switzerland
| | - Mathilde Foglierini
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, 6500 Bellinzona, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Paola Antonello
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, 6500 Bellinzona, Switzerland; Graduate School of Cellular and Molecular Sciences, University of Bern, 3012 Bern, Switzerland
| | - Rolf A K Stahl
- III Medizinische Klinik, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Sylvia Thelen
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, 6500 Bellinzona, Switzerland
| | - David Jarrossay
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, 6500 Bellinzona, Switzerland
| | - Marcus Thelen
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, 6500 Bellinzona, Switzerland.
| |
Collapse
|
5
|
Jensen P, Patel B, Smith S, Sabnis R, Kaboord B. Improved Immunoprecipitation to Mass Spectrometry Method for the Enrichment of Low-Abundant Protein Targets. Methods Mol Biol 2021; 2261:229-246. [PMID: 33420993 DOI: 10.1007/978-1-0716-1186-9_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Immunoprecipitation (IP) is commonly used upstream of mass spectrometry (MS) as an enrichment tool for low-abundant protein targets. However, several aspects of the classical IP procedure such as nonspecific protein binding to the isolation matrix, detergents or high salt concentrations in wash and elution buffers, and antibody chain contamination in elution fractions render it incompatible with downstream mass spectrometry analysis. Here, we discuss an improved IP-MS workflow that is designed to minimize sample prep time and these contaminants. The method employs biotinylated antibodies to the targets of interest and streptavidin magnetic beads that exhibit low background binding. In addition, alterations in the elution protocol and subsequent MS sample prep were made to reduce time and antibody leaching in the eluent, minimizing potential ion suppression effects and thereby maximizing detection of multiple target antigens and interacting proteins.
Collapse
Affiliation(s)
| | | | | | - Renuka Sabnis
- Nisarga Biotech Pvt. Ltd., Satara, Maharashtra, India
| | | |
Collapse
|
6
|
Matti C, D'Uonnolo G, Artinger M, Melgrati S, Salnikov A, Thelen S, Purvanov V, Strobel TD, Spannagel L, Thelen M, Legler DF. CCL20 is a novel ligand for the scavenging atypical chemokine receptor 4. J Leukoc Biol 2020; 107:1137-1154. [PMID: 32533638 DOI: 10.1002/jlb.2ma0420-295rrr] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022] Open
Abstract
The chemokine CCL20 is broadly produced by endothelial cells in the liver, the lung, in lymph nodes and mucosal lymphoid tissues, and recruits CCR6 expressing leukocytes, particularly dendritic cells, mature B cells, and subpopulations of T cells. How CCL20 is systemically scavenged is currently unknown. Here, we identify that fluorescently labeled human and mouse CCL20 are efficiently taken-up by the atypical chemokine receptor ACKR4. CCL20 shares ACKR4 with the homeostatic chemokines CCL19, CCL21, and CCL25, although with a lower affinity. We demonstrate that all 4 human chemokines recruit β-arrestin1 and β-arrestin2 to human ACKR4. Similarly, mouse CCL19, CCL21, and CCL25 equally activate the human receptor. Interestingly, at the same chemokine concentration, mouse CCL20 did not recruit β-arrestins to human ACKR4. Further cross-species analysis suggests that human ACKR4 preferentially takes-up human CCL20, whereas mouse ACKR4 similarly internalizes mouse and human CCL20. Furthermore, we engineered a fluorescently labeled chimeric chemokine consisting of the N-terminus of mouse CCL25 and the body of mouse CCL19, termed CCL25_19, which interacts with and is taken-up by human and mouse ACKR4.
Collapse
Affiliation(s)
- Christoph Matti
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland
| | - Giulia D'Uonnolo
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Marc Artinger
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland
| | - Serena Melgrati
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Angela Salnikov
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland
| | - Sylvia Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Vladimir Purvanov
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland
| | - Tobias D Strobel
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland
| | - Lisa Spannagel
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland.,Faculty of Biology, University of Konstanz, Konstanz, Germany.,Theodor Kocher Institute, University of Bern, Bern, Switzerland
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Stüber JC, Plückthun A. Labeling surface proteins with high specificity: Intrinsic limitations of phosphopantetheinyl transferase systems. PLoS One 2019; 14:e0226579. [PMID: 31856184 PMCID: PMC6922365 DOI: 10.1371/journal.pone.0226579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 11/28/2019] [Indexed: 12/04/2022] Open
Abstract
Objective Fluorescent labeling of specific cell-surface proteins enables a manifold of techniques to study their function in health and disease. A frequently cited family of methods employs phosphopantetheinyl transferases (PPTases) to attach probes, provided as conjugates of Coenzyme A. This method appears attractive, as only short peptide tags genetically fused to the protein of interest are needed as conjugation sites. Here, we describe observations we made when evaluating such protocols for delicate single-molecule applications where we require a particular combination of dyes, low background binding or low labeling of other proteins, and a high degree of labeling. Results When we tested a PPTase-acceptor peptide couple with several experimental protocols and various CoA conjugates for labeling of a protein on the cell surface, we noticed substantial non-specific labeling. For the first time, we provide here a quantification of the non-specific fraction of the signals obtained using appropriate controls. We further present evidence that this background is due to CoA-dye conjugates entering the cell, where they may be covalently attached to endogenous proteins. However, when studying cell-surface proteins, most fluorescent readouts require that labeling is strictly limited to the protein of interest located at the cell surface. While such data have so far been missing in the literature, they suggest that for applications where labeling of unwanted molecules would affect the conclusions, researchers need to be aware of this potential non-specificity of PPTase methods when selecting a labeling strategy. We show, again by quantitative comparison, that the HaloTag is a viable alternative.
Collapse
Affiliation(s)
- Jakob C. Stüber
- Department of Biochemistry, University of Zurich, Winterthurerstrasse, Zurich, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstrasse, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
10
|
Adlere I, Caspar B, Arimont M, Dekkers S, Visser K, Stuijt J, de Graaf C, Stocks M, Kellam B, Briddon S, Wijtmans M, de Esch I, Hill S, Leurs R. Modulators of CXCR4 and CXCR7/ACKR3 Function. Mol Pharmacol 2019; 96:737-752. [PMID: 31548340 DOI: 10.1124/mol.119.117663] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/14/2019] [Indexed: 02/14/2025] Open
Abstract
The two G protein-coupled receptors (GPCRs) C-X-C chemokine receptor type 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3) are part of the class A chemokine GPCR family and represent important drug targets for human immunodeficiency virus (HIV) infection, cancer, and inflammation diseases. CXCR4 is one of only three chemokine receptors with a US Food and Drug Administration approved therapeutic agent, the small-molecule modulator AMD3100. In this review, known modulators of the two receptors are discussed in detail. Initially, the structural relationship between receptors and ligands is reviewed on the basis of common structural motifs and available crystal structures. To date, no atypical chemokine receptor has been crystallized, which makes ligand design and predictions for these receptors more difficult. Next, the selectivity, receptor activation, and the resulting ligand-induced signaling output of chemokines and other peptide ligands are reviewed. Binding of pepducins, a class of lipid-peptides whose basis is the internal loop of a GPCR, to CXCR4 is also discussed. Finally, small-molecule modulators of CXCR4 and ACKR3 are reviewed. These modulators have led to the development of radio- and fluorescently labeled tool compounds, enabling the visualization of ligand binding and receptor characterization both in vitro and in vivo. SIGNIFICANCE STATEMENT: To investigate the pharmacological modulation of CXCR4 and ACKR3, significant effort has been focused on the discovery and development of a range of ligands, including small-molecule modulators, pepducins, and synthetic peptides. Imaging tools, such as fluorescent probes, also play a pivotal role in the field of drug discovery. This review aims to provide an overview of the aforementioned modulators that facilitate the study of CXCR4 and ACKR3 receptors.
Collapse
Affiliation(s)
- Ilze Adlere
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Birgit Caspar
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Marta Arimont
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Sebastian Dekkers
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Kirsten Visser
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Jeffrey Stuijt
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Chris de Graaf
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Michael Stocks
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Barrie Kellam
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Stephen Briddon
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Maikel Wijtmans
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Iwan de Esch
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Stephen Hill
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Rob Leurs
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| |
Collapse
|
11
|
Shao Y, Zhou F, He D, Zhang L, Shen J. Overexpression of CXCR7 promotes mesenchymal stem cells to repair phosgene-induced acute lung injury in rats. Biomed Pharmacother 2018; 109:1233-1239. [PMID: 30551373 DOI: 10.1016/j.biopha.2018.10.108] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/11/2018] [Accepted: 10/20/2018] [Indexed: 12/14/2022] Open
Abstract
Phosgene exposure may result in acute lung injury (ALI) with high mortality. Emerging evidence suggests that mesenchymal stem cells (MSCs) have a therapeutic potential against ALI. CXC chemokine receptor 7 (CXCR7) has been identified as a receptor of stromal-cell-derived factor 1 (SDF1) involved in MSC migration and may be an important mediator of the therapeutic effects of MSCs on ALI. In our study, we initially constructed a lentiviral vector overexpressing CXCR7 and then successfully transduced it into rat bone marrow-derived MSCs (resulting in MSCs-CXCR7). We found that ALI and the wet-to-dry ratio significantly decreased in the phosgene-exposed rats after administration of MSCs-CXCR7 or MSCs-GFP. Indeed, treatment with MSCs-CXCR7 caused further improvement. Moreover, injection of MSCs-CXCR7 significantly facilitated MSC homing to injured lung tissue. Meanwhile, overexpression of CXCR7 promoted differentiation of MSCs into type II alveolar epithelial (AT II) cells and enhanced the ability of MSCs to modulate the inflammatory response in phosgene-induced ALI. Taken together, our findings suggest that CXCR7-overexpressing MSCs may markedly facilitate treatment of phosgene-induced ALI (P-ALI) in rats.
Collapse
Affiliation(s)
- Yiru Shao
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Fangqing Zhou
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Daikun He
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Lin Zhang
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Jie Shen
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China.
| |
Collapse
|
12
|
Szpakowska M, Meyrath M, Reynders N, Counson M, Hanson J, Steyaert J, Chevigné A. Mutational analysis of the extracellular disulphide bridges of the atypical chemokine receptor ACKR3/CXCR7 uncovers multiple binding and activation modes for its chemokine and endogenous non-chemokine agonists. Biochem Pharmacol 2018. [DOI: 10.1016/j.bcp.2018.03.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
13
|
Moreno MJ, Gallardo A, Novelli S, Mozos A, Aragó M, Pavón MÁ, Céspedes MV, Pallarès V, Falgàs A, Alcoceba M, Blanco O, Gonzalez-Díaz M, Sierra J, Mangues R, Casanova I. CXCR7 expression in diffuse large B-cell lymphoma identifies a subgroup of CXCR4+ patients with good prognosis. PLoS One 2018; 13:e0198789. [PMID: 29920526 PMCID: PMC6007902 DOI: 10.1371/journal.pone.0198789] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 04/14/2018] [Indexed: 01/10/2023] Open
Abstract
The CXCR4/CXCL12 axis has been extensively associated with different types of cancer correlating with higher aggressiveness and metastasis. In diffuse large B-cell lymphoma (DLBCL), the expression of the chemokine receptor CXCR4 is involved in the dissemination of malignant B cells and is a marker of poor prognosis. CXCR7 is a chemokine receptor that binds to the same ligand as CXCR4 and regulates de CXCR4-CXCL12 axis. These findings together with the report of CXCR7 prognostic value in several tumor types, led us to evaluate the expression of CXCR7 in diffuse large B-cell lymphoma biopsies. Here, we describe that CXCR7 receptor is an independent prognostic factor that associates with good clinical outcome. Moreover, the expression of CXCR7 associates with increased survival in CXCR4+ but not in CXCR4- DLBCL patients. Thus, the combined immunohistochemical evaluation of both CXCR7 and CXCR4 expression in DLBCL biopsies may improve their prognostic value as single markers. Finally, we show that CXCR7 overexpression in vitro is able to diminish DLBCL cell survival and increase their sensitivity to antitumor drugs. Hence, further studies on the CXCR7 receptor may establish its role in DLBCL and the molecular mechanisms that modulate CXCR4 activity.
Collapse
MESH Headings
- Adult
- Aged
- Biomarkers, Tumor
- Biopsy
- Cell Line, Tumor
- Chemokine CXCL12/physiology
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Kaplan-Meier Estimate
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- Middle Aged
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Prognosis
- Proportional Hazards Models
- Receptors, CXCR/biosynthesis
- Receptors, CXCR/genetics
- Receptors, CXCR/physiology
- Receptors, CXCR4/analysis
Collapse
Affiliation(s)
- María José Moreno
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
| | - Alberto Gallardo
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Silvana Novelli
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ana Mozos
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Marc Aragó
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Miguel Ángel Pavón
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
| | - María Virtudes Céspedes
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
| | - Víctor Pallarès
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Aïda Falgàs
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Miguel Alcoceba
- Department of Hematology and Pathology, IBSAL-University Hospital, Center for Cancer Research-IBMCC (USAL-CSIC), Salamanca, Spain
- CIBER in Oncology (CIBER-ONC), Madrid, Spain
| | - Oscar Blanco
- Department of Hematology and Pathology, IBSAL-University Hospital, Center for Cancer Research-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Marcos Gonzalez-Díaz
- Department of Hematology and Pathology, IBSAL-University Hospital, Center for Cancer Research-IBMCC (USAL-CSIC), Salamanca, Spain
- CIBER in Oncology (CIBER-ONC), Madrid, Spain
| | - Jorge Sierra
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Josep Carreras Research Institute, Barcelona, Spain
| | - Ramon Mangues
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
- Josep Carreras Research Institute, Barcelona, Spain
- * E-mail:
| | - Isolda Casanova
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomecidicina (CIBER-BBN), Barcelona, Spain
- Josep Carreras Research Institute, Barcelona, Spain
| |
Collapse
|
14
|
Ameti R, Melgrati S, Radice E, Cameroni E, Hub E, Thelen S, Rot A, Thelen M. Characterization of a chimeric chemokine as a specific ligand for ACKR3. J Leukoc Biol 2018; 104:391-400. [DOI: 10.1002/jlb.2ma1217-509r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/03/2018] [Accepted: 01/03/2018] [Indexed: 12/19/2022] Open
Affiliation(s)
- Rafet Ameti
- Institute for Research in Biomedicine; Università della Svizzera italiana; Bellinzona Switzerland
- Graduate School for Cellular and Biomedical Sciences; University of Bern; Bern Switzerland
| | - Serena Melgrati
- Institute for Research in Biomedicine; Università della Svizzera italiana; Bellinzona Switzerland
- University of York; York United Kingdom
| | - Egle Radice
- Institute for Research in Biomedicine; Università della Svizzera italiana; Bellinzona Switzerland
- Graduate School for Cellular and Biomedical Sciences; University of Bern; Bern Switzerland
| | - Elisabetta Cameroni
- Institute for Research in Biomedicine; Università della Svizzera italiana; Bellinzona Switzerland
| | - Elin Hub
- The William Harvey Research Institute; Queen Mary University London; London United Kingdom
| | - Sylvia Thelen
- Institute for Research in Biomedicine; Università della Svizzera italiana; Bellinzona Switzerland
| | - Antal Rot
- The William Harvey Research Institute; Queen Mary University London; London United Kingdom
- Institute for Cardiovascular Prevention; Ludwig-Maximilians University (LMU); Munich Germany
| | - Marcus Thelen
- Institute for Research in Biomedicine; Università della Svizzera italiana; Bellinzona Switzerland
| |
Collapse
|
15
|
Mazzotti C, Gagliostro V, Bosisio D, Del Prete A, Tiberio L, Thelen M, Sozzani S. The Atypical Receptor CCRL2 (C-C Chemokine Receptor-Like 2) Does Not Act As a Decoy Receptor in Endothelial Cells. Front Immunol 2017; 8:1233. [PMID: 29056935 PMCID: PMC5635198 DOI: 10.3389/fimmu.2017.01233] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/19/2017] [Indexed: 11/26/2022] Open
Abstract
C-C chemokine receptor-like 2 (CCRL2) is a non-signaling seven-transmembrane domain (7-TMD) receptor related to the atypical chemokine receptor (ACKR) family. ACKRs bind chemokines but do not activate G protein-dependent signaling or cell functions. ACKRs were shown to regulate immune functions in vivo by their ability to scavenge chemokines from the local environment. This study was performed to investigate whether CCRL2 shares two of the main characteristics of ACKRs, namely the ability to internalize and scavenge the ligands. Cell membrane analysis of CCRL2-transfected cells revealed a weak, constitutive, ligand-independent internalization, and recycling of CCRL2, with a kinetics that was slower than those observed with ACKR3, a prototypic ACKR, or other chemotactic signaling receptors [i.e., chemokine-like receptor 1 and C-X-C motif chemokine receptor 2]. Intracellularly, CCRL2 colocalized with early endosome antigen 1-positive and Rab5-positive vesicles and with recycling compartments mainly characterized by Rab11-positive vesicles. CCRL2-transfected cells and activated mouse blood endothelial cells, that endogenously express CCRL2, were used to investigate the scavenging ability of CCRL2. These experiments confirmed the ability of CCRL2 to bind chemerin, the only recognized ligand, but excluded the ability of CCRL2 to perform scavenging. Collectively, these results identify unique functional properties for this member of the non-signaling 7-TMD receptor family.
Collapse
Affiliation(s)
- Chiara Mazzotti
- Laboratory of Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Vincenzo Gagliostro
- Laboratory of Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Daniela Bosisio
- Laboratory of Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Annalisa Del Prete
- Laboratory of Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Humanitas Clinical and Research Centre, Rozzano, Italy
| | - Laura Tiberio
- Laboratory of Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Silvano Sozzani
- Laboratory of Experimental Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Humanitas Clinical and Research Centre, Rozzano, Italy
| |
Collapse
|
16
|
Puddinu V, Casella S, Radice E, Thelen S, Dirnhofer S, Bertoni F, Thelen M. ACKR3 expression on diffuse large B cell lymphoma is required for tumor spreading and tissue infiltration. Oncotarget 2017; 8:85068-85084. [PMID: 29156704 PMCID: PMC5689594 DOI: 10.18632/oncotarget.18844] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/05/2017] [Indexed: 12/30/2022] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is the most frequent lymphoma accounting for more than the 30% of the cases. Involvement of extranodal sites, such as bone marrow and central nervous system, is associated with poor prognosis. A contribution of the chemokine system in these processes is assumed as it is known as a critical regulator of the metastatic process in cancer. The atypical chemokine receptor 3 (ACKR3), which does not couple to G-proteins and does not mediate cell migration, acts as a scavenger for CXCL11 and CXCL12, interfering with the tumor homing CXCL12/CXCR4 axis. Here, functional expression of ACKR3 in DLBCL cells was necessary for colonization of the draining lymph node in an in vivo subcutaneous lymphoma model. Moreover, in a disseminated in vivo lymphoma model, ACKR3 expression was required for bone marrow and brain invasion and local tumor growth. The present data unveil ACKR3 as potential therapeutic target for the control of tumor dissemination in DLBCL.
Collapse
Affiliation(s)
- Viola Puddinu
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Sabrina Casella
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Egle Radice
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Sylvia Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Stefan Dirnhofer
- Institute of Pathology, University Hospital, University of Basel, Basel, Switzerland
| | | | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| |
Collapse
|
17
|
Szpakowska M, Dupuis N, Baragli A, Counson M, Hanson J, Piette J, Chevigné A. Human herpesvirus 8-encoded chemokine vCCL2/vMIP-II is an agonist of the atypical chemokine receptor ACKR3/CXCR7. Biochem Pharmacol 2016; 114:14-21. [DOI: 10.1016/j.bcp.2016.05.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/25/2016] [Indexed: 10/21/2022]
|
18
|
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
|
19
|
Abstract
Migration and positioning of cells is fundamental for complex functioning of multicellular organisms. During an immune response, cells are recruited from remote distances to a distinct location. Cells that are passively transported leave the circulation stimulated by locally produced signals and follow chemotactic cues to reach specific destinations. Such gradients are short (<150 μm) and require a source of production where the concentration is the highest and a sink in apposition where the attractant dissipates and the concentration is the lowest. Several straight forward methods exist to identify in vitro and in vivo cells producing chemoattractants. This can be achieved at the transcriptional level and by measuring secreted proteins. However, to demonstrate the activity of sinks in vitro and in vivo is more challenging. Cell-mediated dissipation of an attractant must be revealed by measuring its uptake and subsequent destruction. Elimination of chemoattractants such as chemokines can be monitored in vitro using radiolabeled ligands or more elegantly with fluorescent-labeled chemoattractants. The latter method can also be used in vivo and enables to monitor the process in real time using time-lapse video microscopy. In this chapter, we describe methods to produce fluorescently labeled chemokines either as fusion proteins secreted from insect cells or as recombinant bacterial proteins that can enzymatically be labeled. We discuss methods that were successfully used to demonstrate sink activities of scavenger receptors. Moreover, fluorescent chemokines can be used to noninvasively analyze receptor expression and activity in living cells.
Collapse
Affiliation(s)
- Barbara Moepps
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.
| |
Collapse
|
20
|
Palmesino E, Apuzzo T, Thelen S, Mueller B, Langen H, Thelen M. Association of eukaryotic translation initiation factor eIF2B with fully solubilized CXCR4. J Leukoc Biol 2015; 99:971-8. [PMID: 26609049 DOI: 10.1189/jlb.2ma0915-415r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/31/2015] [Indexed: 12/31/2022] Open
Abstract
Chemokine receptors are key regulators of leukocyte trafficking but also have an important role in development, tumor growth, and metastasis. Among the chemokine receptors, CXCR4 is the only one that leads to perinatal death when genetically ablated in mice, indicating a more-widespread function in development. To identify pathways that are activated downstream of CXCR4, a solubilization protocol was elaborated, which allows for the isolation of the endogenous receptor from human cells in its near-native conformation. Solubilized CXCR4 is recognized by the conformation-sensitive monoclonal antibody 12G5 and retains the ability to bind CXCL12 in solution, which was abolished in the presence of receptor antagonists. Mass spectrometry of CXCR4 immunoprecipitates revealed a specific interaction with the pentameric eukaryotic translation initiation factor 2B. The observation that the addition of CXCL12 leads to the dissociation of eukaryotic translation initiation factor 2B from CXCR4 suggests that stimulation of the receptor may trigger the local protein synthesis required for efficient cell movement.
Collapse
Affiliation(s)
- Elena Palmesino
- Institute for Research in Biomedicine, Bellinzona, Switzerland; and
| | - Tiziana Apuzzo
- Institute for Research in Biomedicine, Bellinzona, Switzerland; and
| | - Sylvia Thelen
- Institute for Research in Biomedicine, Bellinzona, Switzerland; and
| | - Bernd Mueller
- Protein and Metabolite Technologies, F. Hoffmann-La Roche Ltd, Pharmaceutical Sciences Roche Innovation Center, Basel, Switzerland
| | - Hanno Langen
- Protein and Metabolite Technologies, F. Hoffmann-La Roche Ltd, Pharmaceutical Sciences Roche Innovation Center, Basel, Switzerland
| | - Marcus Thelen
- Institute for Research in Biomedicine, Bellinzona, Switzerland; and
| |
Collapse
|
21
|
Herpes simplex virus enhances chemokine function through modulation of receptor trafficking and oligomerization. Nat Commun 2015; 6:6163. [PMID: 25625471 DOI: 10.1038/ncomms7163] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 12/19/2014] [Indexed: 01/13/2023] Open
Abstract
Glycoprotein G (gG) from herpes simplex virus 1 and 2 (HSV-1 and HSV-2, important human neurotropic pathogens) is the first viral chemokine-binding protein found to potentiate chemokine function. Here we show that gG attaches to cell surface glycosaminoglycans and induces lipid raft clustering, increasing the incorporation of CXCR4 receptors into these microdomains. gG induces conformational rearrangements in CXCR4 homodimers and changes their intracellular partners, leading to sustained, functional chemokine/receptor complexes at the surface. This results in increased chemotaxis dependent on the cholesterol content of the plasma membrane and receptor association to Src-kinases and phosphatidylinositol-3-kinase signalling pathways, but independent of clathrin-mediated endocytosis. Furthermore, using electron microscopy, we show that such enhanced functionality is associated with the accumulation of low-order CXCR4 nanoclusters. Our results provide insights into basic mechanisms of chemokine receptor function and into a viral strategy of immune modulation.
Collapse
|
22
|
Kaboord B, Smith S, Patel B, Meier S. Enrichment of low-abundant protein targets by immunoprecipitation upstream of mass spectrometry. Methods Mol Biol 2015; 1295:135-151. [PMID: 25820720 DOI: 10.1007/978-1-4939-2550-6_12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Immunoprecipitation (IP) is commonly used upstream of mass spectrometry (MS) as an enrichment tool for low-abundant protein targets. However, several aspects of the classical IP procedure such as nonspecific protein binding to the isolation matrix, detergents or high salt concentrations in wash and elution buffers, and antibody chain contamination in elution fractions render it incompatible with downstream mass spectrometry analysis. Here, we discuss two IP workflows that are designed to minimize or eliminate these contaminants: the first employs biotinylated antibodies and streptavidin magnetic beads while the second method utilizes a traditional antibody that is oriented and cross-linked to Protein AG magnetic beads. Both modified magnetic supports have low background binding and both antibody immobilization strategies significantly reduce or eliminate antibody heavy and light chain contamination in the eluent, minimizing potential ion suppression effects and thereby maximizing detection of target antigens and interacting proteins.
Collapse
Affiliation(s)
- Barbara Kaboord
- Research & Development, Thermo Fisher Scientific, 3747 N. Meridian Rd., Rockford, IL, 61101, USA,
| | | | | | | |
Collapse
|
23
|
CXCR4 is dispensable for T cell egress from chronically inflamed skin via the afferent lymph. PLoS One 2014; 9:e95626. [PMID: 24752354 PMCID: PMC3994085 DOI: 10.1371/journal.pone.0095626] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 03/27/2014] [Indexed: 11/19/2022] Open
Abstract
T cell recirculation through extralymphoid tissues is essential to immune surveillance, host defense and inflammation. In this process, T cells enter the tissue from the blood and subsequently leave via the afferent lymph. In the absence of inflammation, T cells require CCR7 expression to egress from the skin or lung, which is consistent with the constitutive expression of the CCR7 ligand CCL21 on lymphatic endothelium. However, during chronic inflammation alternative chemoattractants come into play, allowing Ccr7-deficient (Ccr7−/−) T cells to egress efficiently from affected skin. As T cell egress from inflamed sites is a potential control point of the inflammatory response, we aimed to determine alternative T cell exit receptors using a mouse and a sheep model. We show that CCR7+ and CCR7– T cells exiting from the chronically inflamed skin were highly responsive to the CXCR4 ligand CXCL12, which was induced in the lymphatics in the inflamed site. Based on these findings, we hypothesized that CXCR4 mediates T cell egress from inflamed skin. However, pharmacological inhibition of CXCR4 did not affect the tissue egress of wildtype or Ccr7−/− CD4 and CD8 T cells after adoptive transfer into chronically inflamed skin. Similarly, adoptively transferred Cxcr4−/− Ccr7−/− and Ccr7−/− T cells egressed from the inflamed skin equally well. Based on these data, we conclude that, while CXCR4 might play an essential role for other cell types that enter the afferent lymphatics, it is dispensable for T cell egress from the chronically inflamed skin.
Collapse
|
24
|
Qi Z, Wang J, Han X, Yang J, Zhao G, Cao Y. Listr1 locus regulates innate immunity against Listeria monocytogenes infection in the mouse liver possibly through Cxcl11 polymorphism. Immunogenetics 2014; 66:231-42. [PMID: 24566892 DOI: 10.1007/s00251-014-0761-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/21/2014] [Indexed: 12/31/2022]
Abstract
Inbred stains of mice display differential susceptibility to infection with the common foodborne pathogen Listeria monocytogenes (Lm). Previously, Listr1 and Listr2, two genetic loci that control differential sensitivity to Lm infection between BALB/cByJ and C57BL/6ByJ mice, were identified. To analyze the role of Listr1 in innate immune responses, we employed congenic mice (C.B6By-Listr1/Rag2 (-/-) ) bearing the C57BL/6ByJ-derived Listr1 locus on a BALB/c-Rag2 (-/-) background. Consistent with the results of a previous genetic analysis, the congenic mice showed increased susceptibility to Lm infection. The bacterial burden in the liver between the congenic and control lines was significantly different (P < 0.05) from 24 h postinfection with Lm. Analysis of genes within the Listr1 locus identified a frameshift mutation in the Cxcl11 gene of the C57BL/6 strain that prevents production of the mature chemokine CXCL11. No differences in inflammatory cell infiltration or cells expressing CXCR3 and CXCR7 which are the receptors of CXCL11 occurred because of CXCL11 deficiency in the congenic mice spleens. However, these mice lacked a distinct population of CD14(+) positive resident mononuclear cells that express intermediate levels of CXCR3 and CXCR7 in the liver. There were fewer microabscesses in the liver of CXCL11-deficient mice during the early stage of infection, which is consistent with their decreased ability to resist Lm. Our results, when taken together, show that the Listr1 locus plays an important role in early control of Lm infection in the mouse liver and that Cxcl11 is a candidate gene for disease severity within this locus.
Collapse
Affiliation(s)
- Zanmei Qi
- Department of Immunology, College of Basic Medical Sciences, China Medical University, No. 92 Bei Er Ma Lu, Shenyang, Liaoning, 110001, China,
| | | | | | | | | | | |
Collapse
|
25
|
Humpert ML, Pinto D, Jarrossay D, Thelen M. CXCR7 influences the migration of B cells during maturation. Eur J Immunol 2014; 44:694-705. [PMID: 24259140 DOI: 10.1002/eji.201343907] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 10/31/2013] [Accepted: 11/15/2013] [Indexed: 12/25/2022]
Abstract
The atypical chemokine receptor CXCR7 binds the chemokines CXCL12 and CXCL11. The receptor is widely expressed and was shown to tune CXCR12-induced responses of CXCR4. Here, the function of CXCR7 was examined at late stages of human B-cell maturation, when B cells differentiate into Ab-secreting plasmablasts. We identified two populations of CXCR7(+) cells in tonsillar lymphocytes, one being presumably memory B cells or early plasmablasts (FSC(low) CD19(+) CD38(mid) ) and the other being plasmablasts or early plasma cells (FSC(high) CD19(+) CD38(+) ). CXCR7 is expressed on CD19(+) CD27(+) memory B cells, on CD19(+) CD38(+) CD138(-) and intracellular immunoglobulin high plasmablasts, but not on CD19(+) CD138(+) icIg(high) plasma cells. The differential expression pattern suggests a potential contribution of the scavenger receptor in final B-cell maturation. On in vitro differentiating B cells, we found a marked inverse correlation between CXCR7 and CXCR5 cell surface levels, whereas expression of CXCR4 remained almost constant. Migration assays performed with tonsillar mononuclear cells or in vitro differentiated cells revealed that inhibition of CXCR7 markedly increases chemotaxis toward CXCL12, especially at late stages of B-cell maturation. Chemotaxis was attenuated in the presence of CXCR4 antagonists, confirming that migration is CXCR4 mediated. Our findings unequivocally demonstrate a novel role for CXCR7 in regulating the migration of plasmablasts during B-cell maturation.
Collapse
Affiliation(s)
- Marie-Luise Humpert
- Institute for Research in Biomedicine, Bellinzona, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | | | | |
Collapse
|
26
|
A general method for site specific fluorescent labeling of recombinant chemokines. PLoS One 2014; 9:e81454. [PMID: 24489642 PMCID: PMC3904831 DOI: 10.1371/journal.pone.0081454] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 10/22/2013] [Indexed: 11/19/2022] Open
Abstract
Chemokines control cell migration in many contexts including development, homeostasis, immune surveillance and inflammation. They are also involved in a wide range of pathological conditions ranging from inflammatory diseases and cancer, to HIV. Chemokines function by interacting with two types of receptors: G protein-coupled receptors on the responding cells, which transduce signaling pathways associated with cell migration and activation, and glycosaminoglycans on cell surfaces and the extracellular matrix which organize and present some chemokines on immobilized surface gradients. To probe these interactions, imaging methods and fluorescence-based assays are becoming increasingly desired. Herein, a method for site-specific fluorescence labeling of recombinant chemokines is described. It capitalizes on previously reported 11–12 amino acid tags and phosphopantetheinyl transferase enzymes to install a fluorophore of choice onto a specific serine within the tag through a coenzyme A-fluorophore conjugate. The generality of the method is suggested by our success in labeling several chemokines (CXCL12, CCL2, CCL21 and mutants thereof) and visualizing them bound to chemokine receptors and glycosaminoglycans. CXCL12 and CCL2 showed the expected co-localization on the surface of cells with their respective receptors CXCR4 and CCR2 at 4°C, and co-internalization with their receptors at 37°C. By contrast, CCL21 showed the presence of large discrete puncta that were dependent on the presence of both CCR7 and glycosaminoglycans as co-receptors. These data demonstrate the utility of this labeling approach for the detection of chemokine interactions with GAGs and receptors, which can vary in a chemokine-specific manner as shown here. For some applications, the small size of the fluorescent adduct may prove advantageous compared to other methods (e.g. antibody labeling, GFP fusion) by minimally perturbing native interactions. Other advantages of the method are the ease of bacterial expression, the versatility of labeling with any maleimide-fluorophore conjugate of interest, and the covalent nature of the fluorescent adduct.
Collapse
|
27
|
Abstract
Chemokines have fundamental roles in regulating immune and inflammatory responses, primarily through their control of leukocyte migration and localization. The biological functions of chemokines are typically mediated by signalling through G protein-coupled chemokine receptors, but chemokines are also bound by a small family of atypical chemokine receptors (ACKRs), the members of which are unified by their inability to initiate classical signalling pathways after ligand binding. These ACKRs are emerging as crucial regulatory components of chemokine networks in a wide range of developmental, physiological and pathological contexts. In this Review, we discuss the biochemical and immunological properties of ACKRs and the potential unifying themes in this family, and we highlight recent studies that identify novel roles for these molecules in development , homeostasis, inflammatory disease, infection and cancer.
Collapse
|
28
|
The peculiarities of the SDF-1/CXCL12 system: in some cells, CXCR4 and CXCR7 sing solos, in others, they sing duets. Cell Tissue Res 2013; 355:239-53. [DOI: 10.1007/s00441-013-1747-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 10/17/2013] [Indexed: 12/26/2022]
|
29
|
Bachelerie F, Ben-Baruch A, Burkhardt AM, Combadiere C, Farber JM, Graham GJ, Horuk R, Sparre-Ulrich AH, Locati M, Luster AD, Mantovani A, Matsushima K, Murphy PM, Nibbs R, Nomiyama H, Power CA, Proudfoot AEI, Rosenkilde MM, Rot A, Sozzani S, Thelen M, Yoshie O, Zlotnik A. International Union of Basic and Clinical Pharmacology. [corrected]. LXXXIX. Update on the extended family of chemokine receptors and introducing a new nomenclature for atypical chemokine receptors. Pharmacol Rev 2013; 66:1-79. [PMID: 24218476 PMCID: PMC3880466 DOI: 10.1124/pr.113.007724] [Citation(s) in RCA: 693] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Sixteen years ago, the Nomenclature Committee of the International Union of Pharmacology approved a system for naming human seven-transmembrane (7TM) G protein-coupled chemokine receptors, the large family of leukocyte chemoattractant receptors that regulates immune system development and function, in large part by mediating leukocyte trafficking. This was announced in Pharmacological Reviews in a major overview of the first decade of research in this field [Murphy PM, Baggiolini M, Charo IF, Hébert CA, Horuk R, Matsushima K, Miller LH, Oppenheim JJ, and Power CA (2000) Pharmacol Rev 52:145-176]. Since then, several new receptors have been discovered, and major advances have been made for the others in many areas, including structural biology, signal transduction mechanisms, biology, and pharmacology. New and diverse roles have been identified in infection, immunity, inflammation, development, cancer, and other areas. The first two drugs acting at chemokine receptors have been approved by the U.S. Food and Drug Administration (FDA), maraviroc targeting CCR5 in human immunodeficiency virus (HIV)/AIDS, and plerixafor targeting CXCR4 for stem cell mobilization for transplantation in cancer, and other candidates are now undergoing pivotal clinical trials for diverse disease indications. In addition, a subfamily of atypical chemokine receptors has emerged that may signal through arrestins instead of G proteins to act as chemokine scavengers, and many microbial and invertebrate G protein-coupled chemokine receptors and soluble chemokine-binding proteins have been described. Here, we review this extended family of chemokine receptors and chemokine-binding proteins at the basic, translational, and clinical levels, including an update on drug development. We also introduce a new nomenclature for atypical chemokine receptors with the stem ACKR (atypical chemokine receptor) approved by the Nomenclature Committee of the International Union of Pharmacology and the Human Genome Nomenclature Committee.
Collapse
Affiliation(s)
- Francoise Bachelerie
- Chair, Subcommittee on Chemokine Receptors, Nomenclature Committee-International Union of Pharmacology, Bldg. 10, Room 11N113, NIH, Bethesda, MD 20892.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Berahovich RD, Penfold MET, Miao Z, Walters MJ, Jaen JC, Schall TJ. Differences in CXCR7 protein expression on rat versus mouse and human splenic marginal zone B cells. Immunol Lett 2013; 154:77-9. [PMID: 23954811 DOI: 10.1016/j.imlet.2013.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 08/01/2013] [Accepted: 08/05/2013] [Indexed: 12/27/2022]
Affiliation(s)
- Robert D Berahovich
- ChemoCentryx, Inc., 850 Maude Avenue, Mountain View, CA 94043, United States.
| | | | | | | | | | | |
Collapse
|
31
|
Deregulation of ETS1 and FLI1 contributes to the pathogenesis of diffuse large B-cell lymphoma. Blood 2013; 122:2233-41. [PMID: 23926301 DOI: 10.1182/blood-2013-01-475772] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common form of human lymphoma. DLBCL is a heterogeneous disease characterized by different genetic lesions. We herein report the functional characterization of a recurrent gain mapping on chromosome 11q24.3, found in 23% of 166 DLBCL cases analyzed. The transcription factors ETS1 and FLI1, located within the 11q24.3 region, had significantly higher expression in clinical samples carrying the gain. Functional studies on cell lines showed that ETS1 and FLI1 cooperate in sustaining DLBCL proliferation and viability and regulate genes involved in germinal center differentiation. Taken together, these data identify the 11q24.3 gain as a recurrent lesion in DLBCL leading to ETS1 and FLI1 deregulated expression, which can contribute to the pathogenesis of this disease.
Collapse
|
32
|
Expression of CXCL12 receptors in B cells from Mexican Mestizos patients with systemic Lupus erythematosus. J Transl Med 2012; 10:251. [PMID: 23244336 PMCID: PMC3571925 DOI: 10.1186/1479-5876-10-251] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 12/14/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Systemic Lupus Erythematosus (SLE) is a chronic autoimmune disease characterized by B-cell hyper-reactivity and the production of pathogenic anti-nuclear-directed auto-antibodies (Abs). B-cell ontogeny is partly dependent on the CXCL12/CXCR4 axis for which the contribution to SLE pathogenesis remains unclear. CXCR7, the novel receptor for CXCL12, is differentially expressed among memory B-cell subsets. However, its biological role in SLE remains to be explored. METHODS Relative CXCR4 and CXCR7 expression levels were compared by quantitative PCR in leukocytes from blood samples of 41 Mexican Mestizos patients with SLE and 45 ethnicity-matched healthy subjects. Intracellular and membrane expression of both receptors was analyzed by flow cytometry in naive and Ab-secreting B cells. B-cell responsiveness to CXCL12 was investigated using Transwell-based chemotaxis assays. Data were analyzed using the Kruskal-Wallis test for comparisons of values amongst healthy controls and patients with inactive or active SLE, and non-parametrically using the Mann-Whitney U-test for multiple comparisons and unpaired samples. Correlations were determined by Spearman's ranking. RESULT SLE leukocytes displayed reduced levels of CXCR4 and CXCR7 transcripts. In SLE patients, a significant defect in CXCR4 expression was detected at the surface of naive and Ab-secreting B cells, associated with an abnormal intracellular localization of the receptor. CXCR7 predominantly localized in cytosolic compartments of B cells from healthy and SLE individuals. Disease activity did not impact on these expression patterns. Altered receptor compartmentalization correlated with an impaired CXCL12-promoted migration of SLE B cells. CONCLUSIONS Our data highlight a down-regulation of CXCL12 receptors on circulating B cells from SLE patients that likely influences their migratory behavior and distribution.
Collapse
|
33
|
Sánchez-Martín L, Sánchez-Mateos P, Cabañas C. CXCR7 impact on CXCL12 biology and disease. Trends Mol Med 2012; 19:12-22. [PMID: 23153575 DOI: 10.1016/j.molmed.2012.10.004] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 09/20/2012] [Accepted: 10/11/2012] [Indexed: 12/14/2022]
Abstract
It is known that the chemokine receptor CXCR7 (RDC1) can be engaged by both chemokines CXCL12 (SDF-1) and CXCL11 (I-TAC), but the exact expression pattern and function of CXCR7 is controversial. CXCR7 expression seems to be enhanced during pathological inflammation and tumor development, and emerging data suggest this receptor is an attractive therapeutic target for autoimmune diseases and cancer. CXCR7/CXCR4 heterodimerization, β-arrestin-mediated signaling, and modulation of CXCL12 responsiveness by CXCR7 suggest that the monogamous CXCR4/CXCL12 signaling axis is an oversimplified model that needs to be revisited. Consequently, research into CXCR7 biology is of great interest and further studies are warranted. This review summarizes recent findings about the CXCR7 receptor and analyses its impact on understanding the roles of CXCL12 biology in health and disease.
Collapse
Affiliation(s)
- Lorena Sánchez-Martín
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid 28049, Spain.
| | | | | |
Collapse
|
34
|
Singh AK, Arya RK, Trivedi AK, Sanyal S, Baral R, Dormond O, Briscoe DM, Datta D. Chemokine receptor trio: CXCR3, CXCR4 and CXCR7 crosstalk via CXCL11 and CXCL12. Cytokine Growth Factor Rev 2012; 24:41-9. [PMID: 22989616 DOI: 10.1016/j.cytogfr.2012.08.007] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 08/20/2012] [Indexed: 12/16/2022]
Abstract
Although chemokines are well established to function in immunity and endothelial cell activation and proliferation, a rapidly growing literature suggests that CXC Chemokine receptors CXCR3, CXCR4 and CXCR7 are critical in the development and progression of solid tumors. The effect of these chemokine receptors in tumorigenesis is mediated via interactions with shared ligands I-TAC (CXCL11) and SDF-1 (CXCL12). Over the last decade, CXCR4 has been extensively reported to be overexpressed in most human solid tumors and has earned considerable attention toward elucidating its role in cancer metastasis. To enrich the existing armamentarium of anti-cancerous agents, many inhibitors of CXCL12-CXCR4 axis have emerged as additional or alternative agents for neo-adjuvant treatments and even many of them are in preclinical and clinical stages of their development. However, the discovery of CXCR7 as another receptor for CXCL12 with rather high binding affinity and recent reports about its involvement in cancer progression, has questioned the potential of "selective blockade" of CXCR4 as cancer chemotherapeutics. Interestingly, CXCR7 can also bind another chemokine CXCL11, which is an established ligand for CXCR3. Recent reports have documented that CXCR3 and their ligands are overexpressed in different solid tumors and regulate tumor growth and metastasis. Therefore, it is important to consider the interactions and crosstalk between these three chemokine receptors and their ligand mediated signaling cascades for the development of effective anti-cancer therapies. Emerging evidence also indicates that these receptors are differentially expressed in tumor endothelial cells as well as in cancer stem cells, suggesting their direct role in regulating tumor angiogenesis and metastasis. In this review, we will focus on the signals mediated by this receptor trio via their shared ligands and their role in tumor growth and progression.
Collapse
Affiliation(s)
- Anup Kumar Singh
- Drug Target Discovery and Development (DTDD) Division, CSIR-Central Drug Research Institute, Lucknow 226001, India
| | | | | | | | | | | | | | | |
Collapse
|