1
|
Spatz P, Chen X, Reichau K, Huber ME, Mühlig S, Matsusaka Y, Schiedel M, Higuchi T, Decker M. Development and Initial Characterization of the First 18F-CXCR2-Targeting Radiotracer for PET Imaging of Neutrophils. J Med Chem 2024; 67:6327-6343. [PMID: 38570909 DOI: 10.1021/acs.jmedchem.3c02285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
The interleukin-8 receptor beta (CXCR2) is a highly promising target for molecular imaging of inflammation and inflammatory diseases. This is due to its almost exclusive expression on neutrophils. Modified fluorinated ligands were designed based on a squaramide template, with different modification sites and synthetic strategies explored. Promising candidates were then tested for affinity to CXCR2 in a NanoBRET competition assay, resulting in tracer candidate 16b. As direct 18F-labeling using established tosyl chemistry did not yield the expected radiotracer, an indirect labeling approach was developed. The radiotracer [18F]16b was obtained with a radiochemical yield of 15% using tert-butyl (S)-3-(tosyloxy)pyrrolidine carboxylate and a pentafluorophenol ester. The subsequent time-dependent uptake of [18F]16b in CXCR2-negative and CXCR2-overexpressing human embryonic kidney cells confirmed the radiotracer's specificity. Further studies with human neutrophils revealed its diagnostic potential for functional imaging of neutrophils.
Collapse
Affiliation(s)
- Philipp Spatz
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, University of Würzburg, Würzburg 97074, Germany
| | - Xinyu Chen
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
- Department of Nuclear Medicine and Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg 97080, Germany
| | - Kora Reichau
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, University of Würzburg, Würzburg 97074, Germany
| | - Max E Huber
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen 91058, Germany
| | - Saskia Mühlig
- Department of Nuclear Medicine and Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg 97080, Germany
| | - Yohji Matsusaka
- Department of Nuclear Medicine and Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg 97080, Germany
| | - Matthias Schiedel
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen 91058, Germany
- Pharmaceutical and Medicinal Chemistry, Institute of Medicinal and Pharmaceutical Chemistry, Technical University of Braunschweig, Braunschweig 38106, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine and Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg 97080, Germany
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-0082, Japan
| | - Michael Decker
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, University of Würzburg, Würzburg 97074, Germany
| |
Collapse
|
2
|
Casella B, Farmer JP, Nesheva DN, Williams HEL, Charlton SJ, Holliday ND, Laughton CA, Mistry SN. Design, Synthesis, and Application of Fluorescent Ligands Targeting the Intracellular Allosteric Binding Site of the CXC Chemokine Receptor 2. J Med Chem 2023; 66:12911-12930. [PMID: 37523859 PMCID: PMC10544029 DOI: 10.1021/acs.jmedchem.3c00849] [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: 05/11/2023] [Indexed: 08/02/2023]
Abstract
The inhibition of CXC chemokine receptor 2 (CXCR2), a key inflammatory mediator, is a potential strategy in the treatment of several pulmonary diseases and cancers. The complexity of endogenous chemokine interaction with the orthosteric binding site has led to the development of CXCR2 negative allosteric modulators (NAMs) targeting an intracellular pocket near the G protein binding site. Our understanding of NAM binding and mode of action has been limited by the availability of suitable tracer ligands for competition studies, allowing direct ligand binding measurements. Here, we report the rational design, synthesis, and pharmacological evaluation of a series of fluorescent NAMs, based on navarixin (2), which display high affinity and preferential binding for CXCR2 over CXCR1. We demonstrate their application in fluorescence imaging and NanoBRET binding assays, in whole cells or membranes, capable of kinetic and equilibrium analysis of NAM binding, providing a platform to screen for alternative chemophores targeting these receptors.
Collapse
Affiliation(s)
- Bianca
Maria Casella
- Division
of Biomolecular Sciences and Medicinal Chemistry, School of Pharmacy, University of Nottingham Biodiscovery Institute, Nottingham NG7 2RD, UK
| | - James P. Farmer
- Division
of Physiology, Pharmacology & Neuroscience, Medical School, School
of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Desislava N. Nesheva
- Division
of Physiology, Pharmacology & Neuroscience, Medical School, School
of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Huw E. L. Williams
- School
of Chemistry, University of Nottingham Biodiscovery
Institute, Nottingham NG7 2RD, UK
| | - Steven J. Charlton
- Division
of Physiology, Pharmacology & Neuroscience, Medical School, School
of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
- OMass
Therapeutics Ltd., Oxford OX4 2GX, UK
| | - Nicholas D. Holliday
- Division
of Physiology, Pharmacology & Neuroscience, Medical School, School
of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
- Excellerate
Bioscience Ltd., Biocity, University of
Nottingham, Nottingham NG1 1GF, UK
| | - Charles A. Laughton
- Division
of Biomolecular Sciences and Medicinal Chemistry, School of Pharmacy, University of Nottingham Biodiscovery Institute, Nottingham NG7 2RD, UK
| | - Shailesh N. Mistry
- Division
of Biomolecular Sciences and Medicinal Chemistry, School of Pharmacy, University of Nottingham Biodiscovery Institute, Nottingham NG7 2RD, UK
| |
Collapse
|
3
|
Våbenø J, Oliva-Santiago M, Jørgensen AS, Karlshøj S, Rosenkilde MM. Identification of a Salt Bridge That Is Functionally Important for Chemokine Receptor CXCR1 but not CXCR2. ACS Pharmacol Transl Sci 2023; 6:1120-1128. [PMID: 37588755 PMCID: PMC10425996 DOI: 10.1021/acsptsci.3c00070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Indexed: 08/18/2023]
Abstract
CXC chemokine receptors 1 (CXCR1) and 2 (CXCR2) have high sequence similarity and overlapping chemokine ligand profiles. Residue positions 3.32 and 7.39 are critical for signal transduction in the related CXCR4, and in these positions CXCR1 and CXCR2 contain oppositely charged residues (Lys3.32 and Glu7.39). Experimental and computed receptor structures reveal the possible formation of a salt bridge between transmembrane (TM) helices 3 and 7 via these two residues. To investigate the functional importance of Lys1173.32 and Glu2917.39 in CXCR1, along with the flanking Glu1183.33, we performed a signaling study on 16 CXCR1 mutants using two different CXCL8 isoforms. While single Ala-mutation (K1173.32A, E2917.39A) and charge reversal (K1173.32E, E2917.39K) resulted in nonfunctional receptors, double (K1173.32E-E2917.39K) and triple (K1173.32E-E1183.33A-E2917.39K) mutants rescued CXCR1 function. In contrast, the corresponding mutations did not affect the CXCR2 function to the same extent. Our findings show that the Lys3.32-Glu7.39 salt bridge between TM3 and -7 is functionally important for CXCR1 but not for CXCR2, meaning that signal transduction for these highly homologous receptors is not conserved.
Collapse
Affiliation(s)
- Jon Våbenø
- Helgeland
Hospital Trust, Prestmarkveien
1, 8800 Sandnessjøen, Norway
| | - Marta Oliva-Santiago
- Laboratory
for Molecular Pharmacology, Department of Biomedical Sciences, Faculty
of Health and Medical Sciences, University
of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Astrid S. Jørgensen
- Laboratory
for Molecular Pharmacology, Department of Biomedical Sciences, Faculty
of Health and Medical Sciences, University
of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Stefanie Karlshøj
- Laboratory
for Molecular Pharmacology, Department of Biomedical Sciences, Faculty
of Health and Medical Sciences, University
of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Mette M. Rosenkilde
- Laboratory
for Molecular Pharmacology, Department of Biomedical Sciences, Faculty
of Health and Medical Sciences, University
of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| |
Collapse
|
4
|
Arimont M, Hoffmann C, de Graaf C, Leurs R. Chemokine Receptor Crystal Structures: What Can Be Learned from Them? Mol Pharmacol 2019; 96:765-777. [PMID: 31266800 DOI: 10.1124/mol.119.117168] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/21/2019] [Indexed: 12/18/2022] Open
Abstract
Chemokine receptors belong to the class A of G protein-coupled receptors (GPCRs) and are implicated in a wide variety of physiologic functions, mostly related to the homeostasis of the immune system. Chemokine receptors are also involved in multiple pathologic processes, including immune and autoimmune diseases, as well as cancer. Hence, several members of this GPCR subfamily are considered to be very relevant therapeutic targets. Since drug discovery efforts can be significantly reinforced by the availability of crystal structures, substantial efforts in the area of chemokine receptor structural biology could dramatically increase the outcome of drug discovery campaigns. This short review summarizes the available data on chemokine receptor crystal structures, discusses the numerous applications from chemokine receptor structures that can enhance the daily work of molecular pharmacologists, and describes the challenges and pitfalls to consider when relying on crystal structures for further research applications. SIGNIFICANCE STATEMENT: This short review summarizes the available data on chemokine receptor crystal structures, discusses the numerous applications from chemokine receptor structures that can enhance the daily work of molecular pharmacologists, and describes the challenges and pitfalls to consider when relying on crystal structures for further research applications.
Collapse
Affiliation(s)
- Marta Arimont
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., R.L.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University, Jena, Germany (C.H.); and Sosei Heptares, Great Abington, Cambridge, United Kingdom (C.d.G.)
| | - Carsten Hoffmann
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., R.L.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University, Jena, Germany (C.H.); and Sosei Heptares, Great Abington, Cambridge, United Kingdom (C.d.G.)
| | - Chris de Graaf
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., R.L.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University, Jena, Germany (C.H.); and Sosei Heptares, Great Abington, Cambridge, United Kingdom (C.d.G.)
| | - Rob Leurs
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., R.L.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University, Jena, Germany (C.H.); and Sosei Heptares, Great Abington, Cambridge, United Kingdom (C.d.G.)
| |
Collapse
|
5
|
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
|
6
|
Targeting CXCR1/2: The medicinal potential as cancer immunotherapy agents, antagonists research highlights and challenges ahead. Eur J Med Chem 2019; 185:111853. [PMID: 31732253 DOI: 10.1016/j.ejmech.2019.111853] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/05/2019] [Accepted: 11/04/2019] [Indexed: 12/11/2022]
Abstract
Immune suppression in the tumor microenvironment (TME) is an intractable issue in anti-cancer immunotherapy. The chemokine receptors CXCR1 and CXCR2 recruit immune suppressive cells such as the myeloid derived suppressor cells (MDSCs) to the TME. Therefore, CXCR1/2 antagonists have aroused pharmaceutical interest in recent years. In this review, the medicinal chemistry of CXCR1/2 antagonists and their relevance in cancer immunotherapy have been summarized. The development of the drug candidates, along with their design rationale, clinical status and current challenges have also been discussed.
Collapse
|
7
|
Arimont M, Sun SL, Leurs R, Smit M, de Esch IJP, de Graaf C. Structural Analysis of Chemokine Receptor-Ligand Interactions. J Med Chem 2017; 60:4735-4779. [PMID: 28165741 PMCID: PMC5483895 DOI: 10.1021/acs.jmedchem.6b01309] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
This
review focuses on the construction and application of structural chemokine
receptor models for the elucidation of molecular determinants of chemokine
receptor modulation and the structure-based discovery and design of
chemokine receptor ligands. A comparative analysis of ligand binding
pockets in chemokine receptors is presented, including a detailed
description of the CXCR4, CCR2, CCR5, CCR9, and US28 X-ray structures,
and their implication for modeling molecular interactions of chemokine
receptors with small-molecule ligands, peptide ligands, and large
antibodies and chemokines. These studies demonstrate how the integration
of new structural information on chemokine receptors with extensive
structure–activity relationship and site-directed mutagenesis
data facilitates the prediction of the structure of chemokine receptor–ligand
complexes that have not been crystallized. Finally, a review of structure-based
ligand discovery and design studies based on chemokine receptor crystal
structures and homology models illustrates the possibilities and challenges
to find novel ligands for chemokine receptors.
Collapse
Affiliation(s)
- Marta Arimont
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Shan-Liang Sun
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Rob Leurs
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Martine Smit
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Chris de Graaf
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
8
|
Feng Z, Pearce LV, Zhang Y, Xing C, Herold BKA, Ma S, Hu Z, Turcios NA, Yang P, Tong Q, McCall AK, Blumberg PM, Xie XQ. Multi-Functional Diarylurea Small Molecule Inhibitors of TRPV1 with Therapeutic Potential for Neuroinflammation. AAPS J 2016; 18:898-913. [PMID: 27000851 PMCID: PMC5333490 DOI: 10.1208/s12248-016-9888-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/10/2016] [Indexed: 01/05/2023] Open
Abstract
Transient receptor potential vanilloid type 1 (TRPV1), a heat-sensitive calcium channel protein, contributes to inflammation as well as to acute and persistent pain. Since TRPV1 occupies a central position in pathways of neuronal inflammatory signaling, it represents a highly attractive potential therapeutic target for neuroinflammation. In the present work, we have in silico identified a series of diarylurea analogues for hTRPV1, of which 11 compounds showed activity in the nanomolar to micromolar range as validated by in vitro biological assays. Then, we utilized molecular docking to explore the detailed interactions between TRPV1 and the compounds to understand the contributions of the different substituent groups. Tyr511, Leu518, Leu547, Thr550, Asn551, Arg557, and Leu670 were important for the recognition of the small molecules by TRPV1. A hydrophobic group in R2 or a polar/hydrophilic group in R1 contributed significantly to the activities of the antagonists at TRPV1. In addition, the subtle different binding pose of meta-chloro in place of para-fluoro in the R2 group converted antagonism into partial agonism, as was predicted by our short-term molecular dynamics (MD) simulation and validated by bioassay. Importantly, compound 15, one of our best TRPV1 inhibitors, also showed potential binding affinity (1.39 μM) at cannabinoid receptor 2 (CB2), which is another attractive target for immuno-inflammation diseases. Furthermore, compound 1 and its diarylurea analogues were predicted to target the C-X-C chemokine receptor 2 (CXCR2), although bioassay validation of CXCR2 with these compounds still needs to be performed. This prediction from the modeling is of interest, since CXCR2 is also a potential therapeutic target for chronic inflammatory diseases. Our findings provide novel strategies to develop a small molecule inhibitor to simultaneously target two or more inflammation-related proteins for the treatment of a wide range of inflammatory disorders including neuroinflammation and neurodegenerative diseases with potential synergistic effect.
Collapse
Affiliation(s)
- Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Larry V Pearce
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Yu Zhang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Changrui Xing
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Brienna K A Herold
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Shifan Ma
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Ziheng Hu
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Noe A Turcios
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Peng Yang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Qin Tong
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Anna K McCall
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Peter M Blumberg
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, 20892, USA.
- Laboratory of Cancer Biology and Genetics, National Institutes of Health, Building 37, Room 4048B, 37 Convent Drive MSC 4255, Bethesda, Maryland, 20892-4255, USA.
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.
- NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.
- Departments of Computational Biology and of Structural Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA.
| |
Collapse
|
9
|
Grundmann M, Tikhonova IG, Hudson BD, Smith NJ, Mohr K, Ulven T, Milligan G, Kenakin T, Kostenis E. A Molecular Mechanism for Sequential Activation of a G Protein-Coupled Receptor. Cell Chem Biol 2016; 23:392-403. [PMID: 26991104 DOI: 10.1016/j.chembiol.2016.02.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 02/11/2016] [Accepted: 02/22/2016] [Indexed: 01/05/2023]
Abstract
Ligands targeting G protein-coupled receptors (GPCRs) are currently classified as either orthosteric, allosteric, or dualsteric/bitopic. Here, we introduce a new pharmacological concept for GPCR functional modulation: sequential receptor activation. A hallmark feature of this is a stepwise ligand binding mode with transient activation of a first receptor site followed by sustained activation of a second topographically distinct site. We identify 4-CMTB (2-(4-chlorophenyl)-3-methyl-N-(thiazol-2-yl)butanamide), previously classified as a pure allosteric agonist of the free fatty acid receptor 2, as the first sequential activator and corroborate its two-step activation in living cells by tracking integrated responses with innovative label-free biosensors that visualize multiple signaling inputs in real time. We validate this unique pharmacology with traditional cellular readouts, including mutational and pharmacological perturbations along with computational methods, and propose a kinetic model applicable to the analysis of sequential receptor activation. We envision this form of dynamic agonism as a common principle of nature to spatiotemporally encode cellular information.
Collapse
Affiliation(s)
- Manuel Grundmann
- Molecular-, Cellular- and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany.
| | - Irina G Tikhonova
- Molecular Therapeutics, School of Pharmacy, Medical Biology Centre, Queen's University, Belfast BT7 1NN Northern Ireland
| | - Brian D Hudson
- Molecular Pharmacology Group, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ Scotland
| | - Nicola J Smith
- Molecular Pharmacology Group, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ Scotland; Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW 2010, Australia
| | - Klaus Mohr
- Pharmacology and Toxicology, University of Bonn, 53347 Bonn, Germany
| | - Trond Ulven
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5230 Odense M, Denmark
| | - Graeme Milligan
- Molecular Pharmacology Group, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ Scotland
| | - Terry Kenakin
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Evi Kostenis
- Molecular-, Cellular- and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany.
| |
Collapse
|
10
|
Maeda DY, Peck AM, Schuler AD, Quinn MT, Kirpotina LN, Wicomb WN, Auten RL, Gundla R, Zebala JA. Boronic acid-containing CXCR1/2 antagonists: Optimization of metabolic stability, in vivo evaluation, and a proposed receptor binding model. Bioorg Med Chem Lett 2015; 25:2280-4. [PMID: 25933594 PMCID: PMC4430358 DOI: 10.1016/j.bmcl.2015.04.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/10/2015] [Accepted: 04/13/2015] [Indexed: 12/14/2022]
Abstract
Blockade of undesired neutrophil migration to sites of inflammation remains an area of substantial pharmaceutical interest. To effect this blockade, a validated therapeutic target is antagonism of the chemokine receptor CXCR2. Herein we report the discovery of 6-(2-boronic acid-5-trifluoromethoxy-benzylsulfanyl)-N-(4-fluoro-phenyl)-nicotinamide 6, an antagonist with activity at both CXCR1 and CXCR2 receptors (IC50 values 31 and 21 nM, respectively). Compound 6 exhibited potent inhibition of neutrophil influx in a rat model of pulmonary inflammation, and is hypothesized to interact with a unique intracellular binding site on CXCR2. Compound 6 (SX-576) is undergoing further investigation as a potential therapy for pulmonary inflammation.
Collapse
Affiliation(s)
- Dean Y Maeda
- Syntrix Biosystems, 215 Clay Street Northwest, Suite B5, Auburn, WA 98001, United States.
| | - Angela M Peck
- Syntrix Biosystems, 215 Clay Street Northwest, Suite B5, Auburn, WA 98001, United States
| | - Aaron D Schuler
- Syntrix Biosystems, 215 Clay Street Northwest, Suite B5, Auburn, WA 98001, United States
| | - Mark T Quinn
- Department of Microbiology and Immunology, Montana State University, 960 Technology Boulevard, Bozeman, MT 59717, United States
| | - Liliya N Kirpotina
- Department of Microbiology and Immunology, Montana State University, 960 Technology Boulevard, Bozeman, MT 59717, United States
| | - Winston N Wicomb
- Infectious Disease Research Institute, 1616 Eastlake Avenue East, Seattle, WA 98102, United States
| | - Richard L Auten
- Division of Neonatal Medicine, Department of Pediatrics, Duke University Medical Center, 366 Sands Research Drive, Durham, NC 27710, United States
| | - Rambabu Gundla
- Integrated Drug Discovery Services, GVK Biosciences Private Limited, IDA Nacharam, Hyderabad 500 076, India
| | - John A Zebala
- Syntrix Biosystems, 215 Clay Street Northwest, Suite B5, Auburn, WA 98001, United States
| |
Collapse
|
11
|
Xu L, Li Y, Li D, Xu P, Tian S, Sun H, Liu H, Hou T. Exploring the binding mechanisms of MIF to CXCR2 using theoretical approaches. Phys Chem Chem Phys 2014; 17:3370-82. [PMID: 25526079 DOI: 10.1039/c4cp05095a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a multi-functional protein that acts as a cytokine and as an enzyme. Recently, MIF was identified as a non-canonical ligand of G protein-coupled chemokine receptor CXCR2 with low nanomolar affinity in leukocyte arrest and chemotaxis, but the precise knowledge of the molecular determinants of the MIF-CXCR2 interface has remained unknown. Therefore, we employed homology modeling, protein-protein docking, molecular dynamics (MD) simulations, Molecular Mechanics/Generalized Born Surface Area (MM/GBSA) binding free energy calculations and MM/GBSA binding free energy decomposition to obtain insights into the molecular recognition of MIF with CXCR2. The predicted binding pattern of MIF-CXCR2 is in good agreement with the experimental data and sheds light on the functional role of important MIF-CXCR2 interface residues in association with binding and signaling. According to our predictions, the R11A/D44A double mutations of MIF exhibit a pronounced defect in the binding affinity of MIF to CXCR2, resulting in large conformational changes. The potential two-site binding model for the MIF-CXCR2 recognition was proposed: initialized primarily by the non-polar interactions including the van der Waals and hydrophobic interactions, the N-terminal region of CXCR2 contacts the N-like loop and β-sheet of MIF (site 1), and then the ECL2 and ECL3 regions of CXCR2 form strong interactions with the pseudo-(E)LR motif and C-terminus of MIF, which induces the molecular thermodynamic motion of TMs for signal transduction (site 2). This study will extend our understanding to the binding mechanisms of MIF to CXCR2 and provide useful information for the rational design of potent inhibitors selectively targeting the MIF-CXCR2 interactions.
Collapse
Affiliation(s)
- Lei Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Ha H, Bensman T, Ho H, Beringer PM, Neamati N. A novel phenylcyclohex-1-enecarbothioamide derivative inhibits CXCL8-mediated chemotaxis through selective regulation of CXCR2-mediated signalling. Br J Pharmacol 2014; 171:1551-65. [PMID: 24354854 DOI: 10.1111/bph.12547] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 11/26/2013] [Accepted: 12/02/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Since the CXC chemokine receptor CXCR2 and its cognate ligand CXCL8 (IL-8) critically regulate neutrophil trafficking during inflammation, they have been implicated in a number of inflammatory lung diseases. Several CXCR2 antagonists have been described and the blockade of CXCR2 has shown promise in pre-clinical disease models and early clinical trials. However, given its potential, there are fewer distinct classes of antagonists of CXCR2 than of other clinically relevant molecular targets. Thus, we sought to identify additional classes of compounds that alter CXCR2 function. EXPERIMENTAL APPROACH We used the CXCR2 Tango(TM) assay to screen an in-house library of highly diverse chemical compounds. CX4338 [2-(benzylamino)-4,4-dimethyl-6-oxo-N-phenylcyclohex-1-enecarbothioamide] was identified from our screen and additional studies to characterize the compound were performed. Receptor internalization and second-messenger assays were used to assess the effects of CX4338 on CXCR2-mediated signalling. Wound healing, transwell cell migration and LPS-induced lung inflammation in mice were used to determine the in vitro and in vivo effects of CX4338. KEY RESULTS CX4338 selectively inhibited CXCR2-mediated recruitment of β-arrestin-2 and receptor internalization, while enhancing CXCR2-mediated MAPK activation. Additionally, CX4338 inhibited CXCL8-induced chemotaxis in CXCR2-overexpressing cells and human neutrophils. In vivo, CX4338 significantly reduced neutrophils in bronchoalveolar lavage induced by LPS in mice. CONCLUSIONS AND IMPLICATIONS A novel compound CX4338 inhibited CXCR2-mediated cell migration with a mechanism of action not previously reported. Also, selective inhibition of CXCR2-mediated β-arrestin-2 activation is sufficient to inhibit CXCL8-mediated chemotaxis.
Collapse
Affiliation(s)
- Helen Ha
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | |
Collapse
|
13
|
Role of 3D Structures in Understanding, Predicting, and Designing Molecular Interactions in the Chemokine Receptor Family. ACTA ACUST UNITED AC 2014. [DOI: 10.1007/7355_2014_77] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
14
|
Maeda DY, Peck AM, Schuler A, Quinn MT, Kirpotina LN, Wicomb WN, Fan GH, Zebala JA. Discovery of 2-[5-(4-Fluorophenylcarbamoyl)pyridin-2-ylsulfanylmethyl]phenylboronic Acid (SX-517): Noncompetitive Boronic Acid Antagonist of CXCR1 and CXCR2. J Med Chem 2014; 57:8378-97. [PMID: 25254640 PMCID: PMC4207547 DOI: 10.1021/jm500827t] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Indexed: 12/15/2022]
Abstract
The G protein-coupled chemokine receptors CXCR1 and CXCR2 play key roles in inflammatory diseases and carcinogenesis. In inflammation, they activate and recruit polymorphonuclear cells (PMNs) through binding of the chemokines CXCL1 (CXCR1) and CXCL8 (CXCR1 and CXCR2). Structure-activity studies that examined the effect of a novel series of S-substituted 6-mercapto-N-phenyl-nicotinamides on CXCL1-stimulated Ca(2+) flux in whole human PMNs led to the discovery of 2-[5-(4-fluorophenylcarbamoyl)pyridin-2-ylsulfanylmethyl]phenylboronic acid (SX-517), a potent noncompetitive boronic acid CXCR1/2 antagonist. SX-517 inhibited CXCL1-induced Ca(2+) flux (IC50 = 38 nM) in human PMNs but had no effect on the Ca(2+) flux induced by C5a, fMLF, or PAF. In recombinant HEK293 cells that stably expressed CXCR2, SX-517 antagonized CXCL8-induced [(35)S]GTPγS binding (IC50 = 60 nM) and ERK1/2 phosphorylation. Inhibition was noncompetitive, with SX-517 unable to compete the binding of [(125)I]-CXCL8 to CXCR2 membranes. SX-517 (0.2 mg/kg iv) significantly inhibited inflammation in an in vivo murine model. SX-517 is the first reported boronic acid chemokine antagonist and represents a novel pharmacophore for CXCR1/2 antagonism.
Collapse
Affiliation(s)
- Dean Y. Maeda
- Syntrix
Biosystems, 215 Clay
Street, Auburn, Washington 98001, United States
| | - Angela M. Peck
- Syntrix
Biosystems, 215 Clay
Street, Auburn, Washington 98001, United States
| | - Aaron
D. Schuler
- Syntrix
Biosystems, 215 Clay
Street, Auburn, Washington 98001, United States
| | - Mark T. Quinn
- Department
of Microbiology and Immunology, Montana
State University, 960
Technology Boulevard, Bozeman, Montana 59717, United States
| | - Liliya N. Kirpotina
- Department
of Microbiology and Immunology, Montana
State University, 960
Technology Boulevard, Bozeman, Montana 59717, United States
| | - Winston N. Wicomb
- Infectious
Disease Research Institute, 1616 Eastlake Avenue East, Seattle, Washington 98102, United States
| | - Guo-Huang Fan
- Department
of Pharmacology, Meharry Medical College, 1005 Dr. DB Todd Boulevard, Nashville, Tennessee 37208, United States
| | - John A. Zebala
- Syntrix
Biosystems, 215 Clay
Street, Auburn, Washington 98001, United States
| |
Collapse
|
15
|
Christopoulos A, Changeux JP, Catterall WA, Fabbro D, Burris TP, Cidlowski JA, Olsen RW, Peters JA, Neubig RR, Pin JP, Sexton PM, Kenakin TP, Ehlert FJ, Spedding M, Langmead CJ. International Union of Basic and Clinical Pharmacology. XC. multisite pharmacology: recommendations for the nomenclature of receptor allosterism and allosteric ligands. Pharmacol Rev 2014; 66:918-47. [PMID: 25026896 PMCID: PMC11060431 DOI: 10.1124/pr.114.008862] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Allosteric interactions play vital roles in metabolic processes and signal transduction and, more recently, have become the focus of numerous pharmacological studies because of the potential for discovering more target-selective chemical probes and therapeutic agents. In addition to classic early studies on enzymes, there are now examples of small molecule allosteric modulators for all superfamilies of receptors encoded by the genome, including ligand- and voltage-gated ion channels, G protein-coupled receptors, nuclear hormone receptors, and receptor tyrosine kinases. As a consequence, a vast array of pharmacologic behaviors has been ascribed to allosteric ligands that can vary in a target-, ligand-, and cell-/tissue-dependent manner. The current article presents an overview of allostery as applied to receptor families and approaches for detecting and validating allosteric interactions and gives recommendations for the nomenclature of allosteric ligands and their properties.
Collapse
Affiliation(s)
- Arthur Christopoulos
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Jean-Pierre Changeux
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - William A Catterall
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Doriano Fabbro
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Thomas P Burris
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - John A Cidlowski
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Richard W Olsen
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - John A Peters
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Richard R Neubig
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Jean-Philippe Pin
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Patrick M Sexton
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Terry P Kenakin
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Frederick J Ehlert
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Michael Spedding
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Christopher J Langmead
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| |
Collapse
|
16
|
Roumen L, Scholten DJ, de Kruijf P, de Esch IJP, Leurs R, de Graaf C. C(X)CR in silico: Computer-aided prediction of chemokine receptor-ligand interactions. DRUG DISCOVERY TODAY. TECHNOLOGIES 2014; 9:e281-91. [PMID: 24990665 DOI: 10.1016/j.ddtec.2012.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
This review will focus on the construction, refinement, and validation of chemokine receptor models for the purpose of structure-based virtual screening and ligand design. The review will present a comparative analysis of ligand binding pockets in chemokine receptors, including a review of the recently released CXCR4 X-ray structures, and their implication on chemokine receptor (homology) modeling. The recommended protein-ligand modeling procedure as well as the use of experimental anchors to steer the modeling procedure is discussed and an overview of several successful structure-based ligand discovery and design studies is provided. This review shows that receptor models, despite structural inaccuracies, can be efficiently used to find novel ligands for chemokine receptors.:
Collapse
Affiliation(s)
- L Roumen
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - D J Scholten
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - P de Kruijf
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - I J P de Esch
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - R Leurs
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - C de Graaf
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Ha H, Neamati N. Pyrimidine-based compounds modulate CXCR2-mediated signaling and receptor turnover. Mol Pharm 2014; 11:2431-41. [PMID: 24896229 DOI: 10.1021/mp500180e] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Chemokine receptor CXCR2 is expressed on various immune cells and is essential for neutrophil recruitment and angiogenesis at sites of acute and chronic inflammation caused by tissue injury or infection. Because of its role in inflammation, it has been implicated in a number of immune-mediated inflammatory diseases such as psoriasis, arthritis, COPD, cystic fibrosis, asthma, and various types of cancer. CXCR2 and its ligands are up-regulated in cancer cells as well as the tumor microenvironment, promoting tumor growth, angiogenesis, and invasiveness. Although pharmaceutical companies have pursued the development of CXCR2-specific small-molecule inhibitors as anti-inflammatory agents within the last decades, there are currently no clinically approved CXCR2 inhibitors. Using a high-throughput, cell-based assay specific for CXCR2, we screened an in-house library of structurally diverse compounds and identified a class of pyrimidine-based compounds that alter CXCR2-mediated second messenger signaling. Our lead compound, CX797, inhibited IL8-mediated cAMP signaling and receptor degradation while specifically up-regulating IL8-mediated β-arrestin-2 recruitment. CX797 also inhibited IL8-mediated cell migration. Mechanistic comparison of CX797 and a previously reported CXCR2 inhibitor, SB265610, show these two classes of compounds have a distinct mechanism of action on CXCR2.
Collapse
Affiliation(s)
- Helen Ha
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California , 1985 Zonal Avenue, Los Angeles, California 90033, United States
| | | |
Collapse
|
18
|
Kredel S, Wolff M, Gierschik P, Heilker R. Phenotypic analysis of chemokine-driven actin reorganization in primary human neutrophils. Assay Drug Dev Technol 2014; 12:120-8. [PMID: 24579814 DOI: 10.1089/adt.2013.553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The chemokine-driven activation of CXC-type chemokine receptors 1/2 (CXCR1/2) and the subsequent reorganization of the neutrophilic actin are early key events in the induction of neutrophil migration toward centers of inflammation. In this study, an image analysis algorithm was developed to detect subtle chemokine-induced changes in the actin cytoskeleton of primary human neutrophils. By this means, a discrete early step of neutrophil activation was dissected that could be initiated by concentrations of growth-related oncogen α (Gro-α) or interleukin-8 (IL-8) just above their resting-state plasma levels. The associated half-maximal effective concentration (EC50) values for Gro-α and IL-8 of 8 and 22 pM, respectively, are between two and three orders of magnitude below the so-far reported EC50 values of these chemokines for the induction of neutrophilic calcium release, integrin expression, degranulation, and receptor internalization. Sch527123, a known inhibitor of CXCR2 (KD=49 pM) and with a lower potency/affinity also of CXCR1 (KD=3.9 nM), antagonized actin remodeling with half-maximal inhibitory concentration (IC50) values of 400 pM for the CXCR2-specific agonist Gro-α and of 36 nM for the CXCR1/2-promiscuous agonist IL-8. This observation indicates that the here-described early step of chemokine-driven actin reorganization is modulated by both CXCR1 and CXCR2. Thus, the imaging-based assay format, as developed in this work, may be employed in a phenotypic screening campaign to identify inhibitors of an early step in CXCR1/2-induced neutrophilic chemotaxis.
Collapse
Affiliation(s)
- Simone Kredel
- 1 Institute of Pharmacology and Toxicology, Ulm University Medical Center , Ulm, Germany
| | | | | | | |
Collapse
|
19
|
Exploring the CXCR3 Chemokine Receptor with Small-Molecule Antagonists and Agonists. TOPICS IN MEDICINAL CHEMISTRY 2014. [DOI: 10.1007/7355_2014_75] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
20
|
Chemokine receptor modeling: an interdisciplinary approach to drug design. Future Med Chem 2014; 6:91-114. [DOI: 10.4155/fmc.13.194] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chemokines and their receptors are integral components of the immune response, regulating lymphocyte development, homing and trafficking, and playing a key role in the pathophysiology of many diseases. Chemokine receptors have, therefore, become the target for both small-molecule, peptide and antibody therapeutics. Chemokine receptors belong to the family of seven transmembrane receptor class A G protein-coupled receptors. The publication of the crystal structure of the archetypal class A seven transmembrane receptor protein rhodopsin, and other G protein-coupled receptors, including C-X-C chemokine receptor 4 and C-C chemokine receptor 5, provided the opportunity to create homology models of chemokine receptors. In this review, we describe an interdisciplinary approach to chemokine receptor modeling and the utility of this approach for structure-based drug design of chemokine receptor inhibitors.
Collapse
|
21
|
Scholten DJ, Roumen L, Wijtmans M, Verkade-Vreeker MCA, Custers H, Lai M, de Hooge D, Canals M, de Esch IJP, Smit MJ, de Graaf C, Leurs R. Identification of overlapping but differential binding sites for the high-affinity CXCR3 antagonists NBI-74330 and VUF11211. Mol Pharmacol 2014; 85:116-26. [PMID: 24174496 DOI: 10.1124/mol.113.088633] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
CXC chemokine receptor CXCR3 and/or its main three ligands CXCL9, CXCL10, and CXCL11 are highly upregulated in a variety of diseases. As such, considerable efforts have been made to develop small-molecule receptor CXCR3 antagonists, yielding distinct chemical classes of antagonists blocking binding and/or function of CXCR3 chemokines. Although it is suggested that these compounds bind in an allosteric fashion, thus far no evidence has been provided regarding the molecular details of their interaction with CXCR3. Using site-directed mutagenesis complemented with in silico homology modeling, we report the binding modes of two high-affinity CXCR3 antagonists of distinct chemotypes: VUF11211 [(S)-5-chloro-6-(4-(1-(4-chlorobenzyl)piperidin-4-yl)-3-ethylpiperazin-1-yl)-N-ethylnicotinamide] (piperazinyl-piperidine) with a rigid elongated structure containing two basic groups and NBI-74330 [(R)-N-(1-(3-(4-ethoxyphenyl)-4-oxo-3,4-dihydropyrido[2,3-d]pyrimidin-2-yl)ethyl)-2-(4-fluoro-3-(trifluoromethyl)phenyl)-N-(pyridin-3-ylmethyl)acetamide] (8-azaquinazolinone) without any basic group. Here we show that NBI-74330 is anchored in the transmembrane minor pocket lined by helices 2 (W2.60, D2.63), 3 (F3.32), and 7 (S7.39, Y7.43), whereas VUF11211 extends from the minor pocket into the major pocket of the transmembrane domains, located between residues in helices 1 (Y1.39), 2 (W2.60), 3 (F3.32), 4 (D4.60), 6 (Y6.51), and 7 (S7.39, Y7.43). Mutation of these residues did not affect CXCL11 binding significantly, confirming the allosteric nature of the interaction of these small molecules with CXCR3. Moreover, the model derived from our in silico-guided studies fits well with the already published structure-activity relationship data on these ligands. Altogether, in this study, we show overlapping, yet different binding sites for two high-affinity CXCR3 antagonists, which offer new opportunities for the structure-based design of allosteric modulators for CXCR3.
Collapse
Affiliation(s)
- Danny J Scholten
- Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines, and Systems, VU University Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Raccosta L, Fontana R, Maggioni D, Lanterna C, Villablanca EJ, Paniccia A, Musumeci A, Chiricozzi E, Trincavelli ML, Daniele S, Martini C, Gustafsson JA, Doglioni C, Feo SG, Leiva A, Ciampa MG, Mauri L, Sensi C, Prinetti A, Eberini I, Mora JR, Bordignon C, Steffensen KR, Sonnino S, Sozzani S, Traversari C, Russo V. The oxysterol-CXCR2 axis plays a key role in the recruitment of tumor-promoting neutrophils. J Exp Med 2013; 210:1711-28. [PMID: 23897983 PMCID: PMC3754872 DOI: 10.1084/jem.20130440] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 06/21/2013] [Indexed: 12/19/2022] Open
Abstract
Tumor-infiltrating immune cells can be conditioned by molecules released within the microenvironment to thwart antitumor immune responses, thereby facilitating tumor growth. Among immune cells, neutrophils play an important protumorigenic role by favoring neoangiogenesis and/or by suppressing antitumor immune responses. Tumor-derived oxysterols have recently been shown to favor tumor growth by inhibiting dendritic cell migration toward lymphoid organs. We report that tumor-derived oxysterols recruit protumor neutrophils in a liver X receptor (LXR)-independent, CXCR2-dependent manner, thus favoring tumor growth by promoting neoangiogenesis and immunosuppression. We demonstrate that interfering with the oxysterol-CXCR2 axis delays tumor growth and prolongs the overall survival of tumor-bearing mice. These results identify an unanticipated protumor function of the oxysterol-CXCR2 axis and a possible target for cancer therapy.
Collapse
Affiliation(s)
- Laura Raccosta
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | - Raffaella Fontana
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | - Daniela Maggioni
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | - Claudia Lanterna
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | | | - Aida Paniccia
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | - Andrea Musumeci
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | - Elena Chiricozzi
- Department of Medical Chemistry, Biochemistry and Biotechnology, Center of Excellence on Neurodegenerative Diseases, University of Milan, 20090 Segrate, Italy
| | | | - Simona Daniele
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Jan-Ake Gustafsson
- Department of Biosciences and Nutrition, Karolinska Institute, S-14183 Huddinge, Sweden
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204
| | - Claudio Doglioni
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
- Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | | | - Andrea Leiva
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| | - Maria Grazia Ciampa
- Department of Medical Chemistry, Biochemistry and Biotechnology, Center of Excellence on Neurodegenerative Diseases, University of Milan, 20090 Segrate, Italy
| | - Laura Mauri
- Department of Medical Chemistry, Biochemistry and Biotechnology, Center of Excellence on Neurodegenerative Diseases, University of Milan, 20090 Segrate, Italy
| | - Cristina Sensi
- Proteomics and Protein Structure Study Group, Department of Pharmacological Sciences, University of Milan, 20133 Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Chemistry, Biochemistry and Biotechnology, Center of Excellence on Neurodegenerative Diseases, University of Milan, 20090 Segrate, Italy
| | - Ivano Eberini
- Proteomics and Protein Structure Study Group, Department of Pharmacological Sciences, University of Milan, 20133 Milan, Italy
| | - J. Rodrigo Mora
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA 02114
| | - Claudio Bordignon
- Università Vita-Salute San Raffaele, 20132 Milan, Italy
- MolMed S.p.A., 20132 Milan, Italy
| | - Knut R. Steffensen
- Department of Biosciences and Nutrition, Karolinska Institute, S-14183 Huddinge, Sweden
| | - Sandro Sonnino
- Department of Medical Chemistry, Biochemistry and Biotechnology, Center of Excellence on Neurodegenerative Diseases, University of Milan, 20090 Segrate, Italy
| | - Silvano Sozzani
- Humanitas Clinical and Research Center, 20089 Rozzano, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | | | - Vincenzo Russo
- Cancer Gene Therapy Unit, Program of Immunology and Bio Immuno Gene Therapy of Cancer, Division of Molecular Oncology, and Department of Pathology, Scientific Institute San Raffaele, 20132 Milan, Italy
| |
Collapse
|
23
|
Carter PH. Progress in the discovery of CC chemokine receptor 2 antagonists, 2009 - 2012. Expert Opin Ther Pat 2013; 23:549-68. [PMID: 23428142 DOI: 10.1517/13543776.2013.771168] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION CC chemokine receptor 2 (CCR2) is a key mediator of the activation and migration of inflammatory monocytes. As such, it has been investigated extensively as a target for therapeutic intervention in a diverse range of diseases. AREAS COVERED This article reviews both the patent and peer-reviewed literature on the discovery of CCR2 antagonists from January 2009 to December 2012. Developments have occurred within each of the major chemical families of CCR2 antagonists, and are framed in that context. As has been true historically, a number of the compound families also exhibit substantial activity against the related CC chemokine receptor 5 (CCR5), making them formally CCR2/5-dual antagonists. EXPERT OPINION Significant progress continues to be made in identifying novel, potent CCR2 antagonists. In addition, researchers have had success in addressing issues related to selectivity, cardiac safety, and preclinical pharmacokinetics. Establishing proof-of-concept in clinical trials remains the primary challenge for the field.
Collapse
Affiliation(s)
- Percy H Carter
- Research & Development, Bristol-Myers Squibb Co., Princeton, NJ 08543, USA.
| |
Collapse
|
24
|
Klein MT, Vinson PN, Niswender CM. Approaches for probing allosteric interactions at 7 transmembrane spanning receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 115:1-59. [PMID: 23415091 PMCID: PMC5482179 DOI: 10.1016/b978-0-12-394587-7.00001-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In recent years, allosteric modulation of 7 transmembrane spanning receptors (7TMRs) has become a highly productive and exciting field of receptor pharmacology and drug discovery efforts. Positive and negative allosteric modulators (PAMs and NAMs, respectively) present a number of pharmacological and therapeutic advantages over conventional orthosteric ligands, including improved receptor-subtype selectivity, a lower propensity to induce receptor desensitization, the preservation of endogenous temporal and spatial activation of receptors, greater chemical flexibility for optimization of drug metabolism and pharmacokinetic parameters, and saturability of effect at target receptors, thus improving safety concerns and risk of overdose. Additionally, the relatively new concept of allosteric modulator-mediated receptor signal bias opens up a number of intriguing possibilities for PAMs, NAMs, and allosteric agonists, including the potential to selectively activate therapeutically beneficial signaling cascades, which could yield a superior tissue selectivity and side effect profile of allosteric modulators. However, there are a number of considerations and caveats that must be addressed when screening for and characterizing the properties of 7TMR allosteric modulators. Mode of pharmacology, methodology used to monitor receptor activity, detection of appropriate downstream analytes, selection of orthosteric probe, and assay time-course must all be considered when implementing any high-throughput screening campaign or when characterizing the properties of active compounds. Yet compared to conventional agonist/antagonist drug discovery programs, these elements of assay design are often a great deal more complicated when working with 7TMRs allosteric modulators. Moreover, for classical pharmacological methodologies and analyses, like radioligand binding and the assessment of compound affinity, the properties of allosteric modulators yield data that are more nuanced than orthosteric ligand-receptor interactions. In this review, we discuss the current methodologies being used to identify and characterize allosteric modulators, lending insight into the approaches that have been most successful in accurately and robustly identifying hit compounds. New label-free technologies capable of detecting phenotypic cellular changes in response to receptor activation are powerful tools well suited for assessing subtle or potentially masked cellular responses to allosteric modulation of 7TMRs. Allosteric modulator-induced receptor signal bias and the assay systems available to probe the various downstream signaling outcomes of receptor activation are also discussed.
Collapse
Affiliation(s)
- Michael T Klein
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | |
Collapse
|
25
|
|
26
|
Veenstra M, Ransohoff RM. Chemokine receptor CXCR2: physiology regulator and neuroinflammation controller? J Neuroimmunol 2012; 246:1-9. [PMID: 22445294 PMCID: PMC3335977 DOI: 10.1016/j.jneuroim.2012.02.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 02/23/2012] [Accepted: 02/25/2012] [Indexed: 01/05/2023]
Abstract
The innate immune system is a crucial component of inflammatory reactions, while the central nervous system (CNS) is the most vulnerable site of the body to inflammatory tissue injury. Neuroinflammatory brain pathologies are disorders in which the CNS is threatened by its own immune system. Chemokine receptor CXCR2 and its ligands have been implicated in several neuroinflammatory brain pathologies, as well as in neutrophil recruitment and in the developmental positioning of neural cells. This review focuses on the basics of CXCR2, its regulating role in bone marrow neutrophil recruitment, oligodendrocyte progenitor cell positioning and neural repair mechanisms, as well as its diverse roles in neuroinflammatory brain pathologies.
Collapse
Affiliation(s)
- Mike Veenstra
- Neuroinflammation Research Center, Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
- Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Richard M. Ransohoff
- Neuroinflammation Research Center, Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|