1
|
Comerford I, McColl SR. Atypical chemokine receptors in the immune system. Nat Rev Immunol 2024; 24:753-769. [PMID: 38714818 DOI: 10.1038/s41577-024-01025-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 05/10/2024]
Abstract
Leukocyte migration is a fundamental component of innate and adaptive immune responses as it governs the recruitment and localization of these motile cells, which is crucial for immune cell priming, effector functions, memory responses and immune regulation. This complex cellular trafficking system is controlled to a large extent via highly regulated production of secreted chemokines and the restricted expression of their membrane-tethered G-protein-coupled receptors. The activity of chemokines and their receptors is also regulated by a subfamily of molecules known as atypical chemokine receptors (ACKRs), which are chemokine receptor-like molecules that do not couple to the classical signalling pathways that promote cell migration in response to chemokine ligation. There has been a great deal of progress in understanding the biology of these receptors and their functions in the immune system in the past decade. Here, we describe the contribution of the various ACKRs to innate and adaptive immune responses, focussing specifically on recent progress. This includes recent findings that have defined the role for ACKRs in sculpting extracellular chemokine gradients, findings that broaden the spectrum of chemokine ligands recognized by these receptors, candidate new additions to ACKR family, and our increasing understanding of the role of these receptors in shaping the migration of innate and adaptive immune cells.
Collapse
Affiliation(s)
- Iain Comerford
- The Chemokine Biology Laboratory, School of Molecular & Biomedical Science, The University of Adelaide, Adelaide, South Australia, Australia.
| | - Shaun R McColl
- The Chemokine Biology Laboratory, School of Molecular & Biomedical Science, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
2
|
Saha S, Khanppnavar B, Maharana J, Kim H, Carino CMC, Daly C, Houston S, Sharma S, Zaidi N, Dalal A, Mishra S, Ganguly M, Tiwari D, Kumari P, Jhingan GD, Yadav PN, Plouffe B, Inoue A, Chung KY, Banerjee R, Korkhov VM, Shukla AK. Molecular mechanism of distinct chemokine engagement and functional divergence of the human Duffy antigen receptor. Cell 2024; 187:4751-4769.e25. [PMID: 39089252 DOI: 10.1016/j.cell.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 04/13/2024] [Accepted: 07/03/2024] [Indexed: 08/03/2024]
Abstract
The Duffy antigen receptor is a seven-transmembrane (7TM) protein expressed primarily at the surface of red blood cells and displays strikingly promiscuous binding to multiple inflammatory and homeostatic chemokines. It serves as the basis of the Duffy blood group system in humans and also acts as the primary attachment site for malarial parasite Plasmodium vivax and pore-forming toxins secreted by Staphylococcus aureus. Here, we comprehensively profile transducer coupling of this receptor, discover potential non-canonical signaling pathways, and determine the cryoelectron microscopy (cryo-EM) structure in complex with the chemokine CCL7. The structure reveals a distinct binding mode of chemokines, as reflected by relatively superficial binding and a partially formed orthosteric binding pocket. We also observe a dramatic shortening of TM5 and 6 on the intracellular side, which precludes the formation of the docking site for canonical signal transducers, thereby providing a possible explanation for the distinct pharmacological and functional phenotype of this receptor.
Collapse
Affiliation(s)
- Shirsha Saha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Basavraj Khanppnavar
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland; Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
| | - Jagannath Maharana
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Heeryung Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Carlo Marion C Carino
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Carole Daly
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Shane Houston
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Saloni Sharma
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Nashrah Zaidi
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Annu Dalal
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Sudha Mishra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Manisankar Ganguly
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Divyanshu Tiwari
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Poonam Kumari
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, India
| | | | - Prem N Yadav
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Bianca Plouffe
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Ka Young Chung
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ramanuj Banerjee
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| | - Volodymyr M Korkhov
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland; Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland.
| | - Arun K Shukla
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| |
Collapse
|
3
|
Samus M, Rot A. Atypical chemokine receptors in cancer. Cytokine 2024; 176:156504. [PMID: 38266462 DOI: 10.1016/j.cyto.2024.156504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/28/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Atypical chemokine receptors (ACKRs) are a group of seven-transmembrane spanning serpentine receptors that are structurally homologous to classical G-protein-coupled receptors and bind cognate chemokines with high affinities but do not signal via G-proteins or mediate cell migration. However, ACKRs efficiently modify the availability and function of chemokines in defined microanatomical environments, can signal via intracellular effectors other than G-proteins, and play complex roles in physiology and disease, including in cancer. In this review, we summarize the findings on the diverse contributions of individual ACKRs to cancer development, progression, and tumor-host interactions. We discuss how changes in ACKR expression within tumor affect cancer growth, tumor vascularization, leukocyte infiltration, and metastasis formation, ultimately resulting in differential disease outcomes. Across many studies, ACKR3 expression was shown to support tumor growth and dissemination, whereas ACKR1, ACKR2, and ACKR4 in tumors were more likely to contribute to tumor suppression. With few notable exceptions, the insights on molecular and cellular mechanisms of ACKRs activities in cancer remain sparse, and the intricacies of their involvement are not fully appreciated. This is particularly true for ACKR1, ACKR2 and ACKR4. A better understanding of how ACKR expression and functions impact cancer should pave the way for their future targeting by new and effective therapies.
Collapse
Affiliation(s)
- Maryna Samus
- Centre for Microvascular Research, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Antal Rot
- Centre for Microvascular Research, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK; Institute for Cardiovascular Prevention, Ludwig-Maximilians University, Munich 80336, Germany.
| |
Collapse
|
4
|
Karan D, Wick J, Dubey S, Kumar-Sinha C, Siddiqui J, Kunju LP, Iczkowski KA, Chinnaiyan AM. Racial differences in serum chemokines in prostate cancer patients. Cancer 2023; 129:3783-3789. [PMID: 37698493 DOI: 10.1002/cncr.35012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 09/13/2023]
Abstract
BACKGROUND This study aimed to understand the differential levels of inflammatory chemokines in association with higher prostate cancer incidence and mortality in African American (AA) men than in Caucasians (CA). METHODS The authors used a chemokine assay to simultaneously measure 40 chemokines and cytokines levels in the serum of preoperative prostate cancer patients and healthy controls of AA and CA races. Selected chemokines (CXCL2, CXCL5, and CCL23) serum level was validated in 211 serum samples from prostate cancer patients and healthy controls. Differential expression of CXCL5 and CCL23 was analyzed using immunohistochemistry in a representative cohort of prostate tumor tissues of AA and CA races. RESULTS Race-specific comparisons from 211 serum samples showed significantly higher levels of CXCL2 (control: 3104.0 pg/mL vs. cancer: 2451.0 pg/mL) and CXCL5 (control: 5189.0 pg/mL vs. cancer: 5459.0 pg/mL) in AA men than in CAs (CXCL2; control: 1155.0 pg/mL vs. cancer: 889.3 pg/mL, and CXCL5; control: 1183.0 pg/mL vs. cancer: 977.5 pg/mL). CCL23 differed significantly within and between the races with a lower level in AA cancer cases (454.5 vs. 966.6 pg/mL) than healthy controls (740.5 vs. 1263.0 pg/mL). Patient age, prostate-specific antigen, or Gleason scores were not significantly associated with these chemokines. Immunostaining for CXCL5 and CCL23 in a representative cohort of archival prostate tissues displayed significantly higher CXCL5 in prostate tumors than in adjacent benign tissues, whereas CCL23 was nondetectable in most of the analyzed tumor tissues. CONCLUSION Lower levels of CCL23 in AA prostate cancer patient sera and tumor tissues and high CXCL2 and CXCL5 may contribute to aggressive prostate cancer, as often seen in AA men. The disproportionate levels of serum chemokines associated with race warrant further exploration to improve equitability in precision oncology to benefit prostate cancer patients.
Collapse
Affiliation(s)
- Dev Karan
- Department of Pathology, MCW Cancer Center and Prostate Cancer Center of Excellence, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jo Wick
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Seema Dubey
- Department of Pathology, MCW Cancer Center and Prostate Cancer Center of Excellence, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Chandan Kumar-Sinha
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Javed Siddiqui
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lakshmi P Kunju
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kenneth A Iczkowski
- Department of Pathology, MCW Cancer Center and Prostate Cancer Center of Excellence, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
5
|
Leberzammer J, von Hundelshausen P. Chemokines, molecular drivers of thromboinflammation and immunothrombosis. Front Immunol 2023; 14:1276353. [PMID: 37954596 PMCID: PMC10637585 DOI: 10.3389/fimmu.2023.1276353] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/12/2023] [Indexed: 11/14/2023] Open
Abstract
Blood clotting is a finely regulated process that is essential for hemostasis. However, when dysregulated or spontaneous, it promotes thrombotic disorders. The fact that these are triggered, accompanied and amplified by inflammation is reflected in the term thromboinflammation that includes chemokines. The role of chemokines in thrombosis is therefore illuminated from a cellular perspective, where endothelial cells, platelets, red blood cells, and leukocytes may be both the source and target of chemokines. Chemokine-dependent prothrombotic processes may thereby occur independently of chemokine receptors or be mediated by chemokine receptors, although the binding and activation of classical G protein-coupled receptors and their signaling pathways differ from those of atypical chemokine receptors, which do not function via cell activation and recruitment. Regardless of binding to their receptors, chemokines can induce thrombosis by forming platelet-activating immune complexes with heparin or other polyanions that are pathognomonic for HIT and VITT. In addition, chemokines can bind to NETs and alter their structure. They also change the electrical charge of the cell surface of platelets and interact with coagulation factors, thereby modulating the balance of fibrinolysis and coagulation. Moreover, CXCL12 activates CXCR4 on platelets independently of classical migratory chemokine activity and causes aggregation and thrombosis via the PI3Kβ and Btk signaling pathways. In contrast, typical chemokine-chemokine receptor interactions are involved in the processes that contribute to the adhesiveness of the endothelium in the initial phase of venous thrombosis, where neutrophils and monocytes subsequently accumulate in massive numbers. Later, the reorganization and resolution of a thrombus require coordinated cell migration and invasion of the thrombus, and, as such, indeed, chemokines recruit leukocytes to existing thrombi. Therefore, chemokines contribute in many independent ways to thrombosis.
Collapse
Affiliation(s)
- Julian Leberzammer
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- Department of Cardiology and Angiology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Philipp von Hundelshausen
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Cardiovascular Prevention, Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten (IPEK), Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
6
|
Crawford KS, Volkman BF. Prospects for targeting ACKR1 in cancer and other diseases. Front Immunol 2023; 14:1111960. [PMID: 37006247 PMCID: PMC10050359 DOI: 10.3389/fimmu.2023.1111960] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
The chemokine network is comprised of a family of signal proteins that encode messages for cells displaying chemokine G-protein coupled receptors (GPCRs). The diversity of effects on cellular functions, particularly directed migration of different cell types to sites of inflammation, is enabled by different combinations of chemokines activating signal transduction cascades on cells displaying a combination of receptors. These signals can contribute to autoimmune disease or be hijacked in cancer to stimulate cancer progression and metastatic migration. Thus far, three chemokine receptor-targeting drugs have been approved for clinical use: Maraviroc for HIV, Plerixafor for hematopoietic stem cell mobilization, and Mogalizumab for cutaneous T-cell lymphoma. Numerous compounds have been developed to inhibit specific chemokine GPCRs, but the complexity of the chemokine network has precluded more widespread clinical implementation, particularly as anti-neoplastic and anti-metastatic agents. Drugs that block a single signaling axis may be rendered ineffective or cause adverse reactions because each chemokine and receptor often have multiple context-specific functions. The chemokine network is tightly regulated at multiple levels, including by atypical chemokine receptors (ACKRs) that control chemokine gradients independently of G-proteins. ACKRs have numerous functions linked to chemokine immobilization, movement through and within cells, and recruitment of alternate effectors like β-arrestins. Atypical chemokine receptor 1 (ACKR1), previously known as the Duffy antigen receptor for chemokines (DARC), is a key regulator that binds chemokines involved in inflammatory responses and cancer proliferation, angiogenesis, and metastasis. Understanding more about ACKR1 in different diseases and populations may contribute to the development of therapeutic strategies targeting the chemokine network.
Collapse
Affiliation(s)
- Kyler S. Crawford
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | | |
Collapse
|
7
|
Dillemans L, De Somer L, Neerinckx B, Proost P. A review of the pleiotropic actions of the IFN-inducible CXC chemokine receptor 3 ligands in the synovial microenvironment. Cell Mol Life Sci 2023; 80:78. [PMID: 36862204 PMCID: PMC11071919 DOI: 10.1007/s00018-023-04715-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/09/2023] [Accepted: 02/01/2023] [Indexed: 03/03/2023]
Abstract
Chemokines are pivotal players in instigation and perpetuation of synovitis through leukocytes egress from the blood circulation into the inflamed articulation. Multitudinous literature addressing the involvement of the dual-function interferon (IFN)-inducible chemokines CXCL9, CXCL10 and CXCL11 in diseases characterized by chronic inflammatory arthritis emphasizes the need for detangling their etiopathological relevance. Through interaction with their mutual receptor CXC chemokine receptor 3 (CXCR3), the chemokines CXCL9, CXCL10 and CXCL11 exert their hallmark function of coordinating directional trafficking of CD4+ TH1 cells, CD8+ T cells, NK cells and NKT cells towards inflammatory niches. Among other (patho)physiological processes including infection, cancer, and angiostasis, IFN-inducible CXCR3 ligands have been implicated in autoinflammatory and autoimmune diseases. This review presents a comprehensive overview of the abundant presence of IFN-induced CXCR3 ligands in bodily fluids of patients with inflammatory arthritis, the outcomes of their selective depletion in rodent models, and the attempts at developing candidate drugs targeting the CXCR3 chemokine system. We further propose that the involvement of the CXCR3 binding chemokines in synovitis and joint remodeling encompasses more than solely the directional ingress of CXCR3-expressing leukocytes. The pleotropic actions of the IFN-inducible CXCR3 ligands in the synovial niche reiteratively illustrate the extensive complexity of the CXCR3 chemokine network, which is based on the intercommunion of IFN-inducible CXCR3 ligands with distinct CXCR3 isoforms, enzymes, cytokines, and infiltrated and resident cells present in the inflamed joints.
Collapse
Affiliation(s)
- Luna Dillemans
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Lien De Somer
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Barbara Neerinckx
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
8
|
Parween F, Singh SP, Zhang HH, Kathuria N, Otaizo-Carrasquero FA, Shamsaddini A, Gardina PJ, Ganesan S, Kabat J, Lorenzi HA, Myers TG, Farber JM. Chemokine positioning determines mutually exclusive roles for their receptors in extravasation of pathogenic human T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.25.525561. [PMID: 36789428 PMCID: PMC9928044 DOI: 10.1101/2023.01.25.525561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Pro-inflammatory T cells co-express multiple chemokine receptors, but the distinct functions of individual receptors on these cells are largely unknown. Human Th17 cells uniformly express the chemokine receptor CCR6, and we discovered that the subgroup of CD4+CCR6+ cells that co-express CCR2 possess a pathogenic Th17 signature, can produce inflammatory cytokines independent of TCR activation, and are unusually efficient at transendothelial migration (TEM). The ligand for CCR6, CCL20, was capable of binding to activated endothelial cells (ECs) and inducing firm arrest of CCR6+CCR2+ cells under conditions of flow - but CCR6 could not mediate TEM. By contrast, CCL2 and other ligands for CCR2, despite being secreted from both luminal and basal sides of ECs, failed to bind to the EC surfaces - and CCR2 could not mediate arrest. Nonetheless, CCR2 was required for TEM. To understand if CCR2's inability to mediate arrest was due solely to an absence of EC-bound ligands, we generated a CCL2-CXCL9 chimeric chemokine that could bind to the EC surface. Although display of CCL2 on the ECs did indeed lead to CCR2-mediated arrest of CCR6+CCR2+ cells, activating CCR2 with surface-bound CCL2 blocked TEM. We conclude that mediating arrest and TEM are mutually exclusive activities of chemokine receptors and/or their ligands that depend, respectively, on chemokines that bind to the EC luminal surfaces versus non-binding chemokines that form transendothelial gradients under conditions of flow. Our findings provide fundamental insights into mechanisms of lymphocyte extravasation and may lead to novel strategies to block or enhance their migration into tissue.
Collapse
Affiliation(s)
- Farhat Parween
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Satya P. Singh
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Hongwei H Zhang
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Nausheen Kathuria
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Francisco A. Otaizo-Carrasquero
- Research Technologies Branch, Genomic Technologies, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Amirhossein Shamsaddini
- Research Technologies Branch, Genomic Technologies, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Paul J. Gardina
- Research Technologies Branch, Genomic Technologies, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Sundar Ganesan
- Research Technologies Branch, Biological Imaging, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Juraj Kabat
- Research Technologies Branch, Biological Imaging, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Hernan A. Lorenzi
- Bioinformatics and Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Timothy G. Myers
- Research Technologies Branch, Genomic Technologies, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Joshua M. Farber
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| |
Collapse
|
9
|
Elmansi AM, Eisa NH, Periyasamy-Thandavan S, Kondrikova G, Kondrikov D, Calkins MM, Aguilar-Pérez A, Chen J, Johnson M, Shi XM, Reitman C, McGee-Lawrence ME, Crawford KS, Dwinell MB, Volkman BF, Blumer JB, Luttrell LM, McCorvy JD, Hill WD. DPP4-Truncated CXCL12 Alters CXCR4/ACKR3 Signaling, Osteogenic Cell Differentiation, Migration, and Senescence. ACS Pharmacol Transl Sci 2023; 6:22-39. [PMID: 36659961 PMCID: PMC9844133 DOI: 10.1021/acsptsci.2c00040] [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: 03/07/2022] [Indexed: 12/15/2022]
Abstract
Bone marrow skeletal stem cells (SSCs) secrete many cytokines including stromal derived factor-1 or CXCL12, which influences cell proliferation, migration, and differentiation. All CXCL12 splice variants are rapidly truncated on their N-terminus by dipeptidyl peptidase 4 (DPP4). This includes the common variant CXCL12 alpha (1-68) releasing a much less studied metabolite CXCL12(3-68). Here, we found that CXCL12(3-68) significantly inhibited SSC osteogenic differentiation and RAW-264.7 cell osteoclastogenic differentiation and induced a senescent phenotype in SSCs. Importantly, pre-incubation of SSCs with CXCL12(3-68) significantly diminished their ability to migrate toward CXCL12(1-68) in transwell migration assays. Using a high-throughput G-protein-coupled receptor (GPCR) screen (GPCRome) and bioluminescent resonance energy transfer molecular interaction assays, we revealed that CXCL12(3-68) acts via the atypical cytokine receptor 3-mediated β-arrestin recruitment and as a competitive antagonist to CXCR4-mediated signaling. Finally, a reverse phase protein array assay revealed that DPP4-cleaved CXCL12 possesses a different downstream signaling profile from that of intact CXCL12 or controls. The data presented herein provides insights into regulation of CXCL12 signaling. Importantly, it demonstrates that DPP4 proteolysis of CXCL12 generates a metabolite with significantly different and previously overlooked bioactivity that helps explain discrepancies in the literature. This also contributes to an understanding of the molecular mechanisms of osteoporosis and bone fracture repair and could potentially significantly affect the interpretation of experimental outcomes with clinical consequences in other fields where CXCL12 is vital, including cancer biology, immunology, cardiovascular biology, neurobiology, and associated pathologies.
Collapse
Affiliation(s)
- Ahmed M. Elmansi
- Department of Pathology and Laboratory Medicine,
Medical University of South Carolina, Charleston, South
Carolina 29403, United States
- Johnson Veterans Affairs Medical
Center, Charleston, South Carolina 29403, United
States
- Department of Pathology, University of
Michigan School of Medicine, Ann Arbor, Michigan 48109, United
States
| | - Nada H. Eisa
- Department of Pathology and Laboratory Medicine,
Medical University of South Carolina, Charleston, South
Carolina 29403, United States
- Johnson Veterans Affairs Medical
Center, Charleston, South Carolina 29403, United
States
- Department of Biochemistry, Faculty of Pharmacy,
Mansoura University, Mansoura 35516,
Egypt
| | | | - Galina Kondrikova
- Department of Pathology and Laboratory Medicine,
Medical University of South Carolina, Charleston, South
Carolina 29403, United States
- Johnson Veterans Affairs Medical
Center, Charleston, South Carolina 29403, United
States
| | - Dmitry Kondrikov
- Department of Pathology and Laboratory Medicine,
Medical University of South Carolina, Charleston, South
Carolina 29403, United States
- Johnson Veterans Affairs Medical
Center, Charleston, South Carolina 29403, United
States
| | - Maggie M. Calkins
- Department of Cell Biology, Neurobiology and Anatomy,
Medical College of Wisconsin, 8701 W. Watertown Plank Road,
Milwaukee, Wisconsin 53226, United States
| | - Alexandra Aguilar-Pérez
- Department of Anatomy and Cell Biology,
Indiana University School of Medicine in Indianapolis,
Indianapolis, Indiana 46202, United States
- Department of Cellular and Molecular Biology, School
of Medicine, Universidad Central Del Caribe, Bayamon, Puerto
Rico 00956, United States
- Cellular Biology and Anatomy, Medical College of
Georgia, Augusta University, Augusta, Georgia 30912,
United States
| | - Jie Chen
- Division of Biostatistics and Data Science,
Department of Population Health Science, Medical College of Georgia, Augusta
University, Augusta, Georgia 30912, United States
| | - Maribeth Johnson
- Division of Biostatistics and Data Science,
Department of Population Health Science, Medical College of Georgia, Augusta
University, Augusta, Georgia 30912, United States
| | - Xing-ming Shi
- Department of Orthopaedic Surgery, Medical
College of Georgia, Augusta University, Augusta, Georgia 30912,
United States
- Department of Neuroscience and Regenerative
Medicine, Medical College of Georgia, Augusta University,
Augusta, Georgia 30912, United States
| | - Charles Reitman
- Orthopaedics and Physical Medicine Department,
Medical University of South Carolina, Charleston, South
Carolina 29403, United States
| | - Meghan E. McGee-Lawrence
- Cellular Biology and Anatomy, Medical College of
Georgia, Augusta University, Augusta, Georgia 30912,
United States
- Department of Orthopaedic Surgery, Medical
College of Georgia, Augusta University, Augusta, Georgia 30912,
United States
- Center for Healthy Aging, Medical College of
Georgia, Augusta University, Augusta, Georgia 30912,
United States
| | - Kyler S. Crawford
- Department of Biochemistry,
Medical College of Wisconsin, Milwaukee, Wisconsin 53226,
United States
| | - Michael B. Dwinell
- Department of Microbiology and Immunology,
Medical College of Wisconsin, Milwaukee, Wisconsin 53226,
United States
| | - Brian F. Volkman
- Department of Biochemistry,
Medical College of Wisconsin, Milwaukee, Wisconsin 53226,
United States
| | - Joe B. Blumer
- Department of Cell and Molecular Pharmacology and
Experimental Therapeutics, Medical University of South
Carolina, Charleston, South Carolina 29425, United
States
| | - Louis M. Luttrell
- Division of Endocrinology, Diabetes and
Medical Genetics, Medical University of South Carolina,
Charleston, South Carolina 29403, United States
| | - John D. McCorvy
- Department of Cell Biology, Neurobiology and Anatomy,
Medical College of Wisconsin, 8701 W. Watertown Plank Road,
Milwaukee, Wisconsin 53226, United States
| | - William D. Hill
- Department of Pathology and Laboratory Medicine,
Medical University of South Carolina, Charleston, South
Carolina 29403, United States
- Johnson Veterans Affairs Medical
Center, Charleston, South Carolina 29403, United
States
- Cellular Biology and Anatomy, Medical College of
Georgia, Augusta University, Augusta, Georgia 30912,
United States
- Center for Healthy Aging, Medical College of
Georgia, Augusta University, Augusta, Georgia 30912,
United States
- Charlie Norwood Veterans Affairs
Medical Center, Augusta, Georgia 30904, United
States
| |
Collapse
|
10
|
Gil E, Wall E, Noursadeghi M, Brown JS. Streptococcus pneumoniae meningitis and the CNS barriers. Front Cell Infect Microbiol 2023; 12:1106596. [PMID: 36683708 PMCID: PMC9845635 DOI: 10.3389/fcimb.2022.1106596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/08/2022] [Indexed: 01/05/2023] Open
Abstract
Streptococcus pneumoniae (SPN) is a globally significant cause of meningitis, the pathophysiology of which involves damage to the brain by both bacterial virulence factors and the host inflammatory response. In most cases of SPN meningitis bacteria translocate from the blood into the central nervous system (CNS). The principal site of SPN translocation into the CNS is not known, with possible portals of entry proposed to be the cerebral or meningeal blood vessels or the choroid plexus. All require SPN to bind to and translocate across the vascular endothelial barrier, and subsequently the basement membrane and perivascular structures, including an additional epithelial barrier in the case of the blood-CSF barrier. The presence of SPN in the CNS is highly inflammatory resulting in marked neutrophilic infiltration. The secretion of toxic inflammatory mediators by activated neutrophils within the CNS damages pathogen and host alike, including the non-replicative neurons which drives morbidity and mortality. As with the translocation of SPN, the recruitment of neutrophils into the CNS in SPN meningitis necessitates the translocation of neutrophils from the circulation across the vascular barrier, a process that is tightly regulated under basal conditions - a feature of the 'immune specialization' of the CNS. The brain barriers are therefore central to SPN meningitis, both through a failure to exclude bacteria and maintain CNS sterility, and subsequently through the active recruitment and/or failure to exclude circulating leukocytes. The interactions of SPN with these barriers, barrier inflammatory responses, along with their therapeutic implications, are explored in this review.
Collapse
Affiliation(s)
- Eliza Gil
- Division of Infection and Immunity, University College London, London, United Kingdom,*Correspondence: Eliza Gil,
| | - Emma Wall
- Francis Crick Institute, London, United Kingdom,UCLH Biomedical Research Centre, London, United Kingdom
| | - Mahdad Noursadeghi
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Jeremy S. Brown
- Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
11
|
Szpakowska M, D’Uonnolo G, Luís R, Alonso Bartolomé A, Thelen M, Legler DF, Chevigné A. New pairings and deorphanization among the atypical chemokine receptor family - physiological and clinical relevance. Front Immunol 2023; 14:1133394. [PMID: 37153591 PMCID: PMC10157204 DOI: 10.3389/fimmu.2023.1133394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/30/2023] [Indexed: 05/09/2023] Open
Abstract
Atypical chemokine receptors (ACKRs) form a small subfamily of receptors (ACKR1-4) unable to trigger G protein-dependent signaling in response to their ligands. They do, however, play a crucial regulatory role in chemokine biology by capturing, scavenging or transporting chemokines, thereby regulating their availability and signaling through classical chemokine receptors. ACKRs add thus another layer of complexity to the intricate chemokine-receptor interaction network. Recently, targeted approaches and screening programs aiming at reassessing chemokine activity towards ACKRs identified several new pairings such as the dimeric CXCL12 with ACKR1, CXCL2, CXCL10 and CCL26 with ACKR2, the viral broad-spectrum chemokine vCCL2/vMIP-II, a range of opioid peptides and PAMP-12 with ACKR3 as well as CCL20 and CCL22 with ACKR4. Moreover, GPR182 (ACKR5) has been lately proposed as a new promiscuous atypical chemokine receptor with scavenging activity notably towards CXCL9, CXCL10, CXCL12 and CXCL13. Altogether, these findings reveal new degrees of complexity of the chemokine network and expand the panel of ACKR ligands and regulatory functions. In this minireview, we present and discuss these new pairings, their physiological and clinical relevance as well as the opportunities they open for targeting ACKRs in innovative therapeutic strategies.
Collapse
Affiliation(s)
- Martyna Szpakowska
- Immuno-Pharmacology and Interactomics,Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Giulia D’Uonnolo
- Immuno-Pharmacology and Interactomics,Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Rafael Luís
- Immuno-Pharmacology and Interactomics,Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Tumor Immunotherapy and Microenvironment, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Ana Alonso Bartolomé
- Immuno-Pharmacology and Interactomics,Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Marcus Thelen
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Daniel F. Legler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland
| | - Andy Chevigné
- Immuno-Pharmacology and Interactomics,Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- *Correspondence: Andy Chevigné,
| |
Collapse
|
12
|
Gil E, Venturini C, Stirling D, Turner C, Tezera LB, Ercoli G, Baker T, Best K, Brown JS, Noursadeghi M. Pericyte derived chemokines amplify neutrophil recruitment across the cerebrovascular endothelial barrier. Front Immunol 2022; 13:935798. [PMID: 35967327 PMCID: PMC9371542 DOI: 10.3389/fimmu.2022.935798] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Excessive neutrophil extravasation can drive immunopathology, exemplified in pyogenic meningitis caused by Streptococcus pneumoniae infection. Insufficient knowledge of the mechanisms that amplify neutrophil extravasation has limited innovation in therapeutic targeting of neutrophil mediated pathology. Attention has focussed on neutrophil interactions with endothelia, but data from mouse models also point to a role for the underlying pericyte layer, as well as perivascular macrophages, the only other cell type found within the perivascular space in the cerebral microvasculature. We tested the hypothesis that human brain vascular pericytes (HBVP) contribute to neutrophil extravasation in a transwell model of the cerebral post-capillary venule. We show that pericytes augment endothelial barrier formation. In response to inflammatory cues, they significantly enhance neutrophil transmigration across the endothelial barrier, without increasing the permeability to small molecules. In our model, neither pericytes nor endothelia responded directly to bacterial stimulation. Instead, we show that paracrine signalling by multiple cytokines from monocyte derived macrophages drives transcriptional upregulation of multiple neutrophil chemokines by pericytes. Pericyte mediated amplification of neutrophil transmigration was independent of transcriptional responses by endothelia, but could be mediated by direct chemokine translocation across the endothelial barrier. Our data support a model in which microbial sensing by perivascular macrophages generates an inflammatory cascade where pericytes serve to amplify production of neutrophil chemokines that are translocated across the endothelial barrier to act directly on circulating neutrophils. In view of the striking redundancy in inflammatory cytokines that stimulate pericytes and in the neutrophil chemokines they produce, we propose that the mechanism of chemokine translocation may offer the most effective therapeutic target to reduce neutrophil mediated pathology in pyogenic meningitis.
Collapse
Affiliation(s)
- Eliza Gil
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Cristina Venturini
- Infection, Immunity and Inflammation Department, Institute for Child Health, University College London, London, United Kingdom
| | - David Stirling
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Carolin Turner
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Liku B. Tezera
- Division of Infection and Immunity, University College London, London, United Kingdom
- NIHR Biomedical Research Center, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Giuseppe Ercoli
- Centre for Inflammation and Tissue Repair, Division of Medicine, University College London, London, United Kingdom
| | - Tina Baker
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Katharine Best
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Jeremy S. Brown
- Centre for Inflammation and Tissue Repair, Division of Medicine, University College London, London, United Kingdom
| | - Mahdad Noursadeghi
- Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
13
|
Serum proteomics links suppression of tumor immunity to ancestry and lethal prostate cancer. Nat Commun 2022; 13:1759. [PMID: 35365620 PMCID: PMC8975871 DOI: 10.1038/s41467-022-29235-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 03/07/2022] [Indexed: 12/31/2022] Open
Abstract
There is evidence that tumor immunobiology and immunotherapy response may differ between African American and European American prostate cancer patients. Here, we determine if men of African descent harbor a unique systemic immune-oncological signature and measure 82 circulating proteins in almost 3000 Ghanaian, African American, and European American men. Protein signatures for suppression of tumor immunity and chemotaxis are elevated in men of West African ancestry. Importantly, the suppression of tumor immunity protein signature associates with metastatic and lethal prostate cancer, pointing to clinical importance. Moreover, two markers, pleiotrophin and TNFRSF9, predict poor disease survival specifically among African American men. These findings indicate that immune-oncology marker profiles differ between men of African and European descent. These differences may contribute to the disproportionate burden of lethal prostate cancer in men of African ancestry. The elevated peripheral suppression of tumor immunity may have important implication for guidance of cancer therapy which could particularly benefit African American patients. Ancestry-related differences in immunobiology may explain the health disparities observed in prostate cancer patients, with men of African origin bearing the highest prostate cancer burden. By measuring immune-related proteins in serum samples, here the authors report that systemic cytokines linked to suppression of tumor immunity are upregulated in men of African ancestry and associated with reduced survival.
Collapse
|
14
|
Marchetti L, Francisco D, Soldati S, Haghayegh Jahromi N, Barcos S, Gruber I, Pareja JR, Thiriot A, von Andrian U, Deutsch U, Lyck R, Bruggmann R, Engelhardt B. ACKR1 favors transcellular over paracellular T-cell diapedesis across the blood-brain barrier in neuroinflammation in vitro. Eur J Immunol 2022; 52:161-177. [PMID: 34524684 PMCID: PMC9293480 DOI: 10.1002/eji.202149238] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 08/11/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022]
Abstract
The migration of CD4+ effector/memory T cells across the blood-brain barrier (BBB) is a critical step in MS or its animal model, EAE. T-cell diapedesis across the BBB can occur paracellular, via the complex BBB tight junctions or transcellular via a pore through the brain endothelial cell body. Making use of primary mouse brain microvascular endothelial cells (pMBMECs) as in vitro model of the BBB, we here directly compared the transcriptome profile of pMBMECs favoring transcellular or paracellular T-cell diapedesis by RNA sequencing (RNA-seq). We identified the atypical chemokine receptor 1 (Ackr1) as one of the main candidate genes upregulated in pMBMECs favoring transcellular T-cell diapedesis. We confirmed upregulation of ACKR1 protein in pMBMECs promoting transcellular T-cell diapedesis and in venular endothelial cells in the CNS during EAE. Lack of endothelial ACKR1 reduced transcellular T-cell diapedesis across pMBMECs under physiological flow in vitro. Combining our previous observation that endothelial ACKR1 contributes to EAE pathogenesis by shuttling chemokines across the BBB, the present data support that ACKR1 mediated chemokine shuttling enhances transcellular T-cell diapedesis across the BBB during autoimmune neuroinflammation.
Collapse
Affiliation(s)
- Luca Marchetti
- Theodor Kocher InstituteUniversity of BernBernSwitzerland
| | - David Francisco
- Interfaculty Bioinformatics Unit and Swiss Institute of BioinformaticsUniversity of BernBernSwitzerland
| | - Sasha Soldati
- Theodor Kocher InstituteUniversity of BernBernSwitzerland
| | | | - Sara Barcos
- Theodor Kocher InstituteUniversity of BernBernSwitzerland
| | - Isabelle Gruber
- Theodor Kocher InstituteUniversity of BernBernSwitzerland
- present address: Department of Oncology, Lausanne University HospitalUniversity of LausanneLausanneSwitzerland
| | | | - Aude Thiriot
- Department of Immunology and Center for Immune ImagingHarvard Medical SchoolBostonMassachusettsUSA
- The Ragon Institute of MGH, MIT and HarvardCambridgeMassachusettsUSA
| | - Ulrich von Andrian
- Department of Immunology and Center for Immune ImagingHarvard Medical SchoolBostonMassachusettsUSA
- The Ragon Institute of MGH, MIT and HarvardCambridgeMassachusettsUSA
| | - Urban Deutsch
- Theodor Kocher InstituteUniversity of BernBernSwitzerland
| | - Ruth Lyck
- Theodor Kocher InstituteUniversity of BernBernSwitzerland
| | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of BioinformaticsUniversity of BernBernSwitzerland
| | | |
Collapse
|
15
|
Gutjahr JC, Crawford KS, Jensen DR, Naik P, Peterson FC, Samson GPB, Legler DF, Duchene J, Veldkamp CT, Rot A, Volkman BF. The dimeric form of CXCL12 binds to atypical chemokine receptor 1. Sci Signal 2021; 14:14/696/eabc9012. [PMID: 34404752 DOI: 10.1126/scisignal.abc9012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The pleiotropic chemokine CXCL12 is involved in diverse physiological and pathophysiological processes, including embryogenesis, hematopoiesis, leukocyte migration, and tumor metastasis. It is known to engage the classical receptor CXCR4 and the atypical receptor ACKR3. Differential receptor engagement can transduce distinct cellular signals and effects as well as alter the amount of free, extracellular chemokine. CXCR4 binds both monomeric and the more commonly found dimeric forms of CXCL12, whereas ACKR3 binds monomeric forms. Here, we found that CXCL12 also bound to the atypical receptor ACKR1 (previously known as Duffy antigen/receptor for chemokines or DARC). In vitro nuclear magnetic resonance spectroscopy and isothermal titration calorimetry revealed that dimeric CXCL12 bound to the extracellular N terminus of ACKR1 with low nanomolar affinity, whereas the binding affinity of monomeric CXCL12 was orders of magnitude lower. In transfected MDCK cells and primary human Duffy-positive erythrocytes, a dimeric, but not a monomeric, construct of CXCL12 efficiently bound to and internalized with ACKR1. This interaction between CXCL12 and ACKR1 provides another layer of regulation of the multiple biological functions of CXCL12. The findings also raise the possibility that ACKR1 can bind other dimeric chemokines, thus potentially further expanding the role of ACKR1 in chemokine retention and presentation.
Collapse
Affiliation(s)
- Julia C Gutjahr
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Kyler S Crawford
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Davin R Jensen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Prachi Naik
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Francis C Peterson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Guerric P B Samson
- Biotechnology Institute Thurgau (BITg), University of Konstanz, 8280 Kreuzlingen, Switzerland
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg), University of Konstanz, 8280 Kreuzlingen, Switzerland.,Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Johan Duchene
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University, 80336 Munich, Germany
| | | | - Antal Rot
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK. .,Institute for Cardiovascular Prevention, Ludwig-Maximilians University, 80336 Munich, Germany.,Centre for Inflammation and Therapeutic Innovation, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
16
|
Pan X, Chiwanda Kaminga A, Liu A, Wen SW, Chen J, Luo J. Chemokines in Non-alcoholic Fatty Liver Disease: A Systematic Review and Network Meta-Analysis. Front Immunol 2020; 11:1802. [PMID: 33042108 PMCID: PMC7530185 DOI: 10.3389/fimmu.2020.01802] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/06/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Previous results on the relationship between non-alcoholic fatty liver disease (NAFLD) and chemokine concentrations were inconsistent. The purpose of this network meta-analysis was to evaluate the link between chemokine system and NAFLD. Methods: Relevant data, published not later than June 31, 2019, were searched in the databases of PubMed, Embase, Cochrane Library, and Web of Science. A network meta-analysis was used to rank the chemokines by surface under the cumulative ranking (SUCRA) probabilities. In addition, standardized mean differences (SMDs) with 95% confidence intervals (CIs) were calculated as group differences in the chemokine concentrations. Results: The search in the databases identified 46 relevant studies that investigated the relationship between 15 different chemokines and NAFLD using 4,753 patients and 4,059 controls. Results from the network meta-analysis showed that the concentrations of CCL2 and CXCL8 in the non-alcoholic fatty liver (NAFL) group was significantly higher than that in the control group (SMDs of 1.51 and 1.95, respectively), and the concentrations of CCL3, CCL4, CCL20, CXCL8, and CXCL10 in the non-alcoholic steatohepatitis (NASH) group was significantly higher than that in the control group (SMDs of 0.90, 2.05, 2.16, 0.91, and 1.46, respectively). SUCRA probabilities showed that CXCL8 had the highest rank in NAFL for all chemokines and CCL20 had the highest rank in NASH for all chemokines. Conclusion: Elevated concentrations of CCL2, CCL4, CCL20, CXCL8, and CXCL10 may be associated with NAFL or NASH. In this regard, more population-based studies are needed to ascertain this hypothesis. Systematic Review Registration: PROSPERO: CRD42020139373.
Collapse
Affiliation(s)
- Xiongfeng Pan
- Xiangya School of Public Health, Central South University, Changsha, China
| | - Atipatsa Chiwanda Kaminga
- Xiangya School of Public Health, Central South University, Changsha, China.,Department of Mathematics and Statistics, Mzuzu University, Mzuzu, Malawi
| | - Aizhong Liu
- Xiangya School of Public Health, Central South University, Changsha, China
| | - Shi Wu Wen
- Department of Obstetrics and Gynaecology, University of Ottawa, Ottawa, ON, Canada.,Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Jihua Chen
- Xiangya School of Public Health, Central South University, Changsha, China.,Department of Food Science and Biotechnol, Kagoshima University, Kagoshima, Japan
| | - Jiayou Luo
- Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
17
|
Sjöberg E, Meyrath M, Chevigné A, Östman A, Augsten M, Szpakowska M. The diverse and complex roles of atypical chemokine receptors in cancer: From molecular biology to clinical relevance and therapy. Adv Cancer Res 2020; 145:99-138. [PMID: 32089166 DOI: 10.1016/bs.acr.2019.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chemokines regulate directed cell migration, proliferation and survival and are key components in cancer biology. They exert their functions by interacting with seven-transmembrane domain receptors that signal through G proteins (GPCRs). A subgroup of four chemokine receptors known as the atypical chemokine receptors (ACKRs) has emerged as essential regulators of the chemokine functions. ACKRs play diverse and complex roles in tumor biology from tumor initiation to metastasis, including cancer cell proliferation, adherence to endothelium, epithelial-mesenchymal transition (EMT), extravasation from blood vessels, tumor-associated angiogenesis or protection from immunological responses. This chapter gives an overview on the established and emerging roles that the atypical chemokine receptors ACKR1, ACKR2, ACKR3 and ACKR4 play in the different phases of cancer development and dissemination, their clinical relevance, as well as on the hurdles to overcome in ACKRs targeting as cancer therapy.
Collapse
Affiliation(s)
- Elin Sjöberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Max Meyrath
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Andy Chevigné
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Arne Östman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Martyna Szpakowska
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
18
|
Klei TRL, Aglialoro F, Mul FPJ, Tol S, Ligthart PC, Seignette IM, Geissler J, van den Akker E, van Bruggen R. Differential interaction between DARC and SDF-1 on erythrocytes and their precursors. Sci Rep 2019; 9:16245. [PMID: 31700087 PMCID: PMC6838059 DOI: 10.1038/s41598-019-52186-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/26/2019] [Indexed: 01/21/2023] Open
Abstract
The Duffy Antigen Receptor for Chemokines (DARC) is expressed on erythrocytes and on endothelium of postcapillary venules and splenic sinusoids. Absence of DARC on erythrocytes, but not on endothelium, is referred to as the Duffy negative phenotype and is associated with neutropenia. Here we provide evidence that stromal cell-derived factor 1 (SDF-1), the chemokine that restricts neutrophil precursors to the bone marrow, binds to erythrocyte progenitors in a DARC-dependent manner. Furthermore, we show that SDF-1 binding to DARC is dependent on the conformation of DARC, which gradually changes during erythroid development, resulting in the absence of SDF-1 binding to mature erythrocytes. However, SDF-1 binding to erythrocytes was found to be inducible by pre-treating erythrocytes with IL-8 or with antibodies recognizing specific epitopes on DARC. Taken together, these novel findings identify DARC on erythrocyte precursors as a receptor for SDF-1, which may be of interest in beginning to understand the development of neutropenia in situations where DARC expression is limited.
Collapse
Affiliation(s)
- T R L Klei
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, The Netherlands
| | - F Aglialoro
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, Amsterdam, The Netherlands
| | - F P J Mul
- Research facility, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, The Netherlands
| | - S Tol
- Research facility, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, The Netherlands
| | - P C Ligthart
- Erythrocyte Serology, Sanquin, Amsterdam, The Netherlands
| | - I M Seignette
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, The Netherlands
| | - J Geissler
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, The Netherlands
| | - E van den Akker
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, Amsterdam, The Netherlands
| | - R van Bruggen
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, The Netherlands.
| |
Collapse
|
19
|
Systems Pharmacology and Microbiome Dissection of Shen Ling Bai Zhu San Reveal Multiscale Treatment Strategy for IBD. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8194804. [PMID: 31341536 PMCID: PMC6612409 DOI: 10.1155/2019/8194804] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/19/2019] [Accepted: 05/30/2019] [Indexed: 02/08/2023]
Abstract
Generally, inflammatory bowel disease (IBD) can be caused by psychology, genes, environment, and gut microbiota. Therefore, IBD therapy should be improved to utilize multiple strategies. Shen Ling Bai Zhu San (SLBZS) adheres to the aim of combating complex diseases from an integrative and holistic perspective, which is effective for IBD therapy. Herein, a systems pharmacology and microbiota approach was developed for these molecular mechanisms exemplified by SLBZS. First, by systematic absorption-distribution-metabolism-excretion (ADME) analysis, potential active compounds and their corresponding direct targets were retrieved. Then, the network relationships among the active compounds, targets, and disease were built to deduce the pharmacological actions of the drug. Finally, an “IBD pathway” consisting of several regulatory modules was proposed to dissect the therapeutic effects of SLBZS. In addition, the effects of SLBZS on gut microbiota were evaluated through analysis of the V3-V4 region and multivariate statistical methods. SLBZS significantly shifted the gut microbiota structure in a rat model. Taken together, we found that SLBZS has multidimensionality in the regulation of IBD-related physiological processes, which provides new sights into herbal medicine for the treatment of IBD.
Collapse
|
20
|
William M, Leroux LP, Chaparro V, Graber TE, Alain T, Jaramillo M. Translational repression of Ccl5 and Cxcl10 by 4E-BP1 and 4E-BP2 restrains the ability of mouse macrophages to induce migration of activated T cells. Eur J Immunol 2019; 49:1200-1212. [PMID: 31032899 DOI: 10.1002/eji.201847857] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 04/09/2019] [Accepted: 04/23/2019] [Indexed: 12/27/2022]
Abstract
Signaling through the mechanistic target of rapamycin complex 1 (mTORC1) is a major regulatory node of pro-inflammatory mediator production by macrophages (MΦs). However, it is still unclear whether such regulation relies on selective translational control by two of the main mTORC1 effectors, the eIF4E-binding proteins 1 and 2 (4E-BP1/2). By comparing translational efficiencies of immune-related transcripts of MΦs from WT and 4E-BP1/2 double-KO (DKO) mice, we found that translation of mRNAs encoding the pro-inflammatory chemokines CCL5 and CXCL10 is controlled by 4E-BP1/2. Macrophages deficient in 4E-BP1/2 produced higher levels of CCL5 and CXCL10 upon LPS stimulation, which enhanced chemoattraction of activated T cells. Consistent with this, treatment of WT cells with mTORC1 inhibitors promoted the activation of 4E-BP1/2 and reduced CCL5 and CXCL10 secretion. In contrast, the phosphorylation status of eIF4E did not affect the synthesis of these chemokines since MΦs derived from mice harboring a non-phosphorylatable form of the protein produced similar levels of CCL5 and CXCL10 to WT counterparts. These data provide evidence that the mTORC1-4E-BP1/2 axis contributes to regulate the production of chemoattractants by MΦs by limiting translation efficiency of Ccl5 and Cxcl10 mRNAs, and suggest that 4E-BP1/2 act as immunological safeguards by fine-tuning inflammatory responses in MΦs.
Collapse
Affiliation(s)
| | | | | | - Tyson E Graber
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Tommy Alain
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | | |
Collapse
|
21
|
Jiménez-Sousa MÁ, Gómez-Moreno AZ, Pineda-Tenor D, Sánchez-Ruano JJ, Artaza-Varasa T, Martin-Vicente M, Fernández-Rodríguez A, Martínez I, Resino S. Impact of DARC rs12075 Variants on Liver Fibrosis Progression in Patients with Chronic Hepatitis C: A Retrospective Study. Biomolecules 2019; 9:E143. [PMID: 30970632 PMCID: PMC6523653 DOI: 10.3390/biom9040143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/05/2019] [Accepted: 04/07/2019] [Indexed: 02/07/2023] Open
Abstract
: The Duffy antigen receptor for chemokines (DARC) rs12075 polymorphism regulates leukocyte trafficking and proinflammatory chemokine homeostasis. Hepatitis C virus (HCV)-mediated liver fibrosis is associated with an uncontrolled inflammatory response. In this study, we evaluate the association between the DARC rs12075 polymorphism and liver stiffness progression in HCV-infected patients. We carried out a retrospective cohort study (repeated measures design) in 208 noncirrhotic patients with chronic hepatitis C (CHC) who had at least two liver stiffness measurements (LSM) with a separation of at least 12 months. We used generalized linear models to analyze the association between DARC rs12075 polymorphism and outcome variables. During a follow-up of 46.6 months, the percentage of patients with stages of fibrosis F0/F1 decreased (p < 0.001), while LSM values and the percentage of patients with cirrhosis increased (p < 0.001). This pattern of changes was maintained in each of the groups of patients analyzed according to their rs12075 genotypes (AA or AG/GG). However, the variations in liver stiffness characteristics were lower in patients with the rs12075 AG/GG genotype (AG/GG versus AA). Thereby, in the adjusted analysis, patients with the rs12075 AG/GG genotype had a lower risk of an increased value of LSM2/LSM1 arithmetic mean ratio (AMR = 0.83; p = 0.001) and of an increase in LSM ≥ 5 kPa (odds ratio (OR) = 0.28; p = 0.009). Besides, patients with rs12075 AG/GG had a lower risk of cirrhosis progression (OR = 0.24; p = 0.009). No significant associations were found for an increase in LSM ≥ 10 kPa. We found an association between the DARC rs12075 single nucleotide polymorphism (SNP) and CHC progression. Specifically, patients with the DARC rs12075 AG/GG genotype had a lower risk of liver fibrosis progression and development of cirrhosis.
Collapse
Affiliation(s)
- María Ángeles Jiménez-Sousa
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Spain.
| | | | - Daniel Pineda-Tenor
- Servicio de Laboratorio Clínico, Hospital de Antequera, 29200 Málaga, Spain.
| | | | | | - María Martin-Vicente
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Spain.
| | - Amanda Fernández-Rodríguez
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Spain.
| | - Isidoro Martínez
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Spain.
| | - Salvador Resino
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Spain.
| |
Collapse
|
22
|
Walter A, Herrold AA, Gallagher VT, Lee R, Scaramuzzo M, Bream T, Seidenberg PH, Vandenbergh D, O'Connor K, Talavage TM, Nauman EA, Slobounov SM, Breiter HC. KIAA0319 Genotype Predicts the Number of Past Concussions in a Division I Football Team: A Pilot Study. J Neurotrauma 2019; 36:1115-1124. [DOI: 10.1089/neu.2017.5622] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Alexa Walter
- Department of Kinesiology, The Pennsylvania State University, University Park, Pennsylvania
- Concussion Neuroimaging Consortium, Florida State University, Florida; Harvard University, Massachusetts; Michigan State University, Michigan; Northwestern University, Illinois; Ohio State University, Ohio; Purdue University, Indiana; The Pennsylvania State University, Pennsylvania; University of Central Florida, Florida; University of Nebraska, Nebraska
| | - Amy A. Herrold
- Warren Wright Adolescent Center, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Edward Hines Jr., VA Hospital, Hines, Illinois
- Concussion Neuroimaging Consortium, Florida State University, Florida; Harvard University, Massachusetts; Michigan State University, Michigan; Northwestern University, Illinois; Ohio State University, Ohio; Purdue University, Indiana; The Pennsylvania State University, Pennsylvania; University of Central Florida, Florida; University of Nebraska, Nebraska
| | - Virginia T. Gallagher
- Warren Wright Adolescent Center, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Concussion Neuroimaging Consortium, Florida State University, Florida; Harvard University, Massachusetts; Michigan State University, Michigan; Northwestern University, Illinois; Ohio State University, Ohio; Purdue University, Indiana; The Pennsylvania State University, Pennsylvania; University of Central Florida, Florida; University of Nebraska, Nebraska
| | - Rosa Lee
- Warren Wright Adolescent Center, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Concussion Neuroimaging Consortium, Florida State University, Florida; Harvard University, Massachusetts; Michigan State University, Michigan; Northwestern University, Illinois; Ohio State University, Ohio; Purdue University, Indiana; The Pennsylvania State University, Pennsylvania; University of Central Florida, Florida; University of Nebraska, Nebraska
| | - Madeleine Scaramuzzo
- Athletic Department, The Pennsylvania State University, University Park, Pennsylvania
| | - Tim Bream
- Athletic Department, The Pennsylvania State University, University Park, Pennsylvania
| | - Peter H. Seidenberg
- Athletic Department, The Pennsylvania State University, University Park, Pennsylvania
| | - David Vandenbergh
- Department of Biobehavioral Health, Molecular and Cellular Biosciences Program and Institute for the Neurosciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Kailyn O'Connor
- Warren Wright Adolescent Center, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Thomas M. Talavage
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, Indiana
- Concussion Neuroimaging Consortium, Florida State University, Florida; Harvard University, Massachusetts; Michigan State University, Michigan; Northwestern University, Illinois; Ohio State University, Ohio; Purdue University, Indiana; The Pennsylvania State University, Pennsylvania; University of Central Florida, Florida; University of Nebraska, Nebraska
| | - Eric A. Nauman
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana
- Concussion Neuroimaging Consortium, Florida State University, Florida; Harvard University, Massachusetts; Michigan State University, Michigan; Northwestern University, Illinois; Ohio State University, Ohio; Purdue University, Indiana; The Pennsylvania State University, Pennsylvania; University of Central Florida, Florida; University of Nebraska, Nebraska
| | - Semyon M. Slobounov
- Department of Kinesiology, The Pennsylvania State University, University Park, Pennsylvania
- Concussion Neuroimaging Consortium, Florida State University, Florida; Harvard University, Massachusetts; Michigan State University, Michigan; Northwestern University, Illinois; Ohio State University, Ohio; Purdue University, Indiana; The Pennsylvania State University, Pennsylvania; University of Central Florida, Florida; University of Nebraska, Nebraska
| | - Hans C. Breiter
- Warren Wright Adolescent Center, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Laboratory of Neuroimaging and Genetics, Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts
- Concussion Neuroimaging Consortium, Florida State University, Florida; Harvard University, Massachusetts; Michigan State University, Michigan; Northwestern University, Illinois; Ohio State University, Ohio; Purdue University, Indiana; The Pennsylvania State University, Pennsylvania; University of Central Florida, Florida; University of Nebraska, Nebraska
| |
Collapse
|
23
|
Rappoport N, Simon AJ, Amariglio N, Rechavi G. The Duffy antigen receptor for chemokines, ACKR1,- 'Jeanne DARC' of benign neutropenia. Br J Haematol 2018; 184:497-507. [PMID: 30592023 DOI: 10.1111/bjh.15730] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Benign neutropenia, observed in different ethnic groups, is the most common form of neutropenia worldwide. A specific single nucleotide polymorphism, rs2814778, located at the promoter of the ACKR1 (previously termed DARC) gene, which disrupts a binding site for the GATA1 erythroid transcription factor, resulting in a ACKR1-null phenotype, was found to serve as a predictor of low white blood cell and neutrophil counts in African-Americans and Yemenite Jews. Individuals with benign neutropenia due to the ACKR1-null allele have been found to have an increased susceptibility to human immunodeficiency virus infection and, on the other hand, a protective effect against malaria. The associated protective effect may explain the spread of the ACKR1-null allele by natural selection. The reviewed relationships between ACKR1 polymorphism and various pathological states may have important clinical implications to individuals with and without benign neutropenia. Potential mechanisms for ACKR1 (previously termed DARC) modulation during neutrophil recruitment to inflammation, and chemokine bioavailability in the circulation and in local tissue are reviewed and discussed.
Collapse
Affiliation(s)
- Naama Rappoport
- Cancer Research Centre, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amos J Simon
- Cancer Research Centre, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Institute of Haematology, Sheba Medical Centre, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ninette Amariglio
- Cancer Research Centre, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Institute of Haematology, Sheba Medical Centre, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat-Gan, Israel
| | - Gideon Rechavi
- Cancer Research Centre, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
24
|
Unfolding of hidden white blood cell count phenotypes for gene discovery using latent class mixed modeling. Genes Immun 2018; 20:555-565. [PMID: 30459343 DOI: 10.1038/s41435-018-0051-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 09/24/2018] [Accepted: 10/24/2018] [Indexed: 12/26/2022]
Abstract
Resting-state white blood cell (WBC) count is a marker of inflammation and immune system health. There is evidence that WBC count is not fixed over time and there is heterogeneity in WBC trajectory that is associated with morbidity and mortality. Latent class mixed modeling (LCMM) is a method that can identify unobserved heterogeneity in longitudinal data and attempts to classify individuals into groups based on a linear model of repeated measurements. We applied LCMM to repeated WBC count measures derived from electronic medical records of participants of the National Human Genetics Research Institute (NHRGI) electronic MEdical Record and GEnomics (eMERGE) network study, revealing two WBC count trajectory phenotypes. Advancing these phenotypes to GWAS, we found genetic associations between trajectory class membership and regions on chromosome 1p34.3 and chromosome 11q13.4. The chromosome 1 region contains CSF3R, which encodes the granulocyte colony-stimulating factor receptor. This protein is a major factor in neutrophil stimulation and proliferation. The association on chromosome 11 contain genes RNF169 and XRRA1; both involved in the regulation of double-strand break DNA repair.
Collapse
|
25
|
Pan L, Lv J, Zhang Z, Zhang Y. Adaptation and Constraint in the Atypical Chemokine Receptor Family in Mammals. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9065181. [PMID: 30345310 PMCID: PMC6174752 DOI: 10.1155/2018/9065181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/24/2018] [Accepted: 09/04/2018] [Indexed: 12/22/2022]
Abstract
Atypical chemokine receptors (ACKRs) are a subclass of G protein-coupled receptors characterized by promiscuity of ligand binding and an obvious inability to signal after ligand binding. Although some discoveries regarding this family in Homo sapiens and other species have been reported in some studies, the evolution and function of multiple ACKR in mammals have not yet been clearly understood. We performed an evolutionary analysis of ACKR genes (ACKR1, ACKR2, ACKR3, and ACKR4) in mammals. Ninety-two full-length ACKR genes from 27 mammal species were retrieved from the Genbank and Ensemble databases. Phylogenetic analysis showed that there were four well-conserved subfamilies in mammals. Synteny analysis revealed that ACKR genes formed conserved linkage groups with their adjacent genes across mammalian species, facilitating the identification of ACKRs in as yet unannotated genome datasets. Analysis of the site-specific profiles established by posterior probability revealed the positive-selection sites to be distributed mainly in the ligand binding region of ACKR1. This study highlights the molecular evolution of the ACKR gene family in mammals and identifies the critical amino acid residues likely to be relevant to ligand binding. Further experimental verification of these findings may provide valuable information regarding the ACKR's biochemical and physiological functions.
Collapse
Affiliation(s)
- Li Pan
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No.1 Xujiaping, Yangchangbu, Chengguan District, Lanzhou 730046, Gansu, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
| | - Jianliang Lv
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No.1 Xujiaping, Yangchangbu, Chengguan District, Lanzhou 730046, Gansu, China
| | - Zhongwang Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No.1 Xujiaping, Yangchangbu, Chengguan District, Lanzhou 730046, Gansu, China
| | - Yongguang Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No.1 Xujiaping, Yangchangbu, Chengguan District, Lanzhou 730046, Gansu, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
| |
Collapse
|
26
|
Yao S, Hong CC, Ruiz-Narváez EA, Evans SS, Zhu Q, Schaefer BA, Yan L, Coignet MV, Lunetta KL, Sucheston-Campbell LE, Lee K, Bandera EV, Troester MA, Rosenberg L, Palmer JR, Olshan AF, Ambrosone CB. Genetic ancestry and population differences in levels of inflammatory cytokines in women: Role for evolutionary selection and environmental factors. PLoS Genet 2018; 14:e1007368. [PMID: 29879116 PMCID: PMC5991662 DOI: 10.1371/journal.pgen.1007368] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/18/2018] [Indexed: 01/09/2023] Open
Abstract
Selection pressure due to exposure to infectious pathogens endemic to Africa may explain distinct genetic variations in immune response genes. However, the impact of those genetic variations on human immunity remains understudied, especially within the context of modern lifestyles and living environments, which are drastically different from early humans in sub Saharan Africa. There are few data on population differences in constitutional immune environment, where genetic ancestry and environment are likely two primary sources of variation. In a study integrating genetic, molecular and epidemiologic data, we examined population differences in plasma levels of 14 cytokines involved in innate and adaptive immunity, including those implicated in chronic inflammation, and possible contributing factors to such differences, in 914 AA and 855 EA women. We observed significant differences in 7 cytokines, including higher plasma levels of CCL2, CCL11, IL4 and IL10 in EAs and higher levels of IL1RA and IFNα2 in AAs. Analyses of a wide range of demographic and lifestyle factors showed significant impact, with age, education level, obesity, smoking, and alcohol intake, accounting for some, but not all, observed population differences for the cytokines examined. Levels of two pro-inflammatory chemokines, CCL2 and CCL11, were strongly associated with percent of African ancestry among AAs. Through admixture mapping, the signal was pinpointed to local ancestry at 1q23, with fine-mapping analysis refined to the Duffy-null allele of rs2814778. In AA women, this variant was a major determinant of systemic levels of CCL2 (p = 1.1e-58) and CCL11 (p = 2.2e-110), accounting for 19% and 40% of the phenotypic variance, respectively. Our data reveal strong ancestral footprints in inflammatory chemokine regulation. The Duffy-null allele may indicate a loss of the buffering function for chemokine levels. The substantial immune differences by ancestry may have broad implications to health disparities between AA and EA populations.
Collapse
Affiliation(s)
- Song Yao
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States of America
| | - Chi-Chen Hong
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States of America
| | - Edward A. Ruiz-Narváez
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States of America
| | - Sharon S. Evans
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States of America
| | - Qianqian Zhu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States of America
| | - Beverly A. Schaefer
- Department of Pediatric Hematology & Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States of America
- Department of Pediatric Hematology & Oncology, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, Buffalo, NY, United States of America
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States of America
| | - Marie V. Coignet
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States of America
| | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States of America
| | | | - Kelvin Lee
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States of America
| | - Elisa V. Bandera
- Cancer Prevention and Control Program, Rutgers Cancer Institute of New Jersey, The State University of New Jersey, New Brunswick, NJ, United States of America
| | - Melissa A. Troester
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Lynn Rosenberg
- Slone Epidemiology Center at Boston University, Boston, MA, United States of America
| | - Julie R. Palmer
- Slone Epidemiology Center at Boston University, Boston, MA, United States of America
| | - Andrew F. Olshan
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Christine B. Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States of America
- * E-mail:
| |
Collapse
|
27
|
Metzemaekers M, Vanheule V, Janssens R, Struyf S, Proost P. Overview of the Mechanisms that May Contribute to the Non-Redundant Activities of Interferon-Inducible CXC Chemokine Receptor 3 Ligands. Front Immunol 2018; 8:1970. [PMID: 29379506 PMCID: PMC5775283 DOI: 10.3389/fimmu.2017.01970] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022] Open
Abstract
The inflammatory chemokines CXCL9, CXCL10, and CXCL11 are predominantly induced by interferon (IFN)-γ and share an exclusive chemokine receptor named CXC chemokine receptor 3 (CXCR3). With a prototype function of directing temporal and spatial migration of activated T cells and natural killer cells, and inhibitory effects on angiogenesis, these CXCR3 ligands have been implicated in infection, acute inflammation, autoinflammation and autoimmunity, as well as in cancer. Intense former research efforts led to recent and ongoing clinical trials using CXCR3 and CXCR3 ligand targeting molecules. Scientific evidence has claimed mutual redundancy, ligand dominance, collaboration or even antagonism, depending on the (patho)physiological context. Most research on their in vivo activity, however, illustrates that CXCL9, CXCL10, and CXCL11 each contribute to the activation and trafficking of CXCR3 expressing cells in a non-redundant manner. When looking into detail, one can unravel a multistep machinery behind final CXCR3 ligand functions. Not only can specific cell types secrete individual CXCR3 interacting chemokines in response to certain stimuli, but also the receptor and glycosaminoglycan interactions, major associated intracellular pathways and susceptibility to processing by particular enzymes, among others, seem ligand-specific. Here, we overview major aspects of the molecular properties and regulatory mechanisms of IFN-induced CXCR3 ligands, and propose that their in vivo non-redundancy is a reflection of the unprecedented degree of versatility that seems inherent to the IFN-related CXCR3 chemokine system.
Collapse
Affiliation(s)
- Mieke Metzemaekers
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Vincent Vanheule
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Rik Janssens
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
28
|
Ansel A, Lewis JD, Melnick DJ, Martins C, Valladares-Padua C, Perez-Sweeney B. Genetic variants related to disease susceptibility and immunotolerance in the Duffy antigen receptor for chemokines (DARC, Fy) gene in the black lion tamarin (Leontopithecus chrysopygus, primates). Am J Primatol 2017; 79. [PMID: 28902417 DOI: 10.1002/ajp.22690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/01/2017] [Accepted: 08/03/2017] [Indexed: 12/27/2022]
Abstract
The DARC (Duffy antigen receptor for chemokines) gene encodes the DARC protein, which serves multiple roles in the immune system, as a binding site for the malarial parasites Plasmodium vivax and Plasmodium knowlesi, a promiscuous chemokine receptor and a blood group antigen. Variation in DARC may play particularly significant roles in innate immunity, immunotolerance and pathogen entry in callitrichines, such as the black lion tamarin (Leontopithecus chrysopygus). We compared amino acid sequences of DARC in the black lion tamarin (BLT) to non-human Haplorhine primates and Homo sapiens. Consistent with prior studies in other Haplorhines, we observed that the chemokine receptor experiences two opposing selection forces: (1) positive selection on the Plasmodium binding site and (2) purifying selection. We observed also that D21N, F22L, and V25L differentiated BLT from humans at a critical site for P. vivax and P. knowlesi binding. One amino acid residue, F22L, was subject to both positive selection and fixation in New World monkeys, suggesting a beneficial role as an adaptive barrier to Plasmodium entry. Unlike in humans, we observed no variation in DARC among BLTs, suggesting that the protein does not play a role in immunotolerance. In addition, lion tamarins differed from humans at the blood compatibility Fya /Fyb antigen-binding site 44, as well as at the putative destabilizing residues A61, T68, A187, and L215, further supporting a difference in the functional role of DARC in these primates compared with humans. Further research is needed to determine whether changes in the Plasmodium and Fya /Fyb antigen-binding sites disrupt DARC function in callitrichines.
Collapse
Affiliation(s)
- Ashley Ansel
- Department of Biological Sciences, Fordham University, Bronx, New York
| | - James D Lewis
- Department of Biological Sciences, Fordham University, Bronx, New York.,Louis Calder Center-Biological Field Station, Fordham University, Armonk, New York
| | - Don J Melnick
- Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, New York
| | - Cristiana Martins
- IPÊ Instituto de Pesquisas Ecológicas, Escola Superior de Conservação Ambiental e Sustentabilidade-ESCAS, Nazaré Paulista, Brazil
| | - Claudio Valladares-Padua
- IPÊ Instituto de Pesquisas Ecológicas, Escola Superior de Conservação Ambiental e Sustentabilidade-ESCAS, Nazaré Paulista, Brazil
| | - Beatriz Perez-Sweeney
- Department of Biological Sciences, Fordham University, Bronx, New York.,Louis Calder Center-Biological Field Station, Fordham University, Armonk, New York.,Baylor College of Medicine, Houston, Texas
| |
Collapse
|
29
|
Chapman DG, Mougey EB, Van der Velden JL, Lahue KG, Aliyeva M, Daphtary N, George KL, Hoffman SM, Schneider RW, Tracy RP, Worthen GS, Poynter ME, Peters SP, Lima JJ, Janssen-Heininger YMW, Irvin CG. The Duffy antigen receptor for chemokines regulates asthma pathophysiology. Clin Exp Allergy 2017; 47:1214-1222. [PMID: 28471517 PMCID: PMC5578916 DOI: 10.1111/cea.12949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/21/2017] [Accepted: 03/27/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND The Duffy antigen receptor for chemokines (DARC) is an atypical receptor that regulates pro-inflammatory cytokines. However, the role of DARC in asthma pathophysiology is unknown. OBJECTIVE To determine the role of DARC in allergic airways disease in mice, and the association between DARC single nucleotide polymorphisms (SNPs) and clinical outcomes in patients with asthma. METHODS Mice with targeted disruption of the Darc gene (Darc∆E2 ) or WT mice were challenged over 3 weeks with house dust mite (HDM) antigen. Allergic airways disease was assessed 24 hours and 7 days following the final challenge. Additionally, associations between DARC SNPs and clinical outcomes were analysed in a cohort of poorly controlled asthmatics. RESULTS Total airway inflammation following HDM did not differ between Darc∆E2 and WT mice. At 24 hours, Darc∆E2 mice had increased airway hyperresponsiveness; however, at 7 days airway hyperresponsiveness had completely resolved in Darc∆E2 but persisted in WT mice. In poorly controlled asthmatics, DARC SNPs were associated with worse asthma control at randomization and subsequent increased risk of healthcare utilization (odds ratio 3.13(1.37-7.27), P=.0062). CONCLUSIONS AND CLINICAL RELEVANCE Our animal model and human patient data suggest a novel role for DARC in the temporal regulation in asthma pathophysiology and symptoms.
Collapse
Affiliation(s)
- D G Chapman
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
- Woolcock Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - E B Mougey
- Nemours Pharmacogenetics Center, Nemours Children's Clinic, Jacksonville, FL, USA
| | - J L Van der Velden
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA
| | - K G Lahue
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA
| | - M Aliyeva
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - N Daphtary
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - K L George
- Nemours Pharmacogenetics Center, Nemours Children's Clinic, Jacksonville, FL, USA
| | - S M Hoffman
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA
| | - R W Schneider
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA
| | - R P Tracy
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
- Department of Biochemistry, University of Vermont College of Medicine, Burlington, VT, USA
| | - G S Worthen
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - M E Poynter
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - S P Peters
- Section on Pulmonary, Critical Care, Allergy & Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - J J Lima
- Nemours Pharmacogenetics Center, Nemours Children's Clinic, Jacksonville, FL, USA
| | | | - C G Irvin
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| |
Collapse
|
30
|
Duchene J, Novitzky-Basso I, Thiriot A, Casanova-Acebes M, Bianchini M, Etheridge SL, Hub E, Nitz K, Artinger K, Eller K, Caamaño J, Rülicke T, Moss P, Megens RTA, von Andrian UH, Hidalgo A, Weber C, Rot A. Atypical chemokine receptor 1 on nucleated erythroid cells regulates hematopoiesis. Nat Immunol 2017; 18:753-761. [PMID: 28553950 PMCID: PMC5480598 DOI: 10.1038/ni.3763] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 04/28/2017] [Indexed: 12/14/2022]
Abstract
Healthy individuals of African ancestry have neutropenia that has been linked with the variant rs2814778(G) of the gene encoding atypical chemokine receptor 1 (ACKR1). This polymorphism selectively abolishes the expression of ACKR1 in erythroid cells, causing a Duffy-negative phenotype. Here we describe an unexpected fundamental role for ACKR1 in hematopoiesis and provide the mechanism that links its absence with neutropenia. Nucleated erythroid cells had high expression of ACKR1, which facilitated their direct contact with hematopoietic stem cells. The absence of erythroid ACKR1 altered mouse hematopoiesis including stem and progenitor cells, which ultimately gave rise to phenotypically distinct neutrophils that readily left the circulation, causing neutropenia. Individuals with a Duffy-negative phenotype developed a distinct profile of neutrophil effector molecules that closely reflected the one observed in the ACKR1-deficient mice. Thus, alternative physiological patterns of hematopoiesis and bone marrow cell outputs depend on the expression of ACKR1 in the erythroid lineage, findings with major implications for the selection advantages that have resulted in the paramount fixation of the ACKR1 rs2814778(G) polymorphism in Africa.
Collapse
Affiliation(s)
- Johan Duchene
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Igor Novitzky-Basso
- Blood and Marrow Transplant Unit, Queen Elizabeth University Hospital, Glasgow UK
| | - Aude Thiriot
- Department of Microbiology and Immunobiology and Center for Immune Imaging, Harvard Medical School, Boston, MA, USA
- The Ragon Institute, Cambridge, MA, USA
| | - Maria Casanova-Acebes
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Mariaelvy Bianchini
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - S. Leah Etheridge
- Centre for Immunology and Infection, Department of Biology, University of York, Heslington, York, UK
| | - Elin Hub
- Centre for Immunology and Infection, Department of Biology, University of York, Heslington, York, UK
| | - Katrin Nitz
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Katharina Artinger
- Centre for Immunology and Infection, Department of Biology, University of York, Heslington, York, UK
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Kathrin Eller
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Jorge Caamaño
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Thomas Rülicke
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Paul Moss
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Remco T. A. Megens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
- Cardiovascular Research Institute Maastricht, University of Maastricht, Maastricht, The Netherlands
| | - Ulrich H. von Andrian
- Department of Microbiology and Immunobiology and Center for Immune Imaging, Harvard Medical School, Boston, MA, USA
- The Ragon Institute, Cambridge, MA, USA
| | - Andres Hidalgo
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
- Cardiovascular Research Institute Maastricht, University of Maastricht, Maastricht, The Netherlands
| | - Antal Rot
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
- Centre for Immunology and Infection, Department of Biology, University of York, Heslington, York, UK
- Center for Advanced Studies, Ludwig-Maximilians-University, Munich, Germany
- Address from July 2017: William Harvey Research Institute, Queen Mary University of London. London, UK
| |
Collapse
|
31
|
Abstract
Chemokines are a family of small cytokines that share a typical key structure that is stabilized by disulfide bonds between the cysteine residues at the NH2-terminal of the protein, and they are secreted by a great variety of cells in several different conditions. Their function is directly dependent on their interactions with their receptors. Chemokines are involved in cell maturation and differentiation, infection, autoimmunity, cancer, and, in general, in any situation where immune components are involved. However, their role in postfracture inflammation and fracture healing is not yet well established. In this article, we will discuss the response of chemokines to bone fracture and their potential roles in postfracture inflammation and healing based on data from our studies and from other previously published studies.
Collapse
Affiliation(s)
- Bouchra Edderkaoui
- Musculoskeletal Disease Center, Loma Linda VA Health Care Systems, Loma Linda, CA, USA
- Department of Medicine, Loma Linda University, Loma Linda, CA, USA
- *Correspondence: Bouchra Edderkaoui,
| |
Collapse
|
32
|
Vacchini A, Locati M, Borroni EM. Overview and potential unifying themes of the atypical chemokine receptor family. J Leukoc Biol 2016; 99:883-92. [PMID: 26740381 DOI: 10.1189/jlb.2mr1015-477r] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/12/2015] [Indexed: 12/17/2022] Open
Abstract
Chemokines modulate immune responses through their ability to orchestrate the migration of target cells. Chemokines directly induce cell migration through a distinct set of 7 transmembrane domain G protein-coupled receptors but are also recognized by a small subfamily of atypical chemokine receptors, characterized by their inability to support chemotactic activity. Atypical chemokine receptors are now emerging as crucial regulatory components of chemokine networks in a wide range of physiologic and pathologic contexts. Although a new nomenclature has been approved recently to reflect their functional distinction from their conventional counterparts, a systematic view of this subfamily is still missing. This review discusses their biochemical and immunologic properties to identify potential unifying themes in this emerging family.
Collapse
Affiliation(s)
- Alessandro Vacchini
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, and Humanitas Clinical and Research Center, Milan, Italy
| | - Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, and Humanitas Clinical and Research Center, Milan, Italy
| | - Elena Monica Borroni
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, and Humanitas Clinical and Research Center, Milan, Italy
| |
Collapse
|
33
|
Santulli-Marotto S, Wheeler J, Lacy ER, Boakye K, Luongo J, Wu SJ, Ryan M. CCL22-specific Antibodies Reveal That Engagement of Two Distinct Binding Domains on CCL22 Is Required for CCR4-mediated Function. Monoclon Antib Immunodiagn Immunother 2015; 34:373-80. [DOI: 10.1089/mab.2015.0039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
| | - John Wheeler
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Eilyn R. Lacy
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Ken Boakye
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Jennifer Luongo
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Sheng-Jiun Wu
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Mary Ryan
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| |
Collapse
|
34
|
Association of haemolytic uraemic syndrome with dysregulation of chemokine receptor expression in circulating monocytes. Clin Sci (Lond) 2015; 129:235-44. [PMID: 25748554 DOI: 10.1042/cs20150016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Haemolytic uraemic syndrome (HUS) is the major complication of Escherichia coli gastrointestinal infections that are Shiga toxin (Stx) producing. Monocytes contribute to HUS evolution by producing cytokines that sensitize endothelial cells to Stx action and migration to the injured kidney. As CC chemokine receptors (CCRs) are involved in monocyte recruitment to injured tissue, we analysed the contribution of these receptors to the pathogenesis of HUS. We analysed CCR1, CCR2 and CCR5 expression in peripheral monocytes from HUS patients during the acute period, with healthy children as controls. We observed an increased expression of CCRs per cell in monocytes from HUS patients, accompanied by an increase in the absolute number of monocytes CCR1+, CCR2+ and CCR5+. It is interesting that prospective analysis confirmed that CCR1 expression positively correlated with HUS severity. The evaluation of chemokine levels in plasma showed that regulated on activation of normal T-cell-expressed and -secreted (RANTES) protein was reduced in plasma from patients with severe HUS, and this decrease correlated with thrombocytopenia. Finally, the expression of the higher CCRs was accompanied by a loss of functionality which could be due to a mechanism for desensitization to compensate for altered receptor expression. The increase in CCR expression correlates with HUS severity, suggesting that the dysregulation of these receptors might contribute to an increased risk of renal damage. Activated monocytes could be recruited by chemokines and then receptors could be dysregulated. The dysregulation of CCRs and their ligands observed during the acute period suggests that a chemokine pathway would participate in HUS development.
Collapse
|
35
|
Chen Y, Liao N, Lu F, Peng H, Gao J. The role of Duffy antigen receptor for chemokines in keloids. Gene 2015; 570:44-9. [PMID: 26045366 DOI: 10.1016/j.gene.2015.05.071] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 05/27/2015] [Accepted: 05/30/2015] [Indexed: 11/17/2022]
Abstract
This study aims to determine the relationship between Duffy antigen receptor for chemokines (DARC) and keloid pathogenesis. DARC expression was determined by immunohistochemistry, real-time PCR, and Western blot analysis. Cell proliferation was assessed by CCK-8 assay. Cell migration and invasion abilities were measured by the shift assay. Levels of CC chemokine ligand 2 (CCL2), CXC chemokine ligand 8 (CXCL8), and matrix metalloproteinase 2 (MMP2) were detected by real-time PCR and ELISA. Our results showed that DARC levels were elevated in human keloid fibroblasts. After knocking down DARC, cell proliferation was not altered, whereas the migration and invasion abilities of keloid fibroblasts were significantly elevated. Additionally, the mRNA expression levels of CCL2, CXCL8, and MMP2 were not influenced by DARC knockdown. However, the secretion of CCL2, but not CXCL8 or MMP2, was significantly increased after DARC knockdown. Our results suggest that DARC might inhibit the secretion of CCL2. Moreover, DARC knockdown increases the migration and invasion abilities of keloid fibroblasts.
Collapse
Affiliation(s)
- Ying Chen
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China; Department of Breast Surgery, Guangdong Traditional Chinese Medicine Hospital, Guangzhou 510000, Guangdong, China
| | - Nong Liao
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China; Department of Plastic Surgery, The Third Affiliated Hospital of Guangzhou Medical College, Guangzhou 510150, Guangdong, China
| | - Feng Lu
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Hui Peng
- Department of Pathology, Guangdong Traditional Chinese Medicine Hospital, Guangzhou 510000, Guangdong, China
| | - Jianhua Gao
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China.
| |
Collapse
|
36
|
Van Raemdonck K, Van den Steen PE, Liekens S, Van Damme J, Struyf S. CXCR3 ligands in disease and therapy. Cytokine Growth Factor Rev 2015; 26:311-27. [DOI: 10.1016/j.cytogfr.2014.11.009] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 11/05/2014] [Indexed: 12/19/2022]
|
37
|
Stateman WA, Henkin RI, Knöppel AB, Flegel WA. Erythrocyte membrane antigen frequencies in patients with Type II congenital smell loss. Am J Otolaryngol 2015; 36:146-52. [PMID: 25456515 DOI: 10.1016/j.amjoto.2014.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 10/04/2014] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The objective of this study was to determine whether there are genetic factors associated with Type II congenital smell loss. STUDY DESIGN The expression frequencies of 16 erythrocyte antigens among patients with Type II congenital smell loss were determined and compared to those of a large control group. METHODS Blood samples were obtained from 99 patients with Type II congenital smell loss. Presence of the erythrocyte surface antigens A, B, M, N, S, s, Fy(a), Fy(b), D, C, c, E, e, K, Jk(a), and Jk(b) was analyzed by blood group serology. Comparisons of expression frequencies of these antigens were made between the patients and a large control group. RESULTS Patients tested for the Duffy b antigen (Fy(b) haplotype) exhibited a statistically significant 11% decrease in expression frequency compared to the controls. There were no significant differences between patients and controls in the expression frequencies for all other erythrocyte antigens (A, B, M, N, S, s, Fy(a), D, C, c, E, e, K, Jk(a), or Jk(b)). CONCLUSIONS These findings describe the presence of a previously unrevealed genetic tendency among patients with Type II congenital smell loss related to erythrocyte surface antigen expression. The deviation in expression rate of Duffy b suggests a target gene and chromosome region in which future research into this form of congenital smell loss may reveal a more specific genetic basis for Type II congenital smell loss.
Collapse
|
38
|
Studies of a murine monoclonal antibody directed against DARC: reappraisal of its specificity. PLoS One 2015; 10:e0116472. [PMID: 25706384 PMCID: PMC4338028 DOI: 10.1371/journal.pone.0116472] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 12/09/2014] [Indexed: 12/29/2022] Open
Abstract
Duffy Antigen Receptor for Chemokines (DARC) plays multiple roles in human health as a blood group antigen, a receptor for chemokines and the only known receptor for Plasmodium vivax merozoites. It is the target of the murine anti-Fy6 monoclonal antibody 2C3 which binds to the first extracellular domain (ECD1), but exact nature of the recognized epitope was a subject of contradictory reports. Here, using a set of complex experiments which include expression of DARC with amino acid substitutions within the Fy6 epitope in E. coli and K562 cells, ELISA, surface plasmon resonance (SPR) and flow cytometry, we have resolved discrepancies between previously published reports and show that the basic epitope recognized by 2C3 antibody is 22FEDVW26, with 22F and 26W being the most important residues. In addition, we demonstrated that 30Y plays an auxiliary role in binding, particularly when the residue is sulfated. The STD-NMR studies performed using 2C3-derived Fab and synthetic peptide corroborated most of these results, and together with the molecular modelling suggested that 25V is not involved in direct interactions with the antibody, but determines folding of the epitope backbone.
Collapse
|
39
|
Che JN, Nmorsi OPG, Nkot BP, Isaac C, Okonkwo BC. Chemokines responses to Plasmodium falciparum malaria and co-infections among rural Cameroonians. Parasitol Int 2014; 64:139-44. [PMID: 25462711 DOI: 10.1016/j.parint.2014.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/10/2014] [Accepted: 11/06/2014] [Indexed: 10/24/2022]
Abstract
Malaria remains the major cause of disease morbidity and mortality in sub-Saharan Africa with complex immune responses associated with disease outcomes. Symptoms associated with severe malaria have generally shown chemokine upregulation but little is known of responses to uncomplicated malaria. Eight villages in central Cameroon of 1045 volunteers were screened. Among these, malaria-positive individuals with some healthy controls were selected for chemokine analysis using Enzyme-Linked Immunosorbent Assay (ELISA) kits. Depressed serum levels of CXCL5 and raised CCL28 were observed in malarial positives when compared with healthy controls. The mean concentration of CXCL11 was higher in symptomatic than asymptomatic group, while CCL28 was lower in symptomatic individuals. Lower chemokine levels were associated with symptoms of uncomplicated malaria except for CXCL11 which was upregulated among fever-positive group. The mean CXCL5 level was higher in malaria sole infection than co-infections with HIV and Loa loa. Also, there was a raised mean level of malaria+HIV co-infection for CXCL9. This study hypothesises a situation where depressed chemokines in the face of clinical presentations could indicate an attempt by the immune system in preventing a progression process from uncomplicated to complicated outcomes with CXCL11 being identified as possible biomarker for malarial fever.
Collapse
Affiliation(s)
- Jane Nchangnwi Che
- Tropical Disease Research Unit, Department of Zoology, Ambrose Alli University, Ekpoma, Nigeria; Centre for the Diagnosis and Control of Tropical Disease, Nkolbisson, Yaounde, Cameroon
| | | | - Baleguel Pierre Nkot
- Centre for the Diagnosis and Control of Tropical Disease, Nkolbisson, Yaounde, Cameroon
| | - Clement Isaac
- Tropical Disease Research Unit, Department of Zoology, Ambrose Alli University, Ekpoma, Nigeria.
| | | |
Collapse
|
40
|
Velasco-Velázquez M, Xolalpa W, Pestell RG. The potential to target CCL5/CCR5 in breast cancer. Expert Opin Ther Targets 2014; 18:1265-75. [PMID: 25256399 DOI: 10.1517/14728222.2014.949238] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
41
|
Abstract
Lymphatic vessels (LVs) are involved in a number of physiological and pathophysiological processes such as fluid homoeostasis, immune surveillance, and resolution of inflammation and wound healing. Lymphangiogenesis, the outgrowth of existing LVs and the formation of new ones, has received increasing attention over the past decade on account of its prominence in organ physiology and pathology, which has been enabled by the development of specific tools to study lymph vessel functions. Several studies have been devoted to renal lymphatic vasculature and lymphangiogenesis in kidney diseases, such as chronic renal transplant dysfunction, primary renal fibrotic disorders, proteinuria, diabetic nephropathy and renal inflammation. This review describes the most recent findings on lymphangiogenesis, with a specific focus on renal lymphangiogenesis and its impact on renal diseases. We suggest renal lymphatics as a possible target for therapeutic interventions in renal medicine to dampen tubulointerstitial tissue remodelling and improve renal functioning.
Collapse
|
42
|
Schneider EH, Fowler SC, Lionakis MS, Swamydas M, Holmes G, Diaz V, Munasinghe J, Peiper SC, Gao JL, Murphy PM. Regulation of motor function and behavior by atypical chemokine receptor 1. Behav Genet 2014; 44:498-515. [PMID: 24997773 PMCID: PMC4790732 DOI: 10.1007/s10519-014-9665-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 06/04/2014] [Indexed: 12/14/2022]
Abstract
Atypical Chemokine Receptor 1 (ACKR1), previously known as Duffy Antigen Receptor for Chemokines, stands out among chemokine receptors for high selective expression on cerebellar Purkinje neurons. Although ACKR1 ligands activate Purkinje cells in vitro, evidence for ACKR1 regulation of brain function in vivo is lacking. Here we demonstrate that Ackr1 (-/-) mice have markedly impaired balance and ataxia on a rotating rod and increased tremor when injected with harmaline, which induces whole-body tremor by activating Purkinje cells. Ackr1 (-/-) mice also exhibited impaired exploratory behavior, increased anxiety-like behavior and frequent episodes of marked hypoactivity under low-stress conditions. Surprisingly, Ackr1 (+/-) had similar behavioral abnormalities, indicating pronounced haploinsufficiency. The behavioral phenotype of Ackr1 (-/-) mice was the opposite of mouse models of cerebellar degeneration, and the defects persisted when Ackr1 was deficient only on non-hematopoietic cells. Together, the results suggest that normal motor function and behavior may partly depend on negative regulation of Purkinje cell activity by Ackr1.
Collapse
Affiliation(s)
- Erich H. Schneider
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases (NIAID)/NIH, Bethesda, MD, USA
| | - Stephen C. Fowler
- Department of Pharmacology & Toxicology, University of Kansas, Lawrence, KS, USA
| | - Michail S. Lionakis
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases (NIAID)/NIH, Bethesda, MD, USA
| | - Muthulekha Swamydas
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases (NIAID)/NIH, Bethesda, MD, USA
| | - Gibran Holmes
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases (NIAID)/NIH, Bethesda, MD, USA
| | - Vivian Diaz
- In Vivo NMR Center, National Institute of Neurological Diseases and Stroke (NINDS)/NIH, Bethesda, MD, USA
| | - Jeeva Munasinghe
- In Vivo NMR Center, National Institute of Neurological Diseases and Stroke (NINDS)/NIH, Bethesda, MD, USA
| | - Stephen C. Peiper
- Institute of Pathology, Anatomy & Cell Biology, Jefferson Medical College, Philadelphia, PA, USA
| | - Ji-Liang Gao
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases (NIAID)/NIH, Bethesda, MD, USA
| | - Philip M. Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases (NIAID)/NIH, Bethesda, MD, USA
| |
Collapse
|
43
|
Minten C, Alt C, Gentner M, Frei E, Deutsch U, Lyck R, Schaeren-Wiemers N, Rot A, Engelhardt B. DARC shuttles inflammatory chemokines across the blood-brain barrier during autoimmune central nervous system inflammation. Brain 2014; 137:1454-69. [PMID: 24625696 PMCID: PMC3999718 DOI: 10.1093/brain/awu045] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 12/30/2013] [Accepted: 01/14/2014] [Indexed: 12/14/2022] Open
Abstract
The Duffy antigen/receptor for chemokines, DARC, belongs to the family of atypical heptahelical chemokine receptors that do not couple to G proteins and therefore fail to transmit conventional intracellular signals. Here we show that during experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, the expression of DARC is upregulated at the blood-brain barrier. These findings are corroborated by the presence of a significantly increased number of subcortical white matter microvessels staining positive for DARC in human multiple sclerosis brains as compared to control tissue. Using an in vitro blood-brain barrier model we demonstrated that endothelial DARC mediates the abluminal to luminal transport of inflammatory chemokines across the blood-brain barrier. An involvement of DARC in experimental autoimmune encephalomyelitis pathogenesis was confirmed by the observed ameliorated experimental autoimmune encephalomyelitis in Darc(-/-) C57BL/6 and SJL mice, as compared to wild-type control littermates. Experimental autoimmune encephalomyelitis studies in bone marrow chimeric Darc(-/-) and wild-type mice revealed that increased plasma levels of inflammatory chemokines in experimental autoimmune encephalomyelitis depended on the presence of erythrocyte DARC. However, fully developed experimental autoimmune encephalomyelitis required the expression of endothelial DARC. Taken together, our data show a role for erythrocyte DARC as a chemokine reservoir and that endothelial DARC contributes to the pathogenesis of experimental autoimmune encephalomyelitis by shuttling chemokines across the blood-brain barrier.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Female
- Humans
- Male
- Mice
- Middle Aged
- Antigens, CD/metabolism
- Blood-Brain Barrier/metabolism
- Blood-Brain Barrier/physiopathology
- Capillary Permeability/genetics
- Central Nervous System/immunology
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Cerebellum/metabolism
- Chemokines/genetics
- Chemokines/metabolism
- Disease Models, Animal
- Duffy Blood-Group System/metabolism
- Encephalomyelitis, Autoimmune, Experimental/blood
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- In Vitro Techniques
- Mice, Inbred C57BL
- Mice, Knockout
- Multiple Sclerosis/pathology
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/metabolism
- Up-Regulation/genetics
Collapse
Affiliation(s)
- Carsten Minten
- 1 Theodor Kocher Institute, University of Bern, CH-3012 Bern, Switzerland
| | - Carsten Alt
- 1 Theodor Kocher Institute, University of Bern, CH-3012 Bern, Switzerland
| | - Melanie Gentner
- 2 Neurobiology, Department of Biomedicine, University Hospital Basel, University Basel, Switzerland
| | - Elisabeth Frei
- 1 Theodor Kocher Institute, University of Bern, CH-3012 Bern, Switzerland
| | - Urban Deutsch
- 1 Theodor Kocher Institute, University of Bern, CH-3012 Bern, Switzerland
| | - Ruth Lyck
- 1 Theodor Kocher Institute, University of Bern, CH-3012 Bern, Switzerland
| | - Nicole Schaeren-Wiemers
- 2 Neurobiology, Department of Biomedicine, University Hospital Basel, University Basel, Switzerland
| | - Antal Rot
- 3 MRC Centre for Immune Regulation, School of Immunity and Infection, University of Birmingham, UK
| | - Britta Engelhardt
- 1 Theodor Kocher Institute, University of Bern, CH-3012 Bern, Switzerland
| |
Collapse
|
44
|
Batchelor JD, Malpede BM, Omattage NS, DeKoster GT, Henzler-Wildman KA, Tolia NH. Red blood cell invasion by Plasmodium vivax: structural basis for DBP engagement of DARC. PLoS Pathog 2014; 10:e1003869. [PMID: 24415938 PMCID: PMC3887093 DOI: 10.1371/journal.ppat.1003869] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 11/22/2013] [Indexed: 01/08/2023] Open
Abstract
Plasmodium parasites use specialized ligands which bind to red blood cell (RBC) receptors during invasion. Defining the mechanism of receptor recognition is essential for the design of interventions against malaria. Here, we present the structural basis for Duffy antigen (DARC) engagement by P. vivax Duffy binding protein (DBP). We used NMR to map the core region of the DARC ectodomain contacted by the receptor binding domain of DBP (DBP-RII) and solved two distinct crystal structures of DBP-RII bound to this core region of DARC. Isothermal titration calorimetry studies show these structures are part of a multi-step binding pathway, and individual point mutations of residues contacting DARC result in a complete loss of RBC binding by DBP-RII. Two DBP-RII molecules sandwich either one or two DARC ectodomains, creating distinct heterotrimeric and heterotetrameric architectures. The DARC N-terminus forms an amphipathic helix upon DBP-RII binding. The studies reveal a receptor binding pocket in DBP and critical contacts in DARC, reveal novel targets for intervention, and suggest that targeting the critical DARC binding sites will lead to potent disruption of RBC engagement as complex assembly is dependent on DARC binding. These results allow for models to examine inter-species infection barriers, Plasmodium immune evasion mechanisms, P. knowlesi receptor-ligand specificity, and mechanisms of naturally acquired P. vivax immunity. The step-wise binding model identifies a possible mechanism by which signaling pathways could be activated during invasion. It is anticipated that the structural basis of DBP host-cell engagement will enable development of rational therapeutics targeting this interaction.
Collapse
Affiliation(s)
- Joseph D. Batchelor
- Department of Molecular Microbiology and Microbial Pathogenesis, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Brian M. Malpede
- Department of Molecular Microbiology and Microbial Pathogenesis, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Natalie S. Omattage
- Department of Molecular Microbiology and Microbial Pathogenesis, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Gregory T. DeKoster
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Katherine A. Henzler-Wildman
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Niraj H. Tolia
- Department of Molecular Microbiology and Microbial Pathogenesis, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| |
Collapse
|
45
|
Abstract
Chemokines have fundamental roles in regulating immune and inflammatory responses, primarily through their control of leukocyte migration and localization. The biological functions of chemokines are typically mediated by signalling through G protein-coupled chemokine receptors, but chemokines are also bound by a small family of atypical chemokine receptors (ACKRs), the members of which are unified by their inability to initiate classical signalling pathways after ligand binding. These ACKRs are emerging as crucial regulatory components of chemokine networks in a wide range of developmental, physiological and pathological contexts. In this Review, we discuss the biochemical and immunological properties of ACKRs and the potential unifying themes in this family, and we highlight recent studies that identify novel roles for these molecules in development , homeostasis, inflammatory disease, infection and cancer.
Collapse
|
46
|
Bachelerie F, Ben-Baruch A, Burkhardt AM, Combadiere C, Farber JM, Graham GJ, Horuk R, Sparre-Ulrich AH, Locati M, Luster AD, Mantovani A, Matsushima K, Murphy PM, Nibbs R, Nomiyama H, Power CA, Proudfoot AEI, Rosenkilde MM, Rot A, Sozzani S, Thelen M, Yoshie O, Zlotnik A. International Union of Basic and Clinical Pharmacology. [corrected]. LXXXIX. Update on the extended family of chemokine receptors and introducing a new nomenclature for atypical chemokine receptors. Pharmacol Rev 2013; 66:1-79. [PMID: 24218476 DOI: 10.1124/pr.113.007724] [Citation(s) in RCA: 678] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Sixteen years ago, the Nomenclature Committee of the International Union of Pharmacology approved a system for naming human seven-transmembrane (7TM) G protein-coupled chemokine receptors, the large family of leukocyte chemoattractant receptors that regulates immune system development and function, in large part by mediating leukocyte trafficking. This was announced in Pharmacological Reviews in a major overview of the first decade of research in this field [Murphy PM, Baggiolini M, Charo IF, Hébert CA, Horuk R, Matsushima K, Miller LH, Oppenheim JJ, and Power CA (2000) Pharmacol Rev 52:145-176]. Since then, several new receptors have been discovered, and major advances have been made for the others in many areas, including structural biology, signal transduction mechanisms, biology, and pharmacology. New and diverse roles have been identified in infection, immunity, inflammation, development, cancer, and other areas. The first two drugs acting at chemokine receptors have been approved by the U.S. Food and Drug Administration (FDA), maraviroc targeting CCR5 in human immunodeficiency virus (HIV)/AIDS, and plerixafor targeting CXCR4 for stem cell mobilization for transplantation in cancer, and other candidates are now undergoing pivotal clinical trials for diverse disease indications. In addition, a subfamily of atypical chemokine receptors has emerged that may signal through arrestins instead of G proteins to act as chemokine scavengers, and many microbial and invertebrate G protein-coupled chemokine receptors and soluble chemokine-binding proteins have been described. Here, we review this extended family of chemokine receptors and chemokine-binding proteins at the basic, translational, and clinical levels, including an update on drug development. We also introduce a new nomenclature for atypical chemokine receptors with the stem ACKR (atypical chemokine receptor) approved by the Nomenclature Committee of the International Union of Pharmacology and the Human Genome Nomenclature Committee.
Collapse
Affiliation(s)
- Francoise Bachelerie
- Chair, Subcommittee on Chemokine Receptors, Nomenclature Committee-International Union of Pharmacology, Bldg. 10, Room 11N113, NIH, Bethesda, MD 20892.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
The role of red blood cells in enhancing or preventing HIV infection and other diseases. BIOMED RESEARCH INTERNATIONAL 2013; 2013:758682. [PMID: 24224178 PMCID: PMC3810323 DOI: 10.1155/2013/758682] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 09/13/2013] [Indexed: 01/30/2023]
Abstract
Aim. To highlight the apparently neglected role of erythrocyte antigens in the epidemiology of infectious diseases, especially HIV, with the prime objective of stimulating research in this area. Method. A literature search was performed on the PubMed for relevant papers from 1984 to 2013, the era covering active HIV research. This was achieved by using the phrases “erythrocyte blood groups HIV” (81 papers) or “red cell antigen, blood groups, and HIV” (60 papers). A manual Google Scholar search was done and supplemented by original papers referenced by various authors. However, the review was limited by the relative scarcity of papers on the subject, and only papers written in English were reviewed during the period October 2012 to September 2013. Results. Many communicable and noncommunicable diseases are associated with specific blood groups. Examples of these diseases are discussed in detail. HIV has been shown to bind to erythrocytes, and candidate erythrocyte-binding molecules and mechanisms are also discussed. Moreover, erythrocyte-HIV binding is associated with increased viral infectivity, thus, underscoring the need to study this phenomenon and its implications for HIV epidemiology. Conclusion. Erythrocyte antigens may be important in the pathogenesis and epidemiology of many diseases, including HIV.
Collapse
|
48
|
Ortega-Gómez A, Perretti M, Soehnlein O. Resolution of inflammation: an integrated view. EMBO Mol Med 2013; 5:661-74. [PMID: 23592557 PMCID: PMC3662311 DOI: 10.1002/emmm.201202382] [Citation(s) in RCA: 512] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/22/2013] [Accepted: 03/01/2013] [Indexed: 12/16/2022] Open
Abstract
Resolution of inflammation is a coordinated and active process aimed at restoration of tissue integrity and function. This review integrates the key molecular and cellular mechanisms of resolution. We describe how abrogation of chemokine signalling blocks continued neutrophil tissue infiltration and how apoptotic neutrophils attract monocytes and macrophages to induce their clearance. Uptake of apoptotic neutrophils by macrophages reprograms macrophages towards a resolving phenotype, a key event to restore tissue homeostasis. Finally, we highlight the therapeutic potential that derives from understanding the mechanisms of resolution.
Collapse
Affiliation(s)
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of MedicineLondon, UK
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention, LMUMunich, Germany
- Department of Pathology, AMCAmsterdam, The Netherlands
| |
Collapse
|
49
|
Graham GJ, Locati M. Regulation of the immune and inflammatory responses by the 'atypical' chemokine receptor D6. J Pathol 2013; 229:168-75. [PMID: 23125030 DOI: 10.1002/path.4123] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 10/03/2012] [Accepted: 10/03/2012] [Indexed: 01/19/2023]
Abstract
Chemokines and their receptors are key regulators of leukocyte migration and intra-tissue accumulation under both homeostatic and inflammatory conditions. Regulation of chemokine-dependent responses, particularly those relating to inflammation, is essential to avoid the development of inflammatory and autoimmune pathologies. Recently, a new subfamily of chemokine receptors referred to as the 'atypical' chemokine receptors has emerged, members of which have been shown to play important roles in controlling in vivo chemokine biology. Here we review the basic biology of the chemokine and chemokine receptor family, introduce the topic of 'atypical' chemokine receptor biology and focus specifically on the best-characterized of the 'atypical' chemokine receptors, D6. D6 is a 'scavenging' receptor for inflammatory CC chemokines and plays a central role in the resolution of in vivo inflammatory responses. We describe the biology, biochemistry and pathological relevance of D6 and outline emerging data suggesting that it has additional important roles in integrating innate and adaptive immune responses.
Collapse
Affiliation(s)
- Gerard J Graham
- Institute of Infection, Immunity and Inflammation, University of Glasgow, UK.
| | | |
Collapse
|
50
|
Grodecka M, Bertrand O, Karolak E, Lisowski M, Waśniowska K. One-step immunopurification and lectinochemical characterization of the Duffy atypical chemokine receptor from human erythrocytes. Glycoconj J 2012; 29:93-105. [PMID: 22246380 PMCID: PMC3311851 DOI: 10.1007/s10719-011-9367-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 12/07/2011] [Accepted: 12/18/2011] [Indexed: 12/12/2022]
Abstract
Duffy antigen/receptor for chemokines (DARC) is a glycosylated seven-transmembrane protein acting as a blood group antigen, a chemokine binding protein and a receptor for Plasmodium vivax malaria parasite. It is present on erythrocytes and endothelial cells of postcapillary venules. The N-terminal extracellular domain of the Duffy glycoprotein carries Fy(a)/Fy(b) blood group antigens and Fy6 linear epitope recognized by monoclonal antibodies. Previously, we have shown that recombinant Duffy protein expressed in K562 cells has three N-linked oligosaccharide chains, which are mainly of complex-type. Here we report a one-step purification method of Duffy protein from human erythrocytes. DARC was extracted from erythrocyte membranes in the presence of 1% n-dodecyl-β-D-maltoside (DDM) and 0.05% cholesteryl hemisuccinate (CHS) and purified by affinity chromatography using immobilized anti-Fy6 2C3 mouse monoclonal antibody. Duffy glycoprotein was eluted from the column with synthetic DFEDVWN peptide containing epitope for 2C3 monoclonal antibody. In this single-step immunoaffinity purification method we obtained highly purified DARC, which migrates in SDS-polyacrylamide gel as a major diffuse band corresponding to a molecular mass of 40-47 kDa. In ELISA purified Duffy glycoprotein binds anti-Duffy antibodies recognizing epitopes located on distinct regions of the molecule. Results of circular dichroism measurement indicate that purified DARC has a high content of α-helical secondary structure typical for chemokine receptors. Analysis of DARC glycans performed by means of lectin blotting and glycosidase digestion suggests that native Duffy N-glycans are mostly triantennary complex-type, terminated with α2-3- and α2-6-linked sialic acid residues with bisecting GlcNAc and α1-6-linked fucose at the core.
Collapse
Affiliation(s)
- Magdalena Grodecka
- Department of Immunochemistry, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wrocław, Poland
| | - Olivier Bertrand
- Institut National de la Santé et de la Recherche Médicale, UMR_S 665, F-75015 Paris, France
- Institut National de la Transfusion Sanguine, F-75015 Paris, France
| | - Ewa Karolak
- Department of Immunochemistry, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wrocław, Poland
| | - Marek Lisowski
- Faculty of Chemistry, University of Wrocław, F. Joliot-Curie 14, 50-383 Wrocław, Poland
| | - Kazimiera Waśniowska
- Department of Immunochemistry, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wrocław, Poland
- Faculty of Physical Education and Physiotherapy, Opole University of Technology, Prószkowska 76, 45-758 Opole, Poland
| |
Collapse
|