1
|
Vasquez MT, Lubkin A, Reyes-Robles T, Day CJ, Lacey KA, Jennings MP, Torres VJ. Identification of a domain critical for Staphylococcus aureus LukED receptor targeting and lysis of erythrocytes. J Biol Chem 2020; 295:17241-17250. [PMID: 33051210 PMCID: PMC7863875 DOI: 10.1074/jbc.ra120.015757] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Leukocidin ED (LukED) is a pore-forming toxin produced by Staphylococcus aureus, which lyses host cells and promotes virulence of the bacteria. LukED enables S. aureus to acquire iron by lysing erythrocytes, which depends on targeting the host receptor Duffy antigen receptor for chemokines (DARC). The toxin also targets DARC on the endothelium, contributing to the lethality observed during bloodstream infection in mice. LukED is comprised of two monomers: LukE and LukD. LukE binds to DARC and facilitates hemolysis, but the closely related Panton-Valentine leukocidin S (LukS-PV) does not bind to DARC and is not hemolytic. The interaction of LukE with DARC and the role this plays in hemolysis are incompletely characterized. To determine the domain(s) of LukE that are critical for DARC binding, we studied the hemolytic function of LukE-LukS-PV chimeras, in which areas of sequence divergence (divergence regions, or DRs) were swapped between the toxins. We found that two regions of LukE's rim domain contribute to hemolysis, namely residues 57-75 (DR1) and residues 182-196 (DR4). Interestingly, LukE DR1 is sufficient to render LukS-PV capable of DARC binding and hemolysis. Further, LukE, by binding DARC through DR1, promotes the recruitment of LukD to erythrocytes, likely by facilitating LukED oligomer formation. Finally, we show that LukE targets murine Darc through DR1 in vivo to cause host lethality. These findings expand our biochemical understanding of the LukE-DARC interaction and the role that this toxin-receptor pair plays in S. aureus pathophysiology.
Collapse
Affiliation(s)
- Marilyn T Vasquez
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Ashira Lubkin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Tamara Reyes-Robles
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Christopher J Day
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Keenan A Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Michael P Jennings
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA.
| |
Collapse
|
2
|
Metzemaekers M, Gouwy M, Proost P. Neutrophil chemoattractant receptors in health and disease: double-edged swords. Cell Mol Immunol 2020; 17:433-450. [PMID: 32238918 PMCID: PMC7192912 DOI: 10.1038/s41423-020-0412-0] [Citation(s) in RCA: 238] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/05/2020] [Indexed: 02/08/2023] Open
Abstract
Neutrophils are frontline cells of the innate immune system. These effector leukocytes are equipped with intriguing antimicrobial machinery and consequently display high cytotoxic potential. Accurate neutrophil recruitment is essential to combat microbes and to restore homeostasis, for inflammation modulation and resolution, wound healing and tissue repair. After fulfilling the appropriate effector functions, however, dampening neutrophil activation and infiltration is crucial to prevent damage to the host. In humans, chemoattractant molecules can be categorized into four biochemical families, i.e., chemotactic lipids, formyl peptides, complement anaphylatoxins and chemokines. They are critically involved in the tight regulation of neutrophil bone marrow storage and egress and in spatial and temporal neutrophil trafficking between organs. Chemoattractants function by activating dedicated heptahelical G protein-coupled receptors (GPCRs). In addition, emerging evidence suggests an important role for atypical chemoattractant receptors (ACKRs) that do not couple to G proteins in fine-tuning neutrophil migratory and functional responses. The expression levels of chemoattractant receptors are dependent on the level of neutrophil maturation and state of activation, with a pivotal modulatory role for the (inflammatory) environment. Here, we provide an overview of chemoattractant receptors expressed by neutrophils in health and disease. Depending on the (patho)physiological context, specific chemoattractant receptors may be up- or downregulated on distinct neutrophil subsets with beneficial or detrimental consequences, thus opening new windows for the identification of disease biomarkers and potential drug targets.
Collapse
Affiliation(s)
- Mieke Metzemaekers
- Laboratory of Molecular Immunology, Rega Institute, KU Leuven, Herestraat 49 bus 1042, B-3000, Leuven, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Rega Institute, KU Leuven, Herestraat 49 bus 1042, B-3000, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Rega Institute, KU Leuven, Herestraat 49 bus 1042, B-3000, Leuven, Belgium.
| |
Collapse
|
3
|
Crijns H, Vanheule V, Proost P. Targeting Chemokine-Glycosaminoglycan Interactions to Inhibit Inflammation. Front Immunol 2020; 11:483. [PMID: 32296423 PMCID: PMC7138053 DOI: 10.3389/fimmu.2020.00483] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/02/2020] [Indexed: 12/12/2022] Open
Abstract
Leukocyte migration into tissues depends on the activity of chemokines that form concentration gradients to guide leukocytes to a specific site. Interaction of chemokines with their specific G protein-coupled receptors (GPCRs) on leukocytes induces leukocyte adhesion to the endothelial cells, followed by extravasation of the leukocytes and subsequent directed migration along the chemotactic gradient. Interaction of chemokines with glycosaminoglycans (GAGs) is crucial for extravasation in vivo. Chemokines need to interact with GAGs on endothelial cells and in the extracellular matrix in tissues in order to be presented on the endothelium of blood vessels and to create a concentration gradient. Local chemokine retention establishes a chemokine gradient and prevents diffusion and degradation. During the last two decades, research aiming at reducing chemokine activity mainly focused on the identification of inhibitors of the interaction between chemokines and their cognate GPCRs. This approach only resulted in limited success. However, an alternative strategy, targeting chemokine-GAG interactions, may be a promising approach to inhibit chemokine activity and inflammation. On this line, proteins derived from viruses and parasites that bind chemokines or GAGs may have the potential to interfere with chemokine-GAG interactions. Alternatively, chemokine mimetics, including truncated chemokines and mutant chemokines, can compete with chemokines for binding to GAGs. Such truncated or mutated chemokines are characterized by a strong binding affinity for GAGs and abrogated binding to their chemokine receptors. Finally, Spiegelmers that mask the GAG-binding site on chemokines, thereby preventing chemokine-GAG interactions, were developed. In this review, the importance of GAGs for chemokine activity in vivo and strategies that could be employed to target chemokine-GAG interactions will be discussed in the context of inflammation.
Collapse
Affiliation(s)
- Helena Crijns
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Vincent Vanheule
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
4
|
Miyabe Y, Miyabe C, Mani V, Mempel TR, Luster AD. Atypical complement receptor C5aR2 transports C5a to initiate neutrophil adhesion and inflammation. Sci Immunol 2019; 4:eaav5951. [PMID: 31076525 DOI: 10.1126/sciimmunol.aav5951] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/29/2019] [Indexed: 12/14/2022]
Abstract
Chemoattractant-induced arrest of circulating leukocytes and their subsequent diapedesis is a fundamental component of inflammation. However, how tissue-derived chemoattractants are transported into the blood vessel lumen to induce leukocyte entry into tissue is not well understood. Here, intravital microscopy in live mice has shown that the "atypical" complement C5a receptor 2 (C5aR2) and the atypical chemokine receptor 1 (ACKR1) expressed on endothelial cells were required for the transport of C5a and CXCR2 chemokine ligands, respectively, into the vessel lumen in a murine model of immune complex-induced arthritis. Transported C5a was required to initiate C5aR1-mediated neutrophil arrest, whereas transported chemokines were required to initiate CXCR2-dependent neutrophil transdendothelial migration. These findings provide new insights into how atypical chemoattractant receptors collaborate with "classical" signaling chemoattractant receptors to control distinct steps in the recruitment of neutrophils into tissue sites of inflammation.
Collapse
Affiliation(s)
- Yoshishige Miyabe
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Chie Miyabe
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vinidhra Mani
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Lubkin A, Lee WL, Alonzo F, Wang C, Aligo J, Keller M, Girgis NM, Reyes-Robles T, Chan R, O'Malley A, Buckley P, Vozhilla N, Vasquez MT, Su J, Sugiyama M, Yeung ST, Coffre M, Bajwa S, Chen E, Martin P, Kim SY, Loomis C, Worthen GS, Shopsin B, Khanna KM, Weinstock D, Lynch AS, Koralov SB, Loke P, Cadwell K, Torres VJ. Staphylococcus aureus Leukocidins Target Endothelial DARC to Cause Lethality in Mice. Cell Host Microbe 2019; 25:463-470.e9. [PMID: 30799265 DOI: 10.1016/j.chom.2019.01.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/24/2018] [Accepted: 01/23/2019] [Indexed: 01/16/2023]
Abstract
The pathogenesis of Staphylococcus aureus is thought to depend on the production of pore-forming leukocidins that kill leukocytes and lyse erythrocytes. Two leukocidins, Leukocidin ED (LukED) and γ-Hemolysin AB (HlgAB), are necessary and sufficient to kill mice upon infection and toxin challenge. We demonstrate that LukED and HlgAB cause vascular congestion and derangements in vascular fluid distribution that rapidly cause death in mice. The Duffy antigen receptor for chemokines (DARC) on endothelial cells, rather than leukocytes or erythrocytes, is the critical target for lethality. Consistent with this, LukED and HlgAB injure primary human endothelial cells in a DARC-dependent manner, and mice with DARC-deficient endothelial cells are resistant to toxin-mediated lethality. During bloodstream infection in mice, DARC targeting by S. aureus causes increased tissue damage, organ dysfunction, and host death. The potential for S. aureus leukocidins to manipulate vascular integrity highlights the importance of these virulence factors.
Collapse
Affiliation(s)
- Ashira Lubkin
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Warren L Lee
- Keenan Research Centre, St Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.
| | - Francis Alonzo
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Changsen Wang
- Keenan Research Centre, St Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Jason Aligo
- Janssen Research & Development LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Matthew Keller
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA; Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Natasha M Girgis
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Tamara Reyes-Robles
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Rita Chan
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Aidan O'Malley
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Peter Buckley
- Janssen Research & Development LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Nikollaq Vozhilla
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Marilyn T Vasquez
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Johnny Su
- Keenan Research Centre, St Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Michael Sugiyama
- Keenan Research Centre, St Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Stephen T Yeung
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Maryaline Coffre
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Sofia Bajwa
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Eric Chen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Patricia Martin
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Sang Y Kim
- Department of Pathology, New York University School of Medicine, New York, NY, USA; Office of Collaborative Sciences, NYU School of Medicine, New York, NY, USA; Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Cynthia Loomis
- Department of Pathology, New York University School of Medicine, New York, NY, USA; Office of Collaborative Sciences, NYU School of Medicine, New York, NY, USA; Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - G Scott Worthen
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, USA; Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Bo Shopsin
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA; Division of Infectious Diseases, Department of Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Kamal M Khanna
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Daniel Weinstock
- Janssen Research & Development LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Anthony Simon Lynch
- Janssen Research & Development LLC, 1400 McKean Road, Spring House, PA 19477, USA
| | - Sergei B Koralov
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - P'ng Loke
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Ken Cadwell
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA; Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
6
|
Edderkaoui B, Sargsyan L, Hetrick A, Li H. Deficiency of Duffy Antigen Receptor for Chemokines Ameliorated Cochlear Damage From Noise Exposure. Front Mol Neurosci 2018; 11:173. [PMID: 29899689 PMCID: PMC5988871 DOI: 10.3389/fnmol.2018.00173] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/08/2018] [Indexed: 01/06/2023] Open
Abstract
Cochlear inflammatory response to various environmental insults, including acoustic and ototoxic overexposures, has been increasingly become a topic of interest. As the immune response is associated with both pathology and protection, targeting specific components of the immune response is expected to dissect the relationships between cellular damage and inflammation-associated protection and repair in the cochlea. Duffy antigen receptor for chemokines (DARC) is a member of a group of atypical chemokine receptors, and essential for chemokine-regulated leukocyte/neutrophil trafficking during inflammation. Previous studies have reported that Darc deficiency alters chemokine bioavailability and leukocyte homeostasis, leading to significant anti-inflammatory effects in tissues following injury. In this study, we have used Darc knockout mice to determine the impact of a deficiency in this gene on cochlear development, as well as function in cochlea subjected to various stresses. We observed that DARC is not required for normal development of cochlear function, as evidenced by typical hearing sensitivity in juvenile Darc-KO mice, as compared to wild type (WT) C57BL/6 mice. However, Darc-KO mice exhibited improved hearing recovery after intense noise exposure when compared to wild-type. The auditory brainstem response (ABR) threshold shift between KO and WT mice was most obvious at 1-week post-noise exposure. At cochlear locations above the frequency range of the energy band of damaging noise, both hair cell survival and ribbon synapse density were improved in Darc deficient animals. In addition, the mRNA levels of some major inflammatory effectors, including Mcp-1 and Gdf15, were altered in Darc-KO mice compared to control mice at 1, 3 and 7 days post-noise exposure. These data collectively suggest that the normal Darc-dependent inflammatory response slows down the process of hearing recovery, and exacerbates cellular damage in the cochlea after noise exposure.
Collapse
Affiliation(s)
- Bouchra Edderkaoui
- Research Service, VA Loma Linda Healthcare System, Loma Linda, CA, United States.,Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Liana Sargsyan
- Research Service, VA Loma Linda Healthcare System, Loma Linda, CA, United States
| | - Alisa Hetrick
- Research Service, VA Loma Linda Healthcare System, Loma Linda, CA, United States
| | - Hongzhe Li
- Research Service, VA Loma Linda Healthcare System, Loma Linda, CA, United States.,Loma Linda University School of Medicine, Loma Linda, CA, United States.,Department of Otolaryngology-Head and Neck Surgery, Loma Linda University School of Medicine, Loma Linda, CA, United States
| |
Collapse
|
7
|
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
|
8
|
Thiriot A, Perdomo C, Cheng G, Novitzky-Basso I, McArdle S, Kishimoto JK, Barreiro O, Mazo I, Triboulet R, Ley K, Rot A, von Andrian UH. Differential DARC/ACKR1 expression distinguishes venular from non-venular endothelial cells in murine tissues. BMC Biol 2017; 15:45. [PMID: 28526034 PMCID: PMC5438556 DOI: 10.1186/s12915-017-0381-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/26/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Intravascular leukocyte recruitment in most vertebrate tissues is restricted to postcapillary and collecting venules, whereas capillaries and arterioles usually support little or no leukocyte adhesion. This segmental restriction is thought to be mediated by endothelial, rather than hemodynamic, differences. The underlying mechanisms are largely unknown, in part because effective tools to distinguish, isolate, and analyze venular endothelial cells (V-ECs) and non-venular endothelial cells (NV-ECs) have been unavailable. We hypothesized that the atypical chemokine receptor DARC (Duffy Antigen Receptor for Chemokines, a.k.a. ACKR1 or CD234) may distinguish V-ECs versus NV-ECs in mice. METHODS We generated a rat-anti-mouse monoclonal antibody (MAb) that specifically recognizes the erythroid and endothelial forms of native, surface-expressed DARC. Using this reagent, we characterized DARC expression and distribution in the microvasculature of murine tissues. RESULTS DARC was exquisitely restricted to post-capillary and small collecting venules and completely absent from arteries, arterioles, capillaries, veins, and most lymphatics in every tissue analyzed. Accordingly, intravital microscopy showed that adhesive leukocyte-endothelial interactions were restricted to DARC+ venules. DARC was detectable over the entire circumference of V-ECs, but was more concentrated at cell-cell junctions. Analysis of single-cell suspensions suggested that the frequency of V-ECs among the total microvascular EC pool varies considerably between different tissues. CONCLUSIONS Immunostaining of endothelial DARC allows the identification and isolation of intact V-ECs from multiple murine tissues. This strategy may be useful to dissect the mechanisms underlying segmental microvascular specialization in healthy and diseased tissues and to characterize the role of EC subsets in tissue-homeostasis, immune surveillance, infection, inflammation, and malignancies.
Collapse
Affiliation(s)
- Aude Thiriot
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Carolina Perdomo
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Guiying Cheng
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Igor Novitzky-Basso
- Center for Immunology and Infection, Department of Biology, University of York, YO10 5DD, Heslington, York, UK
- Present address: Blood and Marrow Transplant Unit, Queen Elizabeth University Hospital, Glasgow, UK
| | - Sara McArdle
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Jamie K Kishimoto
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Olga Barreiro
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Irina Mazo
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | | | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Antal Rot
- Center for Immunology and Infection, Department of Biology, University of York, YO10 5DD, Heslington, York, UK
| | - Ulrich H von Andrian
- Department of Microbiology and Immunobiology & HMS Center for Immune Imaging, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA.
| |
Collapse
|
9
|
Alemi F, Elgendy M, Edderkaoui B. Potential Role of DARC-Chemokine Interaction in the Recruitment of Osteoclast Precursors in Response to Bacterial Lipopolysaccharide Challenge. Calcif Tissue Int 2016; 99:481-488. [PMID: 27376530 PMCID: PMC5056128 DOI: 10.1007/s00223-016-0170-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/24/2016] [Indexed: 12/14/2022]
Abstract
Duffy antigen receptor for chemokines (DARC) binds to a number of pro-inflammatory chemokines, and since chemokines are known to regulate trafficking of osteoclast (OC) precursors, we predicted that DARC would regulate OC recruitment to sites of inflammation by modulating chemokine activity. To test this hypothesis, we evaluated the mRNA expression of Darc and the chemokines known to bind to DARC, in endothelial cells treated with bacterial lipopolysaccharide (LPS). The mRNA expression of Mcp-1, Rantes, Darc and Ccr5 was significantly increased in endothelial cells in response to LPS treatment. Blocking the function of DARC with neutralizing antibody partially abrogated the effect of LPS on the mRNA expression of Mcp-1 and Rantes. In vivo, mice with targeted disruption of Darc gene (Darc-KO) and control wild-type (WT) mice were used to assess the role of DARC in response to single LPS application on the top of parietal bones. Five hours post-LPS injection, local expression of Cd14 mRNA (a marker of inflammatory monocytes) was significantly increased in both lines of mice. However, the magnitude of increase was greater in WT mice compared with Darc-KO mice suggesting a role for DARC in mediating the recruitment of monocytes in response to LPS. Histological staining for tartrate-resistant acid phosphatase (TRAP) in calvaria sections taken from the injection sites revealed a significant reduction in TRAP-labeled surface per bone surface in response to LPS in Darc-KO mice compared with WT mice. Based on these findings, we concluded that DARC regulates recruitment of OC precursors at the inflammation site, probably through regulation of chemokines transcytosis across endothelial cell barrier.
Collapse
Affiliation(s)
- Fatemeh Alemi
- Musculoskeletal Disease Center, Research Service, Jerry L Pettis Memorial Veterans Administration Medical Center, 11201 Benton St. (151), Loma Linda, CA, 92357, USA
| | - Mohamed Elgendy
- Musculoskeletal Disease Center, Research Service, Jerry L Pettis Memorial Veterans Administration Medical Center, 11201 Benton St. (151), Loma Linda, CA, 92357, USA
| | - Bouchra Edderkaoui
- Musculoskeletal Disease Center, Research Service, Jerry L Pettis Memorial Veterans Administration Medical Center, 11201 Benton St. (151), Loma Linda, CA, 92357, USA.
- School of Medicine, Loma Linda University, Loma Linda, CA, USA.
| |
Collapse
|
10
|
Wan W, Liu Q, Lionakis MS, Marino APMP, Anderson SA, Swamydas M, Murphy PM. Atypical chemokine receptor 1 deficiency reduces atherogenesis in ApoE-knockout mice. Cardiovasc Res 2015; 106:478-87. [PMID: 25858253 PMCID: PMC4447808 DOI: 10.1093/cvr/cvv124] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 03/18/2015] [Accepted: 03/20/2015] [Indexed: 12/14/2022] Open
Abstract
AIMS Atypical chemokine receptor 1 (Ackr1; previously known as the Duffy antigen receptor for chemokines or Darc) is thought to regulate acute inflammatory responses in part by scavenging inflammatory CC and CXC chemokines; however, evidence for a role in chronic inflammation has been lacking. Here we investigated the role of Ackr1 in chronic inflammation, in particular in the setting of atherogenesis, using the apolipoprotein E-deficient (ApoE(-/-)) mouse model. METHODS AND RESULTS Ackr1(-/-)ApoE(-/-) and Ackr1(+/+)ApoE(-/-) littermates were obtained by crossing ApoE(-/-) mice and Ackr1(-/-) mice on a C57BL/6J background. Ackr1 (+/+)ApoE(-/-)mice fed a Western diet up-regulated Ackr1 expression in the aorta and had markedly increased atherosclerotic lesion size compared with Ackr1(-/-)ApoE(-/-) mice. This difference was observed in both the whole aorta and the aortic root in both early and late stages of the model. Ackr1 deficiency did not affect serum cholesterol levels or macrophage, collagen or smooth muscle cell content in atherosclerotic plaques, but significantly reduced the expression of Ccl2 and Cxcl1 in the whole aorta of ApoE(-/-) mice. In addition, Ackr1 deficiency resulted in a modest decrease in T cell subset frequency and inflammatory mononuclear phagocyte content in aorta and blood in the model. CONCLUSIONS Ackr1 deficiency appears to be protective in the ApoE knockout model of atherogenesis, but it is associated with only modest changes in cytokine and chemokine expression as well as T-cell subset frequency and inflammatory macrophage content.
Collapse
Affiliation(s)
- Wuzhou Wan
- Molecular Signaling Section, Laboratory of Molecular Immunology (LMI), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Qian Liu
- Molecular Signaling Section, Laboratory of Molecular Immunology (LMI), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Michail S Lionakis
- Fungal Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Ana Paula M P Marino
- Molecular Signaling Section, Laboratory of Molecular Immunology (LMI), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Stasia A Anderson
- National Heart, Lung and Blood Institute (NHLBI) Animal MRI Core, NIH, Bethesda, MD, USA
| | - Muthulekha Swamydas
- Fungal Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Philip M Murphy
- Molecular Signaling Section, Laboratory of Molecular Immunology (LMI), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
11
|
Abstract
International Society of Blood Transfusion has recently recognized 33 blood group systems. Apart from ABO and Rhesus system, many other types of antigens have been noticed on the red cell membranes. Blood grouping and cross-matching is one of the few important tests that the anaesthesiologist orders during perioperative period. Hence, a proper understanding of the blood group system, their clinical significance, typing and cross-matching tests, and current perspective are of paramount importance to prevent transfusion-related complications. Nonetheless, the knowledge on blood group system is necessary to approach blood group-linked diseases which are still at the stage of research. This review addresses all these aspects of the blood groups system.
Collapse
Affiliation(s)
- Ranadhir Mitra
- Department of Neuroanaesthesiology, All India Institute of Medical Sciences, New Delhi, India
| | - Nitasha Mishra
- Department of Neuroanaesthesiology, All India Institute of Medical Sciences, New Delhi, India
| | - Girija Prasad Rath
- Department of Neuroanaesthesiology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
12
|
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: 1.2] [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
|
13
|
Kuravi SJ, McGettrick HM, Satchell SC, Saleem MA, Harper L, Williams JM, Rainger GE, Savage COS. Podocytes regulate neutrophil recruitment by glomerular endothelial cells via IL-6-mediated crosstalk. THE JOURNAL OF IMMUNOLOGY 2014; 193:234-43. [PMID: 24872191 PMCID: PMC4067868 DOI: 10.4049/jimmunol.1300229] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stromal cells actively modulate the inflammatory process, in part by influencing the ability of neighboring endothelial cells to support the recruitment of circulating leukocytes. We hypothesized that podocytes influence the ability of glomerular endothelial cells (GEnCs) to recruit neutrophils during inflammation. To address this, human podocytes and human GEnCs were cultured on opposite sides of porous inserts and then treated with or without increasing concentrations of TNF-α prior to addition of neutrophils. The presence of podocytes significantly reduced neutrophil recruitment to GEnCs by up to 50% when cultures were treated with high-dose TNF-α (100 U/ml), when compared with GEnC monocultures. Importantly, this phenomenon was dependent on paracrine actions of soluble IL-6, predominantly released by podocytes. A similar response was absent when HUVECs were cocultured with podocytes, indicating a tissue-specific phenomenon. Suppressor of cytokine signaling 3 elicited the immunosuppressive actions of IL-6 in a process that disrupted the presentation of chemokines on GEnCs by altering the expression of the duffy Ag receptor for chemokines. Interestingly, suppressor of cytokine signaling 3 knockdown in GEnCs upregulated duffy Ag receptor for chemokines and CXCL5 expression, thereby restoring the neutrophil recruitment. In summary, these studies reveal that podocytes can negatively regulate neutrophil recruitment to inflamed GEnCs by modulating IL-6 signaling, identifying a potential novel anti-inflammatory role of IL-6 in renal glomeruli.
Collapse
Affiliation(s)
- Sahithi J Kuravi
- Centre for Translational Inflammation Research, School of Immunity and Infection, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| | - Helen M McGettrick
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Simon C Satchell
- Academic Renal Unit, Southmead Hospital, Bristol BS10 5NB, United Kingdom
| | - Moin A Saleem
- Academic Renal Unit, Southmead Hospital, Bristol BS10 5NB, United Kingdom
| | - Lorraine Harper
- Centre for Translational Inflammation Research, School of Immunity and Infection, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Julie M Williams
- Wellcome Trust Clinical Research Facility, University Hospital Birmingham Foundation Trust, Birmingham B15 2TH, United Kingdom; and
| | - George Ed Rainger
- Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Caroline O S Savage
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
14
|
Manicone AM. Role of the pulmonary epithelium and inflammatory signals in acute lung injury. Expert Rev Clin Immunol 2014. [DOI: 10.1586/1744666x.5.1.63] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
15
|
Yamakawa RH, Saito PK, da Silva Junior WV, de Mattos LC, Borelli SD. Polymorphism of leukocyte and erythrocyte antigens in chronic kidney disease patients in southern Brazil. PLoS One 2014; 9:e84456. [PMID: 24409300 PMCID: PMC3883691 DOI: 10.1371/journal.pone.0084456] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 11/22/2013] [Indexed: 12/02/2022] Open
Abstract
We investigated the polymorphism of human leukocyte antigens (HLA) and Duffy erythrocyte antigens in chronic kidney disease (CKD) patients in southern Brazil. One hundred and eighty-three CKD patients, over 18 years old, on hemodialysis, were included. HLA-A, -B and -DRB1 typing was performed using the LABType®SSO (One Lambda, Inc.). Duffy phenotypes were determined by gel column agglutination using anti-Fya and anti-Fyb monoclonal anti-sera. The patients' predominant ages ranged between 51 and 70 years (43%) and the predominant gender, ethnic group and dialysis period were, respectively, male (62%), white (62%) and 1–3 years (40%). The highest and lowest frequencies of Duffy phenotypes were Fy(a+b+) and Fy(a−b−), respectively. Nineteen HLA-A, 30 HLA-B and 13 HLA-DRB1 allele groups were identified. The most frequent HLA allele groups were HLA-A*01, -A*02, -A*03, -A*11, -A*24; HLA-B*07, -B*15, -B*35, -B*44, -B*51; HLA-DRB1*03, -DRB1*04, -DRB1*07, -DRB1*11 and -DRB1*13. Statistically significant differences were observed in the Duffy and HLA polymorphisms compared between CKD patients and healthy subjects. The Fy(a+b−) phenotype (p<0.0001, OR = 2.56, 95% CI = 1.60–4.07) was the most frequent in the patients (p<0.05), and the Fy(a+b+) phenotype (p = 0.0039, OR = 1.71, 95% CI = 1.18–2.51) was the most frequent in the healthy subjects in the same region of Paraná state (p<0.05). Regarding HLA, the HLA-B*42, -B*45, -B*51 and -DRB1*03 allele groups were the most frequent in the patients (p<0.05), and the HLA-B*44 allele group was the most frequent in the healthy subjects in the same region of Brazil (p<0.05). The polymorphism of these two markers among CKD patients in southern Brazil and healthy subjects of other studies, suggests that these markers might be involved with CKD development. Further studies should be undertaken to analyze the markers' influence on CKD and the long-term results from kidney transplantation.
Collapse
Affiliation(s)
- Roger Haruki Yamakawa
- Departamento de Ciências Básicas da Saúde, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | - Patricia Keiko Saito
- Departamento de Ciências Básicas da Saúde, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| | | | - Luiz Carlos de Mattos
- Departamento de Biologia Molecular, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
| | - Sueli Donizete Borelli
- Departamento de Ciências Básicas da Saúde, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
- * E-mail:
| |
Collapse
|
16
|
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
|
17
|
Rundle CH, Mohan S, Edderkaoui B. Duffy antigen receptor for chemokines regulates post-fracture inflammation. PLoS One 2013; 8:e77362. [PMID: 24146983 PMCID: PMC3798395 DOI: 10.1371/journal.pone.0077362] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 09/02/2013] [Indexed: 12/14/2022] Open
Abstract
There is now considerable experimental data to suggest that inflammatory cells collaborate in the healing of skeletal fractures. In terms of mechanisms that contribute to the recruitment of inflammatory cells to the fracture site, chemokines and their receptors have received considerable attention. Our previous findings have shown that Duffy antigen receptor for chemokines (Darc), the non-classical chemokine receptor that does not signal, but rather acts as a scavenger of chemokines that regulate cell migration, is a negative regulator of peak bone density in mice. Furthermore, because Darc is expressed by inflammatory and endothelial cells, we hypothesized that disruption of Darc action will affect post-fracture inflammation and consequently will affect fracture healing. To test this hypothesis, we evaluated fracture healing in mice with targeted disruption of Darc and corresponding wild type (WT) control mice. We found that fracture callus cartilage formation was significantly greater (33%) at 7 days post-surgery in Darc-KO compared to WT mice. The increased cartilage was associated with greater Collagen (Col) II expression at 3 days post-fracture and Col-X at 7 days post-fracture compared to WT mice, suggesting that Darc deficiency led to early fracture cartilage formation and differentiation. We then compared the expression of cytokine and chemokine genes known to be induced during inflammation. Interleukin (Il)-1β, Il-6, and monocyte chemotactic protein 1 were all down regulated in the fractures derived from Darc-KO mice at one day post-fracture, consistent with an altered inflammatory response. Furthermore, the number of macrophages was significantly reduced around the fractures in Darc-KO compared to WT mice. Based on these data, we concluded that Darc plays a role in modulating the early inflammatory response to bone fracture and subsequent cartilage formation. However, the early cartilage formation was not translated with an early bone formation at the fracture site in Darc-KO compared to WT mice.
Collapse
Affiliation(s)
- Charles H. Rundle
- Musculoskeletal Disease Center, Research Service, Jerry L Pettis Memorial Veterans Administration Medical Center, Loma Linda, California, United States of America
- Department of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Research Service, Jerry L Pettis Memorial Veterans Administration Medical Center, Loma Linda, California, United States of America
- Department of Medicine, Loma Linda University, Loma Linda, California, United States of America
- Department of Biochemistry, Loma Linda University, Loma Linda, California, United States of America
- Department of Physiology, Loma Linda University, Loma Linda, California, United States of America
| | - Bouchra Edderkaoui
- Musculoskeletal Disease Center, Research Service, Jerry L Pettis Memorial Veterans Administration Medical Center, Loma Linda, California, United States of America
- Department of Medicine, Loma Linda University, Loma Linda, California, United States of America
| |
Collapse
|
18
|
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: 497] [Impact Index Per Article: 45.2] [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
|
19
|
Wantha S, Alard JE, Megens RTA, van der Does AM, Döring Y, Drechsler M, Pham CTN, Wang MW, Wang JM, Gallo RL, von Hundelshausen P, Lindbom L, Hackeng T, Weber C, Soehnlein O. Neutrophil-derived cathelicidin promotes adhesion of classical monocytes. Circ Res 2013; 112:792-801. [PMID: 23283724 DOI: 10.1161/circresaha.112.300666] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RATIONALE The leukocyte response in acute inflammation is characterized by an initial recruitment of neutrophils preceding a second wave of monocytes. Neutrophil-derived granule proteins were suggested to hold an important role in this cellular switch. The exact mechanisms by which neutrophils mediate these processes are only partially understood. OBJECTIVE To investigate the role of neutrophils and their granule contents in the adhesion of monocyte subpopulations in acute inflammation. METHODS AND RESULTS Here, we show that neutrophil-derived cathelicidins (human: LL37, mouse: CRAMP) induce adhesion of classical monocytes but not of nonclassical monocytes in the mouse cremaster muscle and in in vitro flow chamber assays. CRAMP is released from emigrated neutrophils and then transported across the endothelium, where it is presented to rolling leukocytes. Endothelial-bound cathelicidin activates formyl-peptide receptor 2 on classical monocytes, resulting in monocytic β1- and β2-integrin conformational change toward an extended, active conformation that allows for adhesion to their respective ligands, vascular cell adhesion molecule 1 and intercellular adhesion molecule 1. CONCLUSIONS These data elucidate a novel mechanism of neutrophil-mediated monocyte recruitment, which could be targeted in conditions where recruitment of classical monocytes plays an unfavorable role.
Collapse
Affiliation(s)
- Sarawuth Wantha
- Institute for Cardiovascular Prevention, LMU Munich, Pettenkoferstrasse 9, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Graham GJ, Locati M, Mantovani A, Rot A, Thelen M. The biochemistry and biology of the atypical chemokine receptors. Immunol Lett 2012; 145:30-8. [PMID: 22698181 DOI: 10.1016/j.imlet.2012.04.004] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 04/13/2012] [Indexed: 01/13/2023]
Abstract
A subset of chemokine receptors, initially called "silent" on the basis of their apparent failure to activate conventional signalling events, has recently attracted growing interest due to their ability to internalize, degrade, or transport ligands and thus modify gradients and create functional chemokine patterns in tissues. These receptors recognize distinct and complementary sets of ligands with high affinity, are strategically expressed in different cellular contexts, and lack structural determinants supporting Gα(i) activation, a key signalling event in cell migration. This is in keeping with the hypothesis that they have evolved to fulfil fundamentally different functions to the classical signalling chemokine receptors. Based on these considerations, these receptors (D6, Duffy antigen receptor for chemokines (DARC), CCX-CKR1 and CXCR7) are now collectively considered as an emerging class of 'atypical' chemokine receptors. In this article, we review the biochemistry and biology of this emerging chemokine receptor subfamily.
Collapse
Affiliation(s)
- G J Graham
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow G12 8TA, UK.
| | | | | | | | | |
Collapse
|
21
|
von Nickisch-Rosenegk M, Teschke T, Bier FF. Construction of an artificial cell membrane anchor using DARC as a fitting for artificial extracellular functionalities of eukaryotic cells. J Nanobiotechnology 2012; 10:1. [PMID: 22221512 PMCID: PMC3271035 DOI: 10.1186/1477-3155-10-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 01/05/2012] [Indexed: 11/10/2022] Open
Abstract
The need to functionalize cell membranes in a directed way for specific applications as single cell arrays or to force close cell-to-cell contact for artificial intercellular interaction and/or induction concerning stem cell manipulation or in general to have a tool for membrane and cell surface-associated processes, we envisaged a neutral inactive membrane anchor for extracellular entities to facillitate the above mentioned functionalities. The silent Duffy antigen/receptor for chemokines (DARC) is a receptor-like membrane protein of erythrocytes and mediates no cell transduction not at least regarding a missing or truncated G-loop and therefore it seemed to be the candidate for our cell membrane anchor. We isolated the genetic information of DARC from human genomic DNA and cloned it in a mammalian cell line as a fusion protein via a suitable plasmid vector. In this report we demonstrate that the human plasma membrane protein DARC can be used as an artificial anchor molecule in cell surface engineering applications. We constructed the fusion protein SNAP-tag-DARC, consisting of DARC and the self-labeling protein tag SNAP-tag® (Covalys). The SNAP-tag® served as an example for a molecular-technological developed protein that is artificially attached to the extracellular side of the plasma membrane through our DARC-anchor. SnapTag should serve as an example for any extracellular entity and was easy to detect by a commercial detection system. The synthesis of SNAP-tag-DARC, its correct incorporation into the cell membrane and the functionality of the SNAP-tag® were verified by RT-PCR, Western blotting and confocal fluorescence microscopy and showed the desired functionality as an membrane anchor for an extracellular application entity.
Collapse
|
22
|
Abstract
Atypical chemokine receptors (ACRs) are cell surface receptors with seven transmembrane domains structurally homologous to chemokine G-protein coupled receptors (GPCRs). However, upon ligation by cognate chemokines, ACRs fail to induce classical signaling and downstream cellular responses characteristic for GPCRs. Despite this, by affecting chemokine availability and function, ACRs impact on a multitude of pathophysiological events and have emerged as important molecular players in health and disease. This review discusses individual characteristics of the currently known ACRs, highlights their similarities and differences and attempts to establish their group identity. It summarizes the progress made in mapping ACR expression, understanding their diverse in vitro and in vivo functions of ACRs and uncovering their contributions to disease pathogeneses.
Collapse
Affiliation(s)
| | | | - Antal Rot
- MRC Centre for Immune Regulation, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|
23
|
Abstract
Chemokine receptors adorn the surface of leukocytes and other cell types ready to translate the extracellular chemokine environment into functional cellular outcomes. However, there are several molecules that, in many respects, look like chemokine receptors, but which do not have the ability to confer chemotactic potential to cell lines. This apparent silence spurred the search for signalling-independent functions and led to the development of new paradigms of chemokine regulation. In this review, we summarise the experimental basis for these ideas focussing on DARC and D6, the most studied members of this group of molecules. We discuss data generated using in vitro systems and genetically deficient mice, include results from observational human studies, and summarise the key findings of recent research. We take a critical look at current models of in vivo function highlighting important gaps in our knowledge and demonstrating that there is still much to find out about these enigmatic molecules.
Collapse
Affiliation(s)
- Chris A H Hansell
- Institute for Infection, Immunity & Inflammation, College of Medical, Veterinary & Life Sciences, Sir Graeme Davis Building, 120 University Place, Glasgow G12 8TA
| | - Catherine E Hurson
- Institute for Infection, Immunity & Inflammation, College of Medical, Veterinary & Life Sciences, Sir Graeme Davis Building, 120 University Place, Glasgow G12 8TA
| | - Robert J B Nibbs
- Institute for Infection, Immunity & Inflammation, College of Medical, Veterinary & Life Sciences, Sir Graeme Davis Building, 120 University Place, Glasgow G12 8TA
| |
Collapse
|
24
|
Mei J, Liu Y, Dai N, Favara M, Greene T, Jeyaseelan S, Poncz M, Lee JS, Worthen GS. CXCL5 regulates chemokine scavenging and pulmonary host defense to bacterial infection. Immunity 2010; 33:106-17. [PMID: 20643340 PMCID: PMC3748840 DOI: 10.1016/j.immuni.2010.07.009] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Revised: 05/13/2010] [Accepted: 07/07/2010] [Indexed: 12/14/2022]
Abstract
The chemokine sink hypothesis pertaining to erythrocyte Duffy Antigen Receptor for Chemokines (DARC) during inflammation has received considerable attention, but lacks direct in vivo evidence. Here we demonstrate, using mice with a targeted deletion in CXCL5, that CXCL5 bound erythrocyte DARC and impaired its chemokine scavenging in blood. CXCL5 increased the plasma concentrations of CXCL1 and CXCL2 in part through inhibiting chemokine scavenging, impairing chemokine gradients and desensitizing CXCR2, which led to decreased neutrophil influx to the lung, increased lung bacterial burden and mortality in an Escherichia coli pneumonia model. In contrast, CXCL5 exerted a predominant role in mediating neutrophil influx to the lung during inflammation after LPS inhalation. Platelets and lung resident cells were the sources of homeostatic CXCL5 in blood and inflammatory CXCL5 in the lung respectively. This study presents a paradigm whereby platelets and red cells alter chemokine scavenging and neutrophil-chemokine interaction during inflammation.
Collapse
Affiliation(s)
- Junjie Mei
- Division of Neonatology, Philadelphia, Pennsylvania, USA
| | - Yuhong Liu
- Division of Neonatology, Philadelphia, Pennsylvania, USA
| | - Ning Dai
- Division of Neonatology, Philadelphia, Pennsylvania, USA
| | - Michael Favara
- Division of Neonatology, Philadelphia, Pennsylvania, USA
| | - Teshell Greene
- Division of Hematology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Samithamby Jeyaseelan
- Department of Pathobiological Sciences and Center for Experimental Infectious Disease Research, Laboratory of Lung Biology, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Mortimer Poncz
- Division of Hematology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Janet S. Lee
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - G. Scott Worthen
- Division of Neonatology, Philadelphia, Pennsylvania, USA
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
25
|
Zarbock A, Bishop J, Müller H, Schmolke M, Buschmann K, Van Aken H, Singbartl K. Chemokine homeostasis vs. chemokine presentation during severe acute lung injury: the other side of the Duffy antigen receptor for chemokines. Am J Physiol Lung Cell Mol Physiol 2010; 298:L462-71. [PMID: 20061440 DOI: 10.1152/ajplung.00224.2009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Acute lung injury (ALI) still poses a major challenge in critical care medicine. Neutrophils, platelets, and chemokines are all considered key components in the development of ALI. The Duffy antigen receptor for chemokines (DARC) is thought to be involved in scavenging, transendothelial transport, and presentation of neutrophil-specific chemokines. DARC is expressed on endothelial cells and erythrocytes but not on leukocytes. Here, we show that DARC is crucial for chemokine-mediated leukocyte recruitment in vivo. However, we also demonstrate that changes in chemokine and chemokine receptor homeostasis, associated with Darc gene deficiency, exert strong anti-inflammatory effects. Neutrophils from Darc gene-deficient (Darc(-/-)) mice display a more prolonged downregulation of CXCR2 during severe inflammation than neutrophils from wild-type mice. In a CXCR2-dependent model of acid-induced ALI, Darc gene deficiency prevents ALI. Darc(-/-) mice demonstrate fully preserved oxygenation, only a small increase in vascular permeability, and a complete lack of pulmonary neutrophil recruitment. Further analysis reveals that only neutrophils but neither endothelial cells nor erythrocytes from Darc(-/-) mice confer protection from ALI. The protection appears to be due to abolished pulmonary recruitment of neutrophils from Darc(-/-) mice. The generation of neutrophil-platelet aggregates, a key mechanism in both pulmonary neutrophil recruitment and thrombus formation, is also affected by altered CXCR2 homeostasis in Darc(-/-) mice. CXCR2 blockade enhances the formation of platelet-neutrophil aggregates and thereby corrects a formerly unknown bleeding defect in Darc(-/-) mice. In summary, our study suggests that chemokine/chemokine receptor homeostasis plays a previously unrecognized and crucial role in severe ALI.
Collapse
Affiliation(s)
- Alexander Zarbock
- Department of Anesthesiology and Critical Care Medicine, University of Münster, Münster, Germany
| | | | | | | | | | | | | |
Collapse
|
26
|
Bonecchi R, Savino B, Borroni EM, Mantovani A, Locati M. Chemokine decoy receptors: structure-function and biological properties. Curr Top Microbiol Immunol 2010; 341:15-36. [PMID: 20373092 DOI: 10.1007/82_2010_19] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Chemokines induce cell migration through the activation of a distinct family of structurally related heterotrimeric G protein-coupled receptors (GPCR). Over the last few years, several receptors in this family that recognize chemokines but do not induce cell migration have been identified. These "atypical" chemokine receptors are unable to activate transduction events that lead directly to cell migration, but appear nonetheless to play a nonredundant role in the control of leukocyte recruitment at inflammatory sites and in tumors by shaping the chemoattractant gradient, either by removing, transporting, or concentrating their cognate ligands.
Collapse
Affiliation(s)
- Raffaella Bonecchi
- Department of Translational Medicine, University of Milan, 20089 Rozzano, Milan, Italy
| | | | | | | | | |
Collapse
|
27
|
Zerfaoui M, Naura AS, Errami Y, Hans CP, Rezk BM, Park J, Elsegeiny W, Kim H, Lord K, Kim JG, Boulares AH. Effects of PARP-1 deficiency on airway inflammatory cell recruitment in response to LPS or TNF: differential effects on CXCR2 ligands and Duffy Antigen Receptor for Chemokines. J Leukoc Biol 2009; 86:1385-92. [PMID: 19741160 PMCID: PMC2780916 DOI: 10.1189/jlb.0309183] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Revised: 07/11/2009] [Accepted: 07/30/2009] [Indexed: 12/14/2022] Open
Abstract
We reported that PARP-1 exhibits differential roles in expression of inflammatory factors. Here, we show that PARP-1 deletion was associated with a significant reduction in inflammatory cell recruitment to mouse airways upon intratracheal administration of LPS. However, PARP-1 deletion exerted little effect in response to TNF exposure. LPS induced massive neutrophilia and moderate recruitment of macrophages, and TNF induced recruitment of primarily macrophages with smaller numbers of neutrophils in the lungs. Following either exposure, macrophage recruitment was blocked severely in PARP-1(-/-) mice, and this was associated with a marked reduction in MCP-1 and MIP-1alpha. This association was corroborated partly by macrophage recruitment in response to intratracheal administration of MCP-1 in PARP-1(-/-) mice. Surprisingly, although neutrophil recruitment was reduced significantly in LPS-treated PARP-1(-/-) mice, neutrophil numbers increased in TNF-treated mice, suggesting that PARP-1 deletion may promote a macrophagic-to-neutrophilic shift in the inflammatory response upon TNF exposure. Neutrophil-specific chemokines mKC and MIP-2 were reduced significantly in lungs of LPS-treated but only partially reduced in TNF-treated PARP-1(-/-) mice. Furthermore, the MIP-2 antagonist abrogated the shift to a neutrophilic response in TNF-exposed PARP-1(-/-) mice. Although CXCR2 expression increased in response to either stimulus in PARP-1(+/+) mice, the DARC increased only in lungs of TNF-treated PARP-1(+/+) mice; both receptors were reduced to basal levels in treated PARP-1(-/-) mice. Our results show that the balance of pro-neutrophilic or pro-macrophagic stimulatory factors and the differential influence of PARP-1 on these factors are critical determinants for the nature of the airway inflammatory response.
Collapse
Affiliation(s)
- Mourad Zerfaoui
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Vielhauer V, Allam R, Lindenmeyer MT, Cohen CD, Draganovici D, Mandelbaum J, Eltrich N, Nelson PJ, Anders HJ, Pruenster M, Rot A, Schlöndorff D, Segerer S. Efficient renal recruitment of macrophages and T cells in mice lacking the duffy antigen/receptor for chemokines. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:119-31. [PMID: 19498001 PMCID: PMC2708800 DOI: 10.2353/ajpath.2009.080590] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/09/2009] [Indexed: 12/14/2022]
Abstract
The Duffy antigen/receptor for chemokines (DARC) is a chemokine-binding protein that is expressed on erythrocytes and renal endothelial cells. DARC-mediated endothelial transcytosis of chemokines may facilitate the renal recruitment of macrophages and T cells, as has been suggested for neutrophils. We studied the role of Darc in two mouse models of prolonged renal inflammation, one that primarily involves the tubulointerstitium (unilateral ureteral obstruction), and one that requires an adaptive immune response that leads to glomerulonephritis (accelerated nephrotoxic nephritis). Renal expression of Darc and its ligands was increased in both models. Leukocytes effectively infiltrated obstructed kidneys in Darc-deficient mice with pronounced T-cell infiltration at early time points. Development of interstitial fibrosis was comparable in both genotypes. Nephrotoxic nephritis was inducible in Darc-deficient mice, with both an increased humoral immune response and functional impairment during the early phase of disease. Leukocytes efficiently infiltrated kidneys of Darc-deficient mice, with increased cell numbers at early but not late time points. Taken together, renal inflammation developed more rapidly in DARC-deficient mice, without affecting the extent of renal injury at later time points. Thus, genetic elimination of Darc in mice does not prevent the development of renal infiltrates and may even enhance such development during the early phases of interstitial and glomerular diseases in mouse models of prolonged renal inflammation.
Collapse
Affiliation(s)
- Volker Vielhauer
- Medizinische Poliklinik, Campus Innenstadt, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Reutershan J, Harry B, Chang D, Bagby GJ, Ley K. DARC on RBC limits lung injury by balancing compartmental distribution of CXC chemokines. Eur J Immunol 2009; 39:1597-607. [PMID: 19499525 PMCID: PMC2733952 DOI: 10.1002/eji.200839089] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The Duffy antigen receptor for chemokines (DARC) has a high affinity for CC and CXC chemokines. However, it lacks the ability to induce cell responses that are typical for classical chemokine receptors. The role of DARC in inflammatory conditions remains to be elucidated. We studied the role of DARC in a murine model of acute lung injury. We found that in Darc-gene-deficient (Darc(-/-)) mice, LPS-induced PMN migration into the alveolar space was elevated more than twofold. In contrast, PMN adhesion to endothelial cells and within the interstitial space was reduced in Darc(-/-) mice. Darc(-/-) mice also exhibited increased microvascular permeability. Elevated PMN migration in Darc(-/-) mice was associated with increased concentrations of two essential CXCR2 ligands, CXCL1 and CXCL2/3 in the alveolar space. In the blood, CXCL1 was mostly associated with RBC in WT mice and with plasma in Darc(-/-) mice. We found that DARC on RBC prevented excessive PMN migration into the alveolar space. In contrast, DARC on non-hematopoietic cells appeared to have only minor effects on leukocyte trafficking in this model. These findings show how DARC regulates lung inflammation by controlling the distribution and presentation of chemokines that bind CXCR2.
Collapse
Affiliation(s)
- Jörg Reutershan
- Department of Anesthesiology and Intensive Care Medicine, University of Tübingen, Tübingen, Germany.
| | | | | | | | | |
Collapse
|
30
|
Abstract
Neutrophil recruitment into inflamed tissue in response to injury or infection is tightly regulated. Reduced neutrophil recruitment can result in a reduced ability to fight invading microorganisms. During inflammation, neutrophils roll along the endothelial wall of postcapillary venules and integrate inflammatory signals. Neutrophil activation by selectins and chemokines regulates integrin adhesiveness. Binding of activated integrins to their counter-receptors on endothelial cells induces neutrophil arrest and firm adhesion. Adherent neutrophils can be further activated to undergo cytoskeletal rearrangement, crawling, transmigration, superoxide production, and respiratory burst. Signaling through G-protein-coupled receptors, selectin ligands, Fc receptors and outside-in signaling through integrins are all involved in neutrophil activation, but their interplay in the multistep process of recruitment is only beginning to emerge. This review provides an overview of signaling in rolling and adherent neutrophils.
Collapse
Affiliation(s)
- Alexander Zarbock
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Hemolytic transfusion reactions (HTRs) are potentially fatal complications of blood transfusions. Many studies, primarily performed in vitro, have provided a great deal of insight into the initiating events of HTRs; however, it is not clear how they are modulated and how they combine to lead to one or more of the final common pathways. Recently developed mouse HTR models now make it possible to enhance our understanding of the pathogenesis of HTRs; this will allow for the rational design of specific therapies to prevent or ameliorate this serious complication in transfusion medicine. RECENT FINDINGS Mouse models support the hypothesis that 'cytokine storm' plays an important role in the pathogenesis of HTRs. Nitric oxide and endothelial cell dysfunction are also implicated in the pathophysiology of these reactions. In addition, the intriguing phenomenon of 'antigen loss,' in which antigen crosslinking by alloantibody leads to antigen removal rather than red blood cell clearance, has been modeled and explored. Finally, these mouse models were used to evaluate new therapeutic targets employing complement receptor 1 peptide homologues and the antimacrophage agent, liposomal clodronate. SUMMARY Models of HTRs are valuable for gaining a better understanding of the pathophysiology of these potentially fatal complications of blood transfusion. The participation of various inflammatory mediators was shown to play a role in these reactions in vivo. This knowledge will lead to novel treatment options.
Collapse
|
32
|
Shedding light on DARC: the role of the Duffy antigen/receptor for chemokines in inflammation, infection and malignancy. Inflamm Res 2009; 58:431-5. [DOI: 10.1007/s00011-009-0023-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2008] [Revised: 01/19/2009] [Accepted: 02/14/2009] [Indexed: 10/21/2022] Open
|
33
|
Reich D, Nalls MA, Kao WHL, Akylbekova EL, Tandon A, Patterson N, Mullikin J, Hsueh WC, Cheng CY, Coresh J, Boerwinkle E, Li M, Waliszewska A, Neubauer J, Li R, Leak TS, Ekunwe L, Files JC, Hardy CL, Zmuda JM, Taylor HA, Ziv E, Harris TB, Wilson JG. Reduced neutrophil count in people of African descent is due to a regulatory variant in the Duffy antigen receptor for chemokines gene. PLoS Genet 2009; 5:e1000360. [PMID: 19180233 PMCID: PMC2628742 DOI: 10.1371/journal.pgen.1000360] [Citation(s) in RCA: 274] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Accepted: 12/30/2008] [Indexed: 11/26/2022] Open
Abstract
Persistently low white blood cell count (WBC) and neutrophil count is a well-described phenomenon in persons of African ancestry, whose etiology remains unknown. We recently used admixture mapping to identify an approximately 1-megabase region on chromosome 1, where ancestry status (African or European) almost entirely accounted for the difference in WBC between African Americans and European Americans. To identify the specific genetic change responsible for this association, we analyzed genotype and phenotype data from 6,005 African Americans from the Jackson Heart Study (JHS), the Health, Aging and Body Composition (Health ABC) Study, and the Atherosclerosis Risk in Communities (ARIC) Study. We demonstrate that the causal variant must be at least 91% different in frequency between West Africans and European Americans. An excellent candidate is the Duffy Null polymorphism (SNP rs2814778 at chromosome 1q23.2), which is the only polymorphism in the region known to be so differentiated in frequency and is already known to protect against Plasmodium vivax malaria. We confirm that rs2814778 is predictive of WBC and neutrophil count in African Americans above beyond the previously described admixture association (P = 3.8 x 10(-5)), establishing a novel phenotype for this genetic variant.
Collapse
Affiliation(s)
- David Reich
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
| | - Michael A. Nalls
- Laboratory of Neurogenetics, Intramural Research Program, National Institute on Aging, Bethesda, Maryland, United States of America
- Laboratory of Epidemiology, Demography and Biometry, Intramural Research Program, National Institute on Aging, Bethesda, Maryland, United States of America
| | - W. H. Linda Kao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Ermeg L. Akylbekova
- Jackson Heart Study Analysis Group, Jackson State University, Jackson, Mississippi, United States of America
| | - Arti Tandon
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
| | - Nick Patterson
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
| | - James Mullikin
- Comparative Genomics Unit, Genome Technology Branch, National Human Genome Research Institute, Rockville, Maryland, United States of America
| | - Wen-Chi Hsueh
- Division of Medical Genetics, Department of Medicine, Department of Epidemiology and Biostatistics, Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Ching-Yu Cheng
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Inherited Disease Research Branch, National Human Genome Research Institute, Baltimore, Maryland, United States of America
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Man Li
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Alicja Waliszewska
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
- Laboratory of Molecular Immunology, Center for Neurologic Disease, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Julie Neubauer
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
| | - Rongling Li
- Department of Preventive Medicine, Center for Genomics and Bioinformatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Tennille S. Leak
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Lynette Ekunwe
- Jackson Heart Study Analysis Group, Jackson State University, Jackson, Mississippi, United States of America
| | - Joe C. Files
- Department of Medicine, Division of Hematology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Cheryl L. Hardy
- Department of Medicine, Division of Hematology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Joseph M. Zmuda
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Herman A. Taylor
- Jackson State University, Jackson, Mississippi, United States of America
- Tougaloo College, Jackson, Mississippi, United States of America
- University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Elad Ziv
- Division of General Internal Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Department of Epidemiology and Biostatistics, Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, United States of America
| | - Tamara B. Harris
- Laboratory of Epidemiology, Demography and Biometry, Intramural Research Program, National Institute on Aging, Bethesda, Maryland, United States of America
| | - James G. Wilson
- V.A. Medical Center, Jackson, Mississippi, United States of America
- University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| |
Collapse
|
34
|
Manicone AM. Role of the pulmonary epithelium and inflammatory signals in acute lung injury. Expert Rev Clin Immunol 2009; 5:63-75. [PMID: 19885383 PMCID: PMC2745180 DOI: 10.1586/177666x.5.1.63] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Acute lung injury (ALI) is a clinical disease marked by respiratory failure due to disruption of the epithelial and endothelial barrier, flooding of the alveolar compartment with protein-rich fluid and recruitment of neutrophils into the alveolar space. ALI affects approximately 200,000 patients annually in the USA and results in approximately 75,000 deaths. It is associated with prolonged mechanical ventilation, intensive medical care, high morbidity and mortality, and rising healthcare costs. Owing to its impact on public health, great strides have been made towards understanding the pathobiology of ALI to affect outcome. This review will focus on the role of the epithelial cell in the pathogenesis and resolution of ALI and the role of various inflammatory mediators in ALI.
Collapse
Affiliation(s)
- Anne M Manicone
- Center for Lung Biology, 815, Mercer Street, Box 358050, Seattle, WA 98115, USA,
| |
Collapse
|
35
|
Ge S, Song L, Serwanski DR, Kuziel WA, Pachter JS. Transcellular transport of CCL2 across brain microvascular endothelial cells. J Neurochem 2008; 104:1219-32. [PMID: 18289346 DOI: 10.1111/j.1471-4159.2007.05056.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The means by which the chemokine CCL2 produced in the brain parenchyma can recruit leukocytes lying behind the highly impervious endothelium of the blood-brain barrier (BBB) has remained a paradox. As other chemokines have been evidenced to stimulate their own synthesis and release by peripheral microvascular endothelial cells, and/or undergo transcytosis in the abluminal-to-luminal direction, we determined whether CCL2 experiences similar fates across brain microvascular endothelial cells (BMEC). Using cultured BMEC as a paradigm of the BBB, it was observed that exogenous unlabeled CCL2 actually depressed the release of endogenous CCL2, and further caused diminished CCL2 mRNA levels in these cells. On the other hand, exogenous (125)I-labeled CCL2 exhibited transport across BMEC in a manner that was sensitive to temperature, competition by excess unlabeled CCL2 but not unlabeled CCL3, knockdown of caveolin-1/caveolae, and elimination of the cognate CCL2 receptor CCR2. These results implied a facet of CCL2 transport by a transcellular mechanism partly involving binding of CCL2 to CCR2, and subsequent transfer to caveolae vesicles for transcytosis. This notion was supported by double-label immuno-electronmicroscopy, which revealed co-localization of caveolin-1 with exogenous CCL2, during this chemokine's transit across BMEC. Collectively, these findings provide a rationale by which CCL2, deposited on the abluminal side of the brain microvasculature during inflammatory episodes, can be relayed across the BBB to foster leukocyte recruitment.
Collapse
Affiliation(s)
- Shujun Ge
- Blood-Brain Barrier Laboratory, Department of Pharmacology, University of Connecticut Health Center, Farmington, Connecticut, USA
| | | | | | | | | |
Collapse
|
36
|
Chakera A, Seeber RM, John AE, Eidne KA, Greaves DR. The Duffy Antigen/Receptor for Chemokines Exists in an Oligomeric Form in Living Cells and Functionally Antagonizes CCR5 Signaling through Hetero-Oligomerization. Mol Pharmacol 2008; 73:1362-70. [DOI: 10.1124/mol.107.040915] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
37
|
Duffy antigen/receptor for chemokines (DARC) attenuates angiogenesis by causing senescence in endothelial cells. Angiogenesis 2007; 10:307-18. [DOI: 10.1007/s10456-007-9084-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Accepted: 10/09/2007] [Indexed: 10/22/2022]
|
38
|
Zarbock A, Schmolke M, Bockhorn SG, Scharte M, Buschmann K, Ley K, Singbartl K. The Duffy antigen receptor for chemokines in acute renal failure: A facilitator of renal chemokine presentation. Crit Care Med 2007; 35:2156-63. [PMID: 17855830 DOI: 10.1097/01.ccm.0000280570.82885.32] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Acute renal failure remains a major challenge in critical care medicine. Both neutrophils and chemokines have been proposed as key components in the development of acute renal failure. Although the Duffy antigen receptor for chemokines (DARC) is present in several tissues and a highly specific ligand for various chemokines, its exact role in vivo remains unclear. DESIGN Prospective, controlled experimental study. SETTING University-based research laboratory. SUBJECTS C57BL/6 wild-type and DARC gene-deficient mice (DARC-/-). INTERVENTIONS To unravel the functional relevance of DARC in vivo, we compared wild-type and DARC-/- using neutrophil-dependent models of acute renal failure, induced by either local (renal ischemia-reperfusion) or systemic (endotoxemia, lipopolysaccharide) injury. MEASUREMENTS AND MAIN RESULTS Plasma creatinine and blood urea nitrogen concentrations served as indicators of renal function or dysfunction. Enzyme-linked immunosorbent assays were used to measure tissue and plasma chemokine concentrations. We also performed immunostaining to localize chemokine expression and flow cytometry to evaluate neutrophil recruitment into the kidney. Following renal injury, wild-type mice developed moderate renal ischemia-reperfusion(lipopolysaccharide, 300% increase in plasma creatinine concentrations) to severe acute renal failure (renal ischemia-reperfusion, 40% mortality) as well as extensive renal neutrophil recruitment. DARC-/- mice exhibited no renal dysfunction (renal ischemia-reperfusion) or only very mild renal dysfunction (lipopolysaccharide, 20% increase in serum creatinine concentrations). DARC-/- mice showed no postischemic neutrophil infiltration. Although DARC-/- and wild-type mice exhibited similar global renal neutrophil-recruitment during endotoxemia, DARC-/- mice showed significantly impaired neutrophil extravasation. Total renal concentrations of the chemokine macrophage inflammatory protein 2, which has been shown to bind to DARC and to be crucial in postischemic acute renal failure, were either identical (lipopolysaccharide) or only moderately different (renal ischemia-reperfusion) between wild-type and DARC-/- mice. Immunostaining revealed an absence of macrophage inflammatory protein-2 in renal endothelial cells of DARC-/- mice. CONCLUSIONS We suggest that DARC predominantly exerts its effects by controlling spatial chemokine distribution, which in turn regulates neutrophil recruitment and subsequent acute renal failure.
Collapse
Affiliation(s)
- Alexander Zarbock
- Klinik und Poliklinik für Anästhesiologie und operative Intensivmedizin, Universitätsklinikum Münster, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
Edderkaoui B, Baylink DJ, Beamer WG, Wergedal JE, Porte R, Chaudhuri A, Mohan S. Identification of mouse Duffy antigen receptor for chemokines (Darc) as a BMD QTL gene. Genes Dev 2007; 17:577-85. [PMID: 17416748 PMCID: PMC1855174 DOI: 10.1101/gr.6009507] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2006] [Accepted: 02/05/2007] [Indexed: 12/14/2022]
Abstract
It is now well known that bone mineral density (BMD) variance is determined by both genetic and environmental factors. Accordingly, studies in human and animal models have revealed evidence for the presence of several quantitative trait loci (QTL) that contribute to BMD variations. However, the identification of BMD QTL genes remains a big challenge. In the current study, we focused our efforts to identify the BMD candidate gene in chromosome 1 (Chr 1) QTL that was detected from a cross involving high BMD CAST/EiJ (CAST) and low BMD C57BL/6J (B6) mice. To this end, we have combined several approaches including: (1) fine mapping the BMD QTL in Chr 1 of the B6.CAST F2 female mice using a large number of polymorphic markers; (2) the generation of congenic sublines of mice by repeated backcrossing of CAST with B6 mice and phenotype characterization; (3) expression profiling genes in the QTL region; and (4) SNP analyses to identify the mouse Duffy Antigen Receptor for Chemokines (Darc) as a candidate gene for Chr 1 BMD QTL2. We verified the involvement of the Darc protein in BMD variation by evaluating the skeletal phenotype of Darc-knockout mice and congenic sublines of mice carrying small chromosomal segments from CAST BMD QTL. Based on the findings that Darc-antibody blocked formation of multinucleated osteoclasts in vitro and that Darc from CAST binds chemokines, known to regulate osteoclast formation, with reduced affinity compared with Darc from B6 mice, we conclude that Darc regulates BMD negatively by increasing osteoclast formation, and that the genetic association between Darc gene polymorphism and BMD variations in humans merits investigation.
Collapse
Affiliation(s)
- Bouchra Edderkaoui
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California 92357, USA
| | - David J. Baylink
- Department of Medicine and Biochemistry, Loma Linda University, Loma Linda, California 92354, USA
| | | | - Jon E. Wergedal
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California 92357, USA
- Department of Medicine and Biochemistry, Loma Linda University, Loma Linda, California 92354, USA
| | - Ryan Porte
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California 92357, USA
| | - Asok Chaudhuri
- Laboratory of Cell Biology, New York Blood Center, New York, New York 10021, USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California 92357, USA
- Department of Medicine and Biochemistry, Loma Linda University, Loma Linda, California 92354, USA
| |
Collapse
|
40
|
Iiizumi M, Mohinta S, Bandyopadhyay S, Watabe K. Tumor-endothelial cell interactions: therapeutic potential. Microvasc Res 2007; 74:114-20. [PMID: 17498748 DOI: 10.1016/j.mvr.2007.04.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 04/02/2007] [Accepted: 04/04/2007] [Indexed: 12/30/2022]
Abstract
Metastasis is the primary cause of death in cancer patients. However, the molecular mechanism of the metastatic process is poorly understood because it involves multiple steps with a high degree of complexity. A critical step for successful establishment of secondary colonization is the hematogenous dissemination of malignant cells. During this process, the attachment of cancer cells to the endothelial cells on microvasculature is considered to be an essential step and many adhesion molecules as well as chemokines have been found to be involved in this process. This interaction of cancer-endothelial cell is considered not only to determine the physical site of metastasis, but also to provide the necessary anchorage to facilitate tumor cell extravasation. However, recent evidence indicates that this interaction also serves as a host defense mechanism and hinders the process of metastasis. The tumor metastases suppressor gene, KAI1, has been known to block metastatic process without affecting the primary tumor growth, and this protein has been found to be able to bind to the chemokine receptor, Duffy antigen receptor for chemokines (DARC), which is expressed on endothelial cells. Importantly, this interaction markedly induces senescence of tumor cells. This novel finding is not only significant in the context of molecular dissection of metastatic process but also in the therapeutic implication to develop drugs inhibiting metastasis.
Collapse
Affiliation(s)
- Megumi Iiizumi
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, 801 N. Rutledge St., P.O. Box 19626, Springfield, IL 62794-9626, USA
| | | | | | | |
Collapse
|
41
|
Iiizumi M, Bandyopadhyay S, Watabe K. Interaction of Duffy Antigen Receptor for Chemokines and KAI1: A Critical Step in Metastasis Suppression: Figure 1. Cancer Res 2007; 67:1411-4. [PMID: 17308076 DOI: 10.1158/0008-5472.can-06-3801] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Tumor metastases suppressor protein KAI1/CD82 is capable of blocking the tumor metastases without affecting the primary tumor formation, and its expression is significantly down-regulated in many types of human cancers. However, the exact molecular mechanism of the suppressor function of KAI1 remains elusive. Evidence from our laboratory supports a model in which tumor cells dislodge from the primary tumor and intravasate into the blood or lymphatic vessels followed by attachment to the endothelial cell surface whereby KAI1 interacts with the Duffy antigen receptor for chemokines (DARC) protein. This interaction transmits a senescent signal to cancer cells expressing KAI1, whereas cells that lost KAI1 expression can proliferate, potentially giving rise to metastases. Our model of the mechanism of action of KAI1 shows that metastasis suppressor activity can be dependent on interaction with host tissue and explains how KAI1 suppresses metastasis without affecting primary tumor formation. Taken together, in vitro and in vivo studies identify the KAI1-DARC interaction as a potential target for cancer therapy.
Collapse
Affiliation(s)
- Megumi Iiizumi
- Department of Medical Microbiology and Immunology, Southern Illinois University School of Medicine, 801 North Rutledge Street, Springfield, IL 62794, USA
| | | | | |
Collapse
|
42
|
Abstract
CXC chemokines are a subset of chemotactic cytokines that possess angiogenic or angiostatic properties. Using genetically engineered mice lacking the receptors for these ligands, recent research has demonstrated a significant role for CXC chemokines in the development and growth of prostate tumors. The Duffy antigen/receptor for chemokines (DARC), which only binds to CXC chemokines that have angiogenic properties, is a nonsignaling receptor expressed on erythrocytes that appears to function by clearing these chemokines from sites of overproduction. The majority of men of African descent lack this receptor on their erythrocytes, suggesting that loss of this receptor may contribute to aggressive tumor phenotypes in these individuals. Thus, CXC chemokines and the erythrocyte DARC may serve as important growth regulators and biomarkers for prostate cancer stage and progression.
Collapse
Affiliation(s)
- Alex B Lentsch
- University of Cincinnati College of Medicine, Department of Surgery, Cincinnati, OH 45267, USA.
| |
Collapse
|
43
|
Comerford I, Litchfield W, Harata-Lee Y, Nibbs RJB, McColl SR. Regulation of chemotactic networks by ‘atypical’ receptors. Bioessays 2007; 29:237-47. [PMID: 17295321 DOI: 10.1002/bies.20537] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Directed cell migration is a fundamental component of numerous biological systems and is critical to the pathology of many diseases. Although the importance of secreted chemoattractant factors in providing navigational cues to migrating cells bearing specific chemoattractant receptors is now well-established, how the function of these factors is regulated is not so well understood and may be of key importance to the design of new therapeutics for numerous human diseases. While regulation of migration clearly takes place on a number of different levels, it is becoming clear that so-called 'atypical' receptors play a role in scavenging, or altering the localisation of, chemoattractant molecules such as chemokines and complement components. These receptors do this through binding and/or internalising their chemoattractant ligands without activating signal transduction cascades leading to cell migration. The atypical chemokine receptor family currently comprises the receptors D6, DARC and CCX-CKR. In this review, we discuss the evidence from in vitro and in vivo studies that these receptors play a role in regulating cell migration, and speculate that other orphan receptors may also belong to this family. Furthermore, with the advent of gene therapy on the horizon, the therapeutic potential of these receptors in human disease is also considered.
Collapse
Affiliation(s)
- Iain Comerford
- School of Molecular and Biomedical Science, University of Adelaide, Australia.
| | | | | | | | | |
Collapse
|
44
|
Zijlstra A, Quigley JP. The DARC side of metastasis: shining a light on KAI1-mediated metastasis suppression in the vascular tunnel. Cancer Cell 2006; 10:177-8. [PMID: 16959609 DOI: 10.1016/j.ccr.2006.08.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Tumor cell metastasis to distant organs is an inefficient process that is limited in part by recently identified metastasis suppressors. Interactions between tumor cells and the surrounding stroma are thought to control much of cancer progression. In the August issue of Nature Medicine, demonstrate that specific cell surface interactions between the metastasis suppressor KAI1 on tumor cells and the decoy cytokine receptor DARC on adjacent vascular cells triggers senescence in the tumor cells and suppresses metastasis. These new observations demonstrate how metastasis suppressors can relay the restraint imposed by the stroma onto disseminating tumor cells.
Collapse
Affiliation(s)
- Andries Zijlstra
- Department of Pathology, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
45
|
Bandyopadhyay S, Zhan R, Chaudhuri A, Watabe M, Pai SK, Hirota S, Hosobe S, Tsukada T, Miura K, Takano Y, Saito K, Pauza ME, Hayashi S, Wang Y, Mohinta S, Mashimo T, Iiizumi M, Furuta E, Watabe K. Interaction of KAI1 on tumor cells with DARC on vascular endothelium leads to metastasis suppression. Nat Med 2006; 12:933-8. [PMID: 16862154 DOI: 10.1038/nm1444] [Citation(s) in RCA: 170] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2006] [Accepted: 06/14/2006] [Indexed: 11/10/2022]
Abstract
CD82, also known as KAI1, was recently identified as a prostate cancer metastasis suppressor gene on human chromosome 11p1.2 (ref. 1). The product of CD82 is KAI1, a 40- to 75-kDa tetraspanin cell-surface protein also known as the leukocyte cell-surface marker CD82 (refs. 1,2). Downregulation of KAI1 has been found to be clinically associated with metastatic progression in a variety of cancers, whereas overexpression of CD82 specifically suppresses tumor metastasis in various animal models. To define the mechanism of action of KAI1, we used a yeast two-hybrid screen and identified an endothelial cell-surface protein, DARC (also known as gp-Fy), as an interacting partner of KAI1. Our results indicate that the cancer cells expressing KAI1 attach to vascular endothelial cells through direct interaction between KAI1 and DARC, and that this interaction leads to inhibition of tumor cell proliferation and induction of senescence by modulating the expression of TBX2 and p21. Furthermore, the metastasis-suppression activity of KAI1 was significantly compromised in DARC knockout mice, whereas KAI1 completely abrogated pulmonary metastasis in wild-type and heterozygous littermates. These results provide direct evidence that DARC is essential for the function of CD82 as a suppressor of metastasis.
Collapse
Affiliation(s)
- Sucharita Bandyopadhyay
- Southern Illinois University School of Medicine, Department of Medical Microbiology, Immunology and Cell Biology, 801 N. Rutledge Street, PO Box 19626, Springfield, Illinois 62794-9626, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
The clinical manifestations of Plasmodium falciparum malaria are directly linked to the blood stage of the parasite life cycle. At the blood stage, the circulating merozoites invade erythrocytes via a specific invasion pathway often identified with its dependence or independence on sialic acid residues of the host receptor. The invasion process involves multiple receptor-ligand interactions that mediate a complex series of events in a period of approximately 1 min. Although the mechanism by which merozoites invade erythrocytes is not fully understood, recent advances have put a new perspective on the importance of developing a multivalent blood stage-malaria vaccine. In this review, we highlight the role of currently identified host invasion receptors in blood-stage malaria.
Collapse
Affiliation(s)
- S S Oh
- Division of Cell Biology, Caritas St. Elizabeth's Medical Center, Tufts University School of Medicine, 736 Cambridge Street, Boston, MA 02135, USA
| | | |
Collapse
|
47
|
Xu L, Niu J, Chaudhuri A. The domain on the mouse Duffy protein for Plasmodium yoelii binding and invasion to mouse erythrocytes. Mol Biochem Parasitol 2006; 146:142-50. [PMID: 16386320 DOI: 10.1016/j.molbiopara.2005.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2005] [Revised: 11/21/2005] [Accepted: 11/23/2005] [Indexed: 12/14/2022]
Abstract
Erythrocyte invasion by malaria parasites is a multi-step process requiring specific molecular interactions between merozoites and erythrocyte surface receptors. Human Duffy blood group protein is the receptor for Plasmodium vivax merozoite invasion to red blood cells. The cognate parasite ligand for Duffy protein is a 135 kDa Duffy binding protein (DBP). Previously, we defined the domain on the N-terminus of human Duffy protein required for DBP binding and showed that a 35-mer N-terminal peptide inhibited DBP binding to Duffy positive red cells in vitro. There is no efficient in vitro culture system or small animal model to study P. vivax ligand binding and invasion to red blood cells. Plasmodium yoelii is frequently used to study the interaction between host receptors and parasite ligands. Similar to human parasite P. vivax, rodent malaria parasite P. yoelii also uses Duffy protein on mouse RBCs for invasion. However, the domain on the mouse Duffy for P. yoelii binding is not known. In this communication, using a mouse model, we show that an antibody against the N-terminus of mouse Duffy protein inhibited P. yoelii invasion in the mouse. In addition, by using small peptides from the N-terminal exocellular domain, we defined the domain on the Duffy protein for P. yoelii binding and invasion to mouse erythrocytes. Our results also indicated that small peptides from the host receptor could act as decoy receptors and may be utilized as potential antimalarial drugs.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Antibodies, Monoclonal
- Antigens, Protozoan/immunology
- Antigens, Protozoan/metabolism
- Antimalarials/metabolism
- Antimalarials/pharmacology
- Disease Models, Animal
- Duffy Blood-Group System/chemistry
- Duffy Blood-Group System/immunology
- Duffy Blood-Group System/metabolism
- Erythrocytes/parasitology
- Malaria/parasitology
- Mice, Inbred C57BL
- Mice, Knockout
- Parasitemia
- Peptides/metabolism
- Peptides/pharmacology
- Plasmodium yoelii/pathogenicity
- Protein Structure, Tertiary
- Protozoan Proteins/immunology
- Protozoan Proteins/metabolism
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/metabolism
Collapse
Affiliation(s)
- Lili Xu
- Laboratory of Cell Biology, New York Blood Center, NY 10021, USA
| | | | | |
Collapse
|
48
|
Laurence ADJ. Location, movement and survival: the role of chemokines in haematopoiesis and malignancy. Br J Haematol 2006; 132:255-67. [PMID: 16409290 DOI: 10.1111/j.1365-2141.2005.05841.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chemokines are a family of over 40 small (8 kDa) related proteins with the function of moving cells along a chemotactic gradient, either to organise cells within an organ or to facilitate the movement of leucocytes around the body. Mouse models have implicated the importance of the chemokine CXCL12 in haematopoiesis and this has lead to the use of the inhibitor AMD3100 for autologous transplantation. This review will briefly discuss the biology of chemokines and their role in haematopoiesis and haematological malignancy together with the possible benefits and hazards of therapeutic modification of the chemokine system.
Collapse
|
49
|
Gardner L, Wilson C, Patterson AM, Bresnihan B, FitzGerald O, Stone MA, Ashton BA, Middleton J. Temporal expression pattern of Duffy antigen in rheumatoid arthritis: Up-regulation in early disease. ACTA ACUST UNITED AC 2006; 54:2022-6. [PMID: 16732566 DOI: 10.1002/art.21909] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Lucy Gardner
- Keele University at Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, UK
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Locati M, Torre YMDL, Galliera E, Bonecchi R, Bodduluri H, Vago G, Vecchi A, Mantovani A. Silent chemoattractant receptors: D6 as a decoy and scavenger receptor for inflammatory CC chemokines. Cytokine Growth Factor Rev 2005; 16:679-86. [PMID: 15996892 DOI: 10.1016/j.cytogfr.2005.05.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2005] [Accepted: 05/13/2005] [Indexed: 10/25/2022]
Abstract
The chemokine system includes at least three "silent" receptors, DARC, D6 and CCX CKR, with distinct specificity and tissue distribution. D6 binds most inflammatory, but not homeostatic, CC chemokines and shuttles in a ligand-independent way from the plasma membrane to endocytic compartments where chemokines are targeted to degradation. In vitro and in vivo evidence, including results with gene-targeted mice, is consistent with the view that D6 acts as a decoy and scavenger for inflammatory CC chemokines. Thus, D6 has unique functional and structural features, which make it ideally adapted to act as a chemokine decoy and scavenger receptor, strategically located on lymphatic endothelium to dampen inflammation in tissues and draining lymph nodes.
Collapse
Affiliation(s)
- Massimo Locati
- Institute of General Pathology, Medical Faculty, University of Milan, I-20133 Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|