51
|
Holmes MC, Zhang P, Nelson S, Summer WR, Bagby GJ. Neutrophil modulation of the pulmonary chemokine response to lipopolysaccharide. Shock 2002; 18:555-60. [PMID: 12462565 DOI: 10.1097/00024382-200212000-00012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
When cells within the intrapulmonary compartment are exposed to pathogens or their products such as lipopolysaccharide, they produce CXC chemokines in order to attract circulating neutrophils into the lower respiratory tract. Previous studies have shown that as neutrophils (PMNs) enter the lung, bronchoalveolar lavage (BAL) chemokine levels are decreased. In this study, we determined the intrapulmonary and systemic responses to two important rat chemokines, cytokine-induced neutrophil chemoattractant (CINC) and macrophage inflammatory protein-2 (MIP-2), to intratracheal (i.t.) LPS (100 microg in 0.5 mL of phosphate-buffered saline) under neutropenic (cyclophosphamide [CPA]) and neutrophilic (G-CSF) conditions. By 4 h after i.t. LPS, CPA pretreatment decreased PMN recruitment 83% and G-CSF increased PMN recruitment 91% compared with recruitment into the lung in vehicle-pretreated rats (42.7 +/- 19.3 million PMNs). Neutropenic rats had increased CINC and MIP-2 concentrations in BAL fluid 4 h after i.t. LPS when compared with levels seen in vehicle controls (P < 0.05). In vitro LPS-stimulated chemokine production by alveolar macrophages obtained from CPA- and vehicle-pretreated animals did not differ. The increase in BAL fluid chemokine levels in neutropenic rats corresponded to increased chemotaxis of neutrophils to BAL fluid from CPA-pretreated rats as compared with the chemotaxis response of PMN to BAL fluid from vehicle-pretreated rats. In contrast, G-CSF enhancement of neutrophil recruitment decreased chemotactic activity of BAL fluid collected 4 h after i.t. LPS. These data show that as neutrophils are recruited into the lung, they alter chemokine levels, which most likely serves to down-regulate the inflammatory response.
Collapse
Affiliation(s)
- Michael C Holmes
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | |
Collapse
|
52
|
Doroshenko T, Chaly Y, Savitskiy V, Maslakova O, Portyanko A, Gorudko I, Voitenok NN. Phagocytosing neutrophils down-regulate the expression of chemokine receptors CXCR1 and CXCR2. Blood 2002; 100:2668-71. [PMID: 12239185 DOI: 10.1182/blood.100.7.2668] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CXC chemokines play a central role in regulation of neutrophil activation and chemotaxis. Because the chemotactic responses of neutrophils are impaired after phagocytosis, we explored the effect of phagocytic stimuli on the expression of interleukin-8 (IL-8) receptors, CXCR1 and CXCR2, in human neutrophils. After phagocytosis of opsonized yeast, the expression of CXCR1 and CXCR2 was substantially down-regulated and was accompanied by reduced Ca(++) responses to corresponding ligands, IL-8 and neutrophil-activating peptide-2 (NAP-2). The levels of CXCR1 and CXCR2 mRNA were constant during phagocytic stimulation of neutrophils. Confocal microscopy revealed that CXCR reduction was not via internalization. Metalloproteinase inhibitor, 1,10-phenantroline, prevented the reduction of CXCRs induced by phagocytosis, indicating that proteolytic degradation may be responsible for down-regulation. These observations suggest that down-regulation of CXCR expression may substantially reduce the responsiveness of phagocytosing neutrophils to CXC chemokines.
Collapse
Affiliation(s)
- Tatyana Doroshenko
- Laboratory of Cellular and Molecular Immunology, Institute of Hematology and Blood Transfusion, Minsk, Belarus, Russia
| | | | | | | | | | | | | |
Collapse
|
53
|
Beaulieu S, Robbiani DF, Du X, Rodrigues E, Ignatius R, Wei Y, Ponath P, Young JW, Pope M, Steinman RM, Mojsov S. Expression of a functional eotaxin (CC chemokine ligand 11) receptor CCR3 by human dendritic cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:2925-36. [PMID: 12218106 DOI: 10.4049/jimmunol.169.6.2925] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Critical to the function of Ag-presenting dendritic cells (DCs) is their capacity to migrate to lymphoid organs and to sites of inflammation. A final stage of development, termed maturation, yields DCs that are strong stimulators of T cell-mediated immunity and is associated with a remodeling of the cell surface that includes a change in the levels of expression of many molecules, including chemokine receptors. We show in this study that CCR3, a chemokine receptor initially discovered on eosinophils, is also expressed by human DCs that differentiate from blood monocytes, DCs that emigrate from skin (epidermal and dermal DCs), and DCs derived from CD34+ hemopoietic precursors in bone marrow, umbilical cord blood, and cytokine-elicited peripheral blood leukapheresis. Unlike other chemokine receptors, such as CCR5 and CCR7, the expression of CCR3 is not dependent on the state of maturation. All DC subsets contain a large intracellular pool of CCR3. The surface expression of CCR3 is not modulated following uptake of particulate substances such as zymosan or latex beads. CCR3 mediates in vitro chemotactic responses to the known ligands, eotaxin and eotaxin-2, because the DC response to these chemokines is inhibited by CCR3-specific mAbs. We postulate that expression of CCR3 may underlie situations where both DCs and eosinophils accumulate in vivo, such as the lesions of patients with Langerhans cell granulomatosis.
Collapse
Affiliation(s)
- Sylvie Beaulieu
- Laboratory of Cellular Physiology and Immunology, The Rockefeller University and Department of Medicine, Memorial Sloan-Kettering Cancer Center, Weill Medical College, Cornell University, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Vita C, Drakopoulou E, Ylisastigui L, Bakri Y, Vizzavona J, Martin L, Parmentier M, Gluckman JC, Benjouad A. Synthesis and characterization of biologically functional biotinylated RANTES. J Immunol Methods 2002; 266:53-65. [PMID: 12133622 DOI: 10.1016/s0022-1759(02)00096-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Development of specifically labeled chemokines that retain their biological properties should be useful for analyzing their mechanisms of action both under physiological and pathological conditions. Here, we report the chemical synthesis and characterization of RANTES (regulated upon activation normal T cell expressed and secreted) derivatives that were biotinylated at residues 1, 25, 33, 45, or 67. Gel filtration and ultracentrifugation experiments showed that biotinylation at position 45 or 67 decreased the aggregation tendency of the chemokine to a dimeric state. Competition experiments, using a stably transfected CHO-K1 cell line overexpressing human CCR5, a RANTES receptor, indicated that derivatives biotinylated at positions 1, 25, and 67 bound to CCR5 with the same affinity as native RANTES. Flow cytometry analysis showed that RANTES biotinylated at residue 67 (B67-RANTES) bound more efficiently to primary macrophages than the other derivatives. Such binding was dependent on cell surface glycosaminoglycans (GAGs) since it was reduced when macrophages or HeLa cells expressing or not CCR5 were first treated with GAG-specific enzymes. In addition, B67-RANTES modulated CCR5 expression on lymphocytes and elicited chemotaxis of monocytes in the same manner as unmodified RANTES. Thus, B67-RANTES acts as a CCR5 agonist and may be useful to study the role of RANTES in pathologies such as, for example, human immunodeficiency virus (HIV) infection and inflammatory disorders.
Collapse
Affiliation(s)
- Claudio Vita
- CEA, Département d'Ingénierie et d'Etudes des Protéines, CE Saclay, F-91190 Gif-sur-Yvette, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Seely AJE, Naud JF, Campisi G, Giannias B, Liu S, DiCarlo A, Ferri LE, Pascual JL, Tchervenkov J, Christou NV. Alteration of chemoattractant receptor expression regulates human neutrophil chemotaxis in vivo. Ann Surg 2002; 235:550-9. [PMID: 11923612 PMCID: PMC1422471 DOI: 10.1097/00000658-200204000-00014] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To elucidate the mechanisms that regulate human neutrophil delivery in vivo, as well as the mechanisms that lead to observed reduction in polymorphonuclear (PMN) delivery to remote sites in septic patients. METHODS Alterations in human PMN chemoattractant receptor expression and chemotactic function in vivo were evaluated in two distinct experiments: exudate PMNs (PMNs that have undergone transmigration to skin window blisters in controls) and septic PMNs (circulating PMNs from septic patients in the intensive care unit) were both separately compared with control circulating PMNs. RESULTS Exudate PMNs displayed increased C5a receptors and C5a chemotaxis, and reduced interleukin-8 receptors (both IL-8 RA and IL-8 RB) and IL-8 chemotaxis. Septic PMNs displayed reduced C5a and IL-8 receptors and decreased C5a chemotaxis but no change in IL-8 chemotaxis. IL-8 but not C5a receptor gene expression decreased in parallel to receptor alteration. CONCLUSIONS These results suggest that change in PMN chemoattractant receptor expression serves to regulate PMN chemotaxis in vivo; that exudate PMN chemotaxis depends more on C5a than IL-8; and that diminished chemoattractant receptors and chemotaxis in septic PMNs may explain decreased PMN delivery in these patients.
Collapse
Affiliation(s)
- Andrew J E Seely
- Division of General Surgery, McGill University Health Center, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Schwartzkopff F, Brandt E, Petersen F, Flad HD, Bock L, Ludwig A. The CXC chemokine NAP-2 mediates differential heterologous desensitization of neutrophil effector functions elicited by platelet-activating factor. J Interferon Cytokine Res 2002; 22:257-67. [PMID: 11911809 DOI: 10.1089/107999002753536239] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
During early inflammation, the chemoattractants neutrophil-activating peptide-2 (NAP-2), platelet-activating factor (PAF), and complement component C5a are rapidly generated within the vasculature and potently induce effector functions in neutrophils, such as chemotaxis and degranulation. We investigated whether these mediators would cross-desensitize each other's activities on isolated neutrophils. We demonstrate that NAP-2 and C5a desensitize degranulation of neutrophils in response to PAF. However, whereas C5a-mediated desensitization correlated with the downregulation of PAF binding sites on neutrophils, NAP-2 did not regulate PAF receptors, nor did it modulate their calcium signaling. In further contrast to C5a, which desensitized PAF-induced neutrophil chemotaxis, NAP-2 did not affect the chemotatic response to PAF. These observations indicate that NAP-2 mediates selective desensitization of PAF-induced neutrophil degranulation by a mechanism downstream from PAF receptors, still allowing migration toward PAF. Thus, a role for NAP-2 may be to limit PAF-dependent vascular tissue damage by preventing degranulation of neutrophils without affecting their migration into the inflamed tissue.
Collapse
Affiliation(s)
- Franziska Schwartzkopff
- Department of Immunology and Cell Biology, Research Center Borstel, D-23845 Borstel, Germany
| | | | | | | | | | | |
Collapse
|
57
|
Zhou N, Luo Z, Luo J, Liu D, Hall JW, Pomerantz RJ, Huang Z. Structural and functional characterization of human CXCR4 as a chemokine receptor and HIV-1 co-receptor by mutagenesis and molecular modeling studies. J Biol Chem 2001; 276:42826-33. [PMID: 11551942 DOI: 10.1074/jbc.m106582200] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The human CXC chemokine receptor 4 (CXCR4) is a receptor for the chemokine stromal cell-derived factor (SDF-1alpha) and a co-receptor for the entry of specific strains of human immunodeficiency virus type I (HIV-1). CXCR4 is also recognized by an antagonistic chemokine, the viral macrophage inflammatory protein II (vMIP-II) encoded by human herpesvirus type VIII. SDF-1alpha or vMIP-II binding to CXCR4 can inhibit HIV-1 entry via this co-receptor. An approach combining protein structural modeling and site-directed mutagenesis was used to probe the structure-function relationship of CXCR4, and interactions with its ligands SDF-1alpha and vMIP-II and HIV-1 envelope protein gp120. Hypothetical three-dimensional structures were proposed by molecular modeling studies of the CXCR4.SDF-1alpha complex, which rationalize extensive biological information on the role of CXCR4 in its interactions with HIV-1 envelope protein gp120. With site-directed mutagenesis, we have identified that the amino acid residues Asp (D20A) and Tyr (Y21A) in the N-terminal domain and the residue Glu (E268A) in extracellular loop 3 (ECL3) are involved in ligand binding, whereas the mutation Y190A in extracellular loop 2 (ECL2) impairs the signaling mediated by SDF-1alpha. As an HIV-1 co-receptor, we found that the N-terminal domain, ECL2, and ECL3 of CXCR4 are involved in HIV-1 entry. These structural and mutational studies provide valuable information regarding the structural basis for CXCR4 activity in chemokine binding and HIV-1 viral entry, and could guide the design of novel targeted inhibitors.
Collapse
Affiliation(s)
- N Zhou
- Kimmel Cancer Center, Department of Medicine, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | |
Collapse
|
58
|
Schaniel C, Rolink AG, Melchers F. Attractions and migrations of lymphoid cells in the organization of humoral immune responses. Adv Immunol 2001; 78:111-68. [PMID: 11432203 DOI: 10.1016/s0065-2776(01)78003-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- C Schaniel
- Basel Institute for Immunology, CH-4005 Basel, Switzerland.
| | | | | |
Collapse
|
59
|
Dulkys Y, Kluthe C, Buschermöhle T, Barg I, Knöss S, Kapp A, Proudfoot AE, Elsner J. IL-3 induces down-regulation of CCR3 protein and mRNA in human eosinophils. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:3443-53. [PMID: 11544337 DOI: 10.4049/jimmunol.167.6.3443] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cytokines and chemokines are responsible for the attraction and activation of eosinophils in allergic and inflammatory diseases. Whereas cytokines such as IL-3, IL-5, and GM-CSF activate eosinophils via heterodimeric receptors containing a distinct alpha-chain (binding domain) and a common beta-chain (signaling domain), chemokines such as eotaxin activate eosinophils via seven-transmembrane G(i) protein-coupled CCRs. Recent studies have demonstrated the importance of CCR3 on human eosinophils that undergo receptor recycling after chemokine activation, but the modulation of this receptor by cytokines has not yet been addressed. In this study, we demonstrate that IL-3 induces a dose- and time-dependent down-regulation of CCR3 from the surface of human eosinophils comparable to the CCR3-specific ligand eotaxin, whereas IL-5, GM-CSF, IL-4, IL-10, IL-13, IFN-gamma, and TNF-alpha had no effect. Maximal down-regulation of CCR3 in response to IL-3 was reached at 24 h. Reduction of CCR3 surface protein in response to IL-3 could be prevented by an anti-IL-3 mAb and was neither due to the release of CC chemokines nor to nonspecific binding of IL-3 to CCR3. Moreover, down-regulation was prevented by phenylarsine oxide, a nonspecific inhibitor of receptor internalization. After 24 h, IL-3-induced decrease of CCR3 surface expression correlated with diminished mRNA expression, suggesting a transcriptional regulation mechanism. Since wortmannin partially inhibited IL-3- but not eotaxin-induced CCR3 down-regulation, receptor down-modulation seems to underlie different signaling events. Therefore, these data suggest a novel role for the cytokine IL-3 in the activation process of eosinophils and its predominant chemokine receptor CCR3.
Collapse
Affiliation(s)
- Y Dulkys
- Department of Dermatology and Allergology, Hannover Medical University, Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|
60
|
Glynn PC, Henney EM, Hall IP. Peripheral blood neutrophils are hyperresponsive to IL-8 and Gro-alpha in cryptogenic fibrosing alveolitis. Eur Respir J 2001; 18:522-9. [PMID: 11589350 DOI: 10.1183/09031936.01.00057901] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Cryptogenic fibrosing alveolitis (CFA) is characterized by increased pulmonary recruitment of peripheral blood neutrophils (PBNs) by interleukin (IL)-8 and other chemotactic mediators. This study investigated whether, in CFA, the PBN motility response is primed by IL-8 and growth-related oncogene (Gro)-alpha, as demonstrated in other neutrophilic inflammatory diseases, and whether the motility response of PBNs to IL-8 and Gro-alpha can be abrogated using a selective antagonist for the neutrophil receptor for IL-8 and Gro-alpha, CXCR2. The percentage of PBNs to undergo shape change (%SC), spontaneously and in response to IL-8 and Gro-alpha, was measured in patients with CFA (n=10) and controls (n=10), and the effect of the CXCR2 antagonist SB272844 studied. Plasma levels of IL-8, and Gro-alpha were measured using an enzyme-linked immunosorbent assay (ELISA). The %SC of unstimulated PBNs and the potency of Gro-alpha and IL-8 to produce neutrophil polarization was greater in CFA than in controls; dose which produces 50% of maximal effect (EC50) of IL-8 was 3.6 +/- 0.7 nM for CFA versus 6.3 +/- 1.0 nM for controls; p<0.05. SB272844 inhibited Gro-alpha induced but not IL-8 induced neutrophil shape change (equilibrium constant (KD) 123 +/- 18 nM). Plasma concentrations of Gro-alpha were increased in patients with CFA. PBNs are spontaneously activated and undergo a greater motility response to IL-8 and Gro-alpha in CFA. Interleukin-8 and growth-related oncogene-alpha, circulating in substimulatory amounts in cryptogenic fibrosing alveolitis, may prime the peripheral blood neutrophils motility response, thus increasing their capacity for migration to the lung. Selective CXCR2 antagonists may be useful to block the Gro-alpha-induced priming response whilst preserving neutrophil functions mediated by CXCR1, the alternative neutrophil receptor for interleukin-8.
Collapse
Affiliation(s)
- P C Glynn
- Division of Therapeutics, University Hospital of Nottingham, UK
| | | | | |
Collapse
|
61
|
Ethuin F, Delarche C, Benslama S, Gougerot‐Pocidalo M, Jacob L, Chollet‐Martin S. Interleukin‐12 increases interleukin 8 production and release by human polymorphonuclear neutrophils. J Leukoc Biol 2001. [DOI: 10.1189/jlb.70.3.439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Frédéric Ethuin
- Laboratoire d’Immunologie and Unité INSERM 479, Hôpital Bichat, and
- Département d’Anesthésie‐Réanimation, Hôpital Saint‐Louis, Paris, France
| | | | - Sylvie Benslama
- Laboratoire d’Immunologie and Unité INSERM 479, Hôpital Bichat, and
| | | | - Laurent Jacob
- Département d’Anesthésie‐Réanimation, Hôpital Saint‐Louis, Paris, France
| | | |
Collapse
|
62
|
Remick DG, Green LB, Newcomb DE, Garg SJ, Bolgos GL, Call DR. CXC chemokine redundancy ensures local neutrophil recruitment during acute inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 159:1149-57. [PMID: 11549608 PMCID: PMC1850476 DOI: 10.1016/s0002-9440(10)61791-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Previous publications demonstrated that elevated systemic levels of interleukin (IL)-8 decrease local neutrophil recruitment. We tested whether sustained, high plasma levels of IL-8 would prevent local inflammation after inflammatory insults. Mice carrying the transgene for human IL-8 were separated on the basis of their plasma levels of IL-8 into IL-8-positive (plasma levels >90 ng/ml) and IL-8-negative (IL-8 below detection). Presence of the IL-8 transgene did not improve survival or morbidity nor did it alter peritoneal neutrophil recruitment induced by the cecal ligation and puncture model of sepsis. In an acute lung injury model created by intratracheal injection of acid, IL-8-positive mice showed no reduction in alveolar neutrophil recruitment. There was no difference in the local recruitment of neutrophils when either thioglycollate or glycogen was injected intraperitoneally. We examined the chemotactic response to murine chemokines to test how neutrophil recruitment occurs in the setting of elevated plasma IL-8 and found that neutrophils from both IL-8-positive and -negative mice respond equally well to recombinant KC or macrophage inflammatory protein (MIP)-2. We measured KC and MIP-2 in the peritoneum after thioglycollate injection and demonstrated that IL-8-positive mice have significantly higher levels of the chemokines compared to the IL-8-negative mice. Antibody inhibition of KC and MIP-2 in the IL-8-positive mice significantly decreased peritoneal neutrophil recruitment in response to thioglycollate, clarifying their important role in the local neutrophil recruitment. Our data demonstrate that despite the presence of high plasma levels of IL-8, neutrophils may still be recruited to sites of local inflammation because of chemokine redundancy.
Collapse
Affiliation(s)
- D G Remick
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0602, USA.
| | | | | | | | | | | |
Collapse
|
63
|
Weber K, Gröne HJ, Röcken M, Klier C, Gu S, Wank R, Proudfoot A, Nelson P, Weber C. Selective recruitment of Th2-type cells and evasion from a cytotoxic immune response mediated by viral macrophage inhibitory protein-II. Eur J Immunol 2001. [DOI: 10.1002/1521-4141(200108)31:8<2458::aid-immu2458>3.0.co;2-l] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
64
|
Yousefi S, Cooper PR, Potter SL, Mueck B, Jarai G. Cloning and expression analysis of a novel G‐protein‐coupled receptor selectively expressed on granulocytes. J Leukoc Biol 2001. [DOI: 10.1189/jlb.69.6.1045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Shida Yousefi
- Novartis Horsham Research Centre, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Paul R Cooper
- Novartis Horsham Research Centre, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Suzanne L Potter
- Novartis Horsham Research Centre, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Beatrice Mueck
- Novartis Horsham Research Centre, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Gabor Jarai
- Novartis Horsham Research Centre, Horsham, West Sussex RH12 5AB, United Kingdom
| |
Collapse
|
65
|
Hofman V, Ricci V, Mograbi B, Brest P, Luciano F, Boquet P, Rossi B, Auberger P, Hofman P. Helicobacter pylori lipopolysaccharide hinders polymorphonuclear leucocyte apoptosis. J Transl Med 2001; 81:375-84. [PMID: 11310830 DOI: 10.1038/labinvest.3780245] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
A prominent histologic feature of Helicobacter pylori infection is a dense infiltration of polymorphonuclear leukocytes (PMNL) in gastric mucosa. H. pylori lipopolysaccharide (LPS) has been recognized as a primary virulence factor evoking acute mucosal inflammatory reaction. Previous works have shown that H. pylori LPS immunologic activities are lower than those of enterobacterial LPS. However, the effect of H. pylori LPS on spontaneous PMNL apoptosis, and mechanisms by which this H. pylori LPS may promote PMNL survival remain to be established. In this study, we investigated, by both morphologic and biochemical approaches, the action of H. pylori LPS on PMNL apoptosis in vitro, using broth culture filtrates (BCF) of H. pylori strains with different genotypes. We found that BCF from H. pylori caused a significant delay in spontaneous PMNL apoptosis and this delay was independent of the VacA, cag pathogenicity island and urease status. We demonstrated that LPS in BCF is responsible for this effect because it was abrogated by the LPS antagonist B287 (a synthetic analog of Rhodobactersphaeroides lipid A). Moreover, BCF from H. pylori induced P42/44MAP kinase activation in PMNL. Similar results were obtained with BCF of an Escherichia coli strain. Taken together these data suggest that longer survival of PMNL induced by H. pylori LPS may increase gastric epithelium injury in H. pylori-associated diseases.
Collapse
Affiliation(s)
- V Hofman
- Laboratoire d'Anatomie-Pathologique, Faculté de Médecine, Nice, France
| | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Weber C, Weber KS, Klier C, Gu S, Wank R, Horuk R, Nelson PJ. Specialized roles of the chemokine receptors CCR1 and CCR5 in the recruitment of monocytes and T(H)1-like/CD45RO(+) T cells. Blood 2001; 97:1144-6. [PMID: 11159551 DOI: 10.1182/blood.v97.4.1144] [Citation(s) in RCA: 185] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chemokines and their receptors control the emigration of leukocytes during inflammation. The role of the RANTES (regulated on activation normal T-cell expressed and secreted) receptors CCR1 and CCR5 in the selective recruitment of monocytes, T(H)1-like T-cell clones, and peripheral T cells enriched for CD45RO(+) "memory" cells were tested in a system in which arrest under flow conditions is triggered by RANTES immobilized to activated endothelium. With the use of selective nonpeptide receptor antagonists or blocking antibodies, it was found that the RANTES-induced arrest of these cells was mediated predominantly by CCR1. In contrast, CCR5 mainly contributed to the spreading in shear flow, and both CCR1 and CCR5 supported transendothelial chemotaxis toward RANTES. The data in this study reveal specialized roles of apparently redundant receptors in distinct steps of leukocyte trafficking and suggest that not all receptors currently used to define mononuclear cell subsets are involved in their direct recruitment from the circulation.
Collapse
Affiliation(s)
- C Weber
- Institut für Prophylaxe der Kreislaufkrankheiten, Medizinische Poliklinik, and Institut für Immunologie, Ludwig-Maximilians-Universität München, Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
67
|
Loetscher P, Pellegrino A, Gong JH, Mattioli I, Loetscher M, Bardi G, Baggiolini M, Clark-Lewis I. The ligands of CXC chemokine receptor 3, I-TAC, Mig, and IP10, are natural antagonists for CCR3. J Biol Chem 2001; 276:2986-91. [PMID: 11110785 DOI: 10.1074/jbc.m005652200] [Citation(s) in RCA: 227] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Th1 and Th2 lymphocytes express a different repertoire of chemokine receptors (CCRs). CXCR3, the receptor for I-TAC (interferon-inducible T cell alpha-chemoattractant), Mig (monokine induced by gamma-interferon), and IP10 (interferon-inducible protein 10), is expressed preferentially on Th1 cells, whereas CCR3, the receptor for eotaxin and several other CC chemokines, is characteristic of Th2 cells. While studying responses that are mediated by these two receptors, we found that the agonists for CXCR3 act as antagonists for CCR3. I-TAC, Mig, and IP10 compete for the binding of eotaxin to CCR3-bearing cells and inhibit migration and Ca(2+) changes induced in such cells by stimulation with eotaxin, eotaxin-2, MCP-2 (monocyte chemottractant protein-2), MCP-3, MCP-4, and RANTES (regulated on activation normal T cell expressed and secreted). A hybrid chemokine generated by substituting the first eight NH(2)-terminal residues of eotaxin with those of I-TAC bound CCR3 with higher affinity than eotaxin or I-TAC (3- and 10-fold, respectively). The hybrid was 5-fold more potent than I-TAC as an inhibitor of eotaxin activity and was effective at concentrations as low as 5 nm. None of the antagonists described induced the internalization of CCR3, indicating that they lack agonistic effects and thus qualify as pure antagonists. These results suggest that chemokines that attract Th1 cells via CXCR3 can concomitantly block the migration of Th2 cells in response to CCR3 ligands, thus enhancing the polarization of T cell recruitment.
Collapse
Affiliation(s)
- P Loetscher
- Theodor-Kocher Institute, University of Bern, P. O. Box 99, CH-3000 Bern 9, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
68
|
Zaslaver A, Feniger-Barish R, Ben-Baruch A. Actin filaments are involved in the regulation of trafficking of two closely related chemokine receptors, CXCR1 and CXCR2. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:1272-84. [PMID: 11145710 DOI: 10.4049/jimmunol.166.2.1272] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The ligand-induced internalization and recycling of chemokine receptors play a significant role in their regulation. In this study, we analyzed the involvement of actin filaments and of microtubules in the control of ligand-induced internalization and recycling of CXC chemokine receptor (CXCR)1 and CXCR2, two closely related G protein-coupled receptors that mediate ELR-expressing CXC chemokine-induced cellular responses. Nocodazole, a microtubule-disrupting agent, did not affect the IL-8-induced reduction in cell surface expression of CXCR1 and CXCR2, nor did it affect the recycling of these receptors following ligand removal and cell recovery at 37 degrees C. In contrast, cytochalasin D, an actin filament depolymerizing agent, promoted the IL-8-induced reduction in cell surface expression of both CXCR1 and CXCR2. Cytochalasin D significantly inhibited the recycling of both CXCR1 and CXCR2 following IL-8-induced internalization, the inhibition being more pronounced for CXCR2 than for CXCR1. Potent inhibition of recycling was observed also when internalization of CXCR2 was induced by another ELR-expressing CXC chemokine, granulocyte chemotactic protein-2. By the use of carboxyl terminus-truncated CXCR1 and CXCR2 it was observed that the carboxyl terminus domains of CXCR1 and CXCR2 were partially involved in the regulation of the actin-mediated process of receptor recycling. The cytochalasin D-mediated inhibition of CXCR2 recycling had a functional relevance because it impaired the ability of CXCR2-expressing cells to mediate cellular responses. These results suggest that actin filaments, but not microtubules, are involved in the regulation of the intracellular trafficking of CXCR1 and CXCR2, and that actin filaments may be required to enable cellular resensitization following a desensitized refractory period.
Collapse
MESH Headings
- Actins/antagonists & inhibitors
- Actins/physiology
- Amino Acid Sequence
- Biological Transport/drug effects
- Biological Transport/genetics
- Biological Transport/immunology
- Cell Line
- Cell Membrane/drug effects
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Cell Migration Inhibition
- Chemotaxis/drug effects
- Chemotaxis/genetics
- Chemotaxis/immunology
- Cytochalasin D/pharmacology
- Down-Regulation/drug effects
- Down-Regulation/genetics
- Down-Regulation/immunology
- Humans
- Interleukin-8/pharmacology
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Neutrophils/drug effects
- Neutrophils/immunology
- Neutrophils/metabolism
- Nocodazole/pharmacology
- Peptide Fragments/genetics
- Peptide Fragments/physiology
- Protein Structure, Tertiary/genetics
- Protein Structure, Tertiary/physiology
- Receptors, Interleukin-8A/antagonists & inhibitors
- Receptors, Interleukin-8A/blood
- Receptors, Interleukin-8A/genetics
- Receptors, Interleukin-8A/metabolism
- Receptors, Interleukin-8B/antagonists & inhibitors
- Receptors, Interleukin-8B/blood
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Transfection
Collapse
Affiliation(s)
- A Zaslaver
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | | | | |
Collapse
|
69
|
Ylisastigui L, Bakri Y, Amzazi S, Gluckman JC, Benjouad A. Soluble glycosaminoglycans Do not potentiate RANTES antiviral activity on the infection of primary macrophages by human immunodeficiency virus type 1. Virology 2000; 278:412-22. [PMID: 11118364 DOI: 10.1006/viro.2000.0670] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Macrophages play an important role in human immunodeficiency virus (HIV)-1 infection. They exist in various differentiation and activation states in vivo, a heterogeneity that may affect their interactions with HIV-1 and susceptibility to drugs. Here, we found that RANTES and MIP-1beta, heparin, or soluble chondroitin sulfate B, but not chondroitin sulfate A, inhibited HIV-1(BaL) infection of macrophages obtained as the adherent cells of 5-day cultures of blood mononuclear cells (PBMC), followed by 2 days without either nonadherent PBMC or added cytokines (MDM-5d), whereas they did not affect infection of macrophages obtained as the adherent cells recovered from 1-h incubation of PBMC and subsequent 7-day culture with macrophage colony-stimulating factor (MDM-MCSF). Such different behavior was not related to differences in HIV-1 binding but rather to postbinding steps, as HIV-1(BaL) attached similarly to MDM-5d and MDM-MCSF, a binding that was affected by soluble glycosaminoglycans but not by RANTES. Of note, CCR5 expression on both types of MDM was comparable, and it was not downregulated by RANTES on either. Mixing RANTES with each of the glycosaminoglycans did not restore inhibition of MDM-MCSF infection by HIV-1; however, heparin at concentrations that had low antiviral activity for MDM-5d counteracted RANTES anti-HIV-1 activity for these cells, whereas chondroitin sulfate B had no additive effect on that of RANTES. Both glycosaminoglycans affected RANTES binding to MDM. Thus, in contrast to cell surface proteoglycans that contribute to the attachment of RANTES to macrophages and enhance its anti-HIV-1 activity, soluble glycosaminoglycans do not facilitate, and may even offset, the anti-HIV-1 activity of RANTES.
Collapse
Affiliation(s)
- L Ylisastigui
- Institut National de la Santé et de la Recherche Médicale (INSERM) E0013, Laboratoire d'Immunologie Cellulaire et Immunopathologie de l'Ecole Pratique des Hautes Etudes, Hôpital de la Pitié-Salp etrière, Paris, France
| | | | | | | | | |
Collapse
|
70
|
Godaly G, Hang L, Frendéus B, Svanborg C. Transepithelial neutrophil migration is CXCR1 dependent in vitro and is defective in IL-8 receptor knockout mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:5287-94. [PMID: 11046063 DOI: 10.4049/jimmunol.165.9.5287] [Citation(s) in RCA: 130] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neutrophil migration across infected mucosal surfaces is chemokine dependent, but the role of chemokine receptors has not been investigated. In this study, chemokine receptors were shown to be expressed by epithelial cells lining the urinary tract, and to play an essential role for neutrophil migration across the mucosal barrier. Uroepithelial CXCR1 and CXCR2 expression was detected in human urinary tract biopsies, and in vitro infection of human uroepithelial cell lines caused a dramatic increase in both receptors. As a consequence, there was higher binding of IL-8 to the cells and the IL-8-dependent neutrophil migration across the infected epithelial cell layers was enhanced. Abs to IL-8 or to the CXCR1 receptor inhibited this increase by 60% (p<0.004), but anti-CXCR2 Abs had no effect, suggesting that CXCR1 was the more essential receptor in this process. Similar observations were made in the mouse urinary tract, where experimental infection stimulated epithelial expression of the murine IL-8 receptor, followed by a rapid flux of neutrophils into the lumen. IL-8 receptor knockout mice, in contrast, failed to express the receptor, their neutrophils were unable to cross the epithelial barrier, and accumulated in massive numbers in the tissues. These results demonstrate that epithelial cells express CXC receptors and that infection increases receptor expression. Furthermore, we show that CXCR1 is required for neutrophil migration across infected epithelial cell layers in vitro, and that the murine IL-8 receptor is needed for neutrophils to cross the infected mucosa of the urinary tract in vivo.
Collapse
Affiliation(s)
- G Godaly
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Lund, Sweden
| | | | | | | |
Collapse
|
71
|
Ludwig A, Ehlert JE, Flad HD, Brandt E. Identification of distinct surface-expressed and intracellular CXC-chemokine receptor 2 glycoforms in neutrophils: N-glycosylation is essential for maintenance of receptor surface expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:1044-52. [PMID: 10878382 DOI: 10.4049/jimmunol.165.2.1044] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The G protein-coupled CXC-chemokine receptor CXCR-2 mediates activation of neutrophil effector functions in response to multiple ligands, including IL-8 and neutrophil-activating peptide 2 (NAP-2). Although CXCR-2 has been successfully cloned and expressed in several cell lines, the molecular properties of the native neutrophil-expressed receptor have remained largely undefined. Here we report on the identification and characterization of distinct CXCR-2 glycoforms and their subcellular distribution in neutrophils. Immunoprecipitation and Western blot analyses of surface-expressed receptors covalently linked to IL-8 or NAP-2 as well as in their unloaded state revealed the occurrence of a single CXCR-2 variant with an apparent size of 56 kDa. According to deglycosylation experiments surface-expressed CXCR-2 carries two N-linked 9-kDa carbohydrate moieties that are both of complex structure. In addition, two other CXCR-2 variants of 38 and 40 kDa were found to occur exclusively intracellular and to carry N-glycosylations of high mannose or hybrid type. These receptors did not participate in ligand-induced receptor trafficking, while surface-expressed CXCR-2 was internalized and re-expressed following stimulation with NAP-2. By enzymatic removal of one 9-kDa carbohydrate moiety in surface-expressed CXCR-2 we can show that neither NAP-2-induced trafficking nor signaling of the receptor is dependent on its full glycosylation. Instead, glycosylation was found to protect CXCR-2 from proteolytic attack, as even partial deglycosylation is associated with serine protease-mediated disappearance of the receptor from the neutrophil surface. Thus, although not directly involved in signaling, glycosylation appears to be required to maintain neutrophil responsiveness to CXC-chemokines during inflammation.
Collapse
Affiliation(s)
- A Ludwig
- Department of Immunology, Forschungszentrum Borstel, Borstel, Germany.
| | | | | | | |
Collapse
|
72
|
Abstract
AbstractChemokines are small peptides that are potent activators and chemoattractants for leukocyte subpopulations and some nonhemopoietic cells. Their actions are mediated by a family of 7-transmembrane G-protein–coupled receptors, the size of which has grown considerably in recent years and now includes 18 members. Chemokine receptor expression on different cell types and their binding and response to specific chemokines are highly variable. Significant advances have been made in understanding the regulation of chemokine receptor expression and the intracellular signaling mechanisms used in bringing about cell activation. Chemokine receptors have also recently been implicated in several disease states including allergy, psoriasis, atherosclerosis, and malaria. However, most fascinating has been the observation that some of these receptors are used by human immunodeficiency virus type 1 in gaining entry into permissive cells. This review will discuss structural and functional aspects of chemokine receptor biology and will consider the roles these receptors play in inflammation and in infectious diseases.
Collapse
|
73
|
Abstract
Chemokines are small peptides that are potent activators and chemoattractants for leukocyte subpopulations and some nonhemopoietic cells. Their actions are mediated by a family of 7-transmembrane G-protein–coupled receptors, the size of which has grown considerably in recent years and now includes 18 members. Chemokine receptor expression on different cell types and their binding and response to specific chemokines are highly variable. Significant advances have been made in understanding the regulation of chemokine receptor expression and the intracellular signaling mechanisms used in bringing about cell activation. Chemokine receptors have also recently been implicated in several disease states including allergy, psoriasis, atherosclerosis, and malaria. However, most fascinating has been the observation that some of these receptors are used by human immunodeficiency virus type 1 in gaining entry into permissive cells. This review will discuss structural and functional aspects of chemokine receptor biology and will consider the roles these receptors play in inflammation and in infectious diseases.
Collapse
|
74
|
Abstract
Interleukin-8 was originally discovered as one of the first chemokines activating neutrophil granulocytes (neutrophils) after secretion by lipopolysaccharide-stimulated monocytes. A wealth of information has been gathered concerning the intracellular events mediated by interleukin-8 and the role of interleukin-8 in numerous physiologic and pathophysiologic processes. We discuss recent advances in the understanding of the initial intracellular signals elicited by interleukin-8. Detailed investigation of these events has led to the identification of subtle but significant differences in the signal transduction processes evoked by interleukin-8 receptors. In particular, much has been learned concerning differences in the cellular mechanisms leading to desensitization, internalization, and recycling of interleukin-8 receptors, and functional consequences of interleukin-8 receptor diversity are now being unraveled.
Collapse
Affiliation(s)
- H U Zeilhofer
- Department of Experimental and Clinical Pharmacology and Toxicology, University of Erlangen-Nürnberg, Erlangen, Germany.
| | | |
Collapse
|
75
|
Dunican A, Grutkoski P, Leuenroth S, Ayala A, Simms HH. Neutrophils regulate their own apoptosis via preservation of CXC receptors. J Surg Res 2000; 90:32-8. [PMID: 10781372 DOI: 10.1006/jsre.2000.5829] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Dysregulated neutrophil (PMN) apoptosis is thought to contribute to an exaggerated inflammatory response in diseases such as acute respiratory distress syndrome (ARDS) and multiple organ dysfunction syndrome (MODS). The CXC chemokines, interleukin-8 (IL-8) and growth-related oncogene alpha (Gro-alpha), contribute to the inflammatory response and suppress PMN apoptosis. We hypothesized that PMN generation of CXC chemokines is an autocrine/paracrine mechanism for amplification of the PMN inflammatory response via suppression of apoptosis. METHODS Freshly isolated human PMNs from healthy donors were incubated with IL-8 or Gro-alpha (100 ng/ml) for 0-12 h, and apoptosis was analyzed at 24 h. De novo synthesis of IL-8 or Gro-alpha was measured using an ELISA. To determine if receptors were available to bind these newly synthesized ligands (125)I radiolabeled monoclonal antibodies specific for each receptor (CXCRI, CXCRII) were used to determine PMN receptor density. Comparison was by one-way ANOVA. RESULTS Significant suppression of apoptosis was seen at 24 h with only 4 h exposure to IL-8 or Gro-alpha (n = 5, P < 0.05). PMNs cultured with IL-8 for 4 h produced 31 +/- 4.3 ng/ml IL-8 by 24 h; PMNs cultured with Gro-alpha produced 19.7 +/- 4.0 ng/ml Gro-alpha (n = 6, P < 0. 05). Neither chemokine induced significant production of the other chemokine. The addition of either ligand promoted upregulation of CXCR1 (n = 4, P < 0.05) at 24 h. However, CXCR2 was downregulated by Gro-alpha and IL-8 to 71 +/- 7.5 and 79 +/- 6.3% of control, respectively (P < 0.05). CONCLUSION IL-8 and Gro-alpha, which suppress apoptosis, stimulate their own production after short-term incubation with PMNs. PMNs maintain the ability to respond to these chemokines through expression of the CXC receptors which suggests that PMNs are active participants in the suppression of apoptosis at inflammatory sites. CXCRI remains upregulated after prolonged stimulation and may be an important target for mediating neutrophil responses to IL-8.
Collapse
Affiliation(s)
- A Dunican
- Division of Surgical Research, Department of Surgery, Brown University School of Medicine, Rhode Island Hospital Department of Surgery, Providence, RI 02903, USA
| | | | | | | | | |
Collapse
|
76
|
Zimmermann N, Daugherty BL, Stark JM, Rothenberg ME. Molecular analysis of CCR-3 events in eosinophilic cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:1055-64. [PMID: 10623856 DOI: 10.4049/jimmunol.164.2.1055] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CCR-3 is a major receptor involved in regulating eosinophil trafficking. Initial analysis of chemokine receptors has demonstrated unique receptor events in different cell types, indicating the importance of investigating CCR-3 events in eosinophilic cell lines. We now report that the eosinophilic cell line, acute myelogenous leukemia (AML) 14.3D10, expresses eosinophil granule proteins and eotaxin, but has no detectable expression of eosinophil chemokine receptors. Treatment of the cell line with butyric acid and IL-5 results in a dose-dependent synergistic induction of CCR-3 and, to a lesser extent, CCR-1 and CCR-5. Interestingly, using a luciferase reporter construct under the control of the hCCR-3 promoter, the uninduced and induced cells display high, but comparable, levels of promoter activity. Differentiated AML cells developed enhanced functional activation, as indicated by adhesion to respiratory epithelial cells and chemokine-induced transepithelial migration. Chemokine signaling did not inhibit adenylate cyclase activity even though calcium transients were blocked by pertussis toxin. Additionally, chemokine-induced calcium transients were inhibited by pretreatment with PMA, but not forskolin. Eotaxin treatment of differentiated AML cells resulted in marked down-modulation of CCR-3 expression for at least 18 h. Receptor internalization was not dependent upon chronic ligand exposure and was not accompanied by receptor degradation. Thus, CCR-3 is a late differentiation marker on AML cells and uses a signal transduction pathway involving rapid and prolonged receptor internalization, calcium transients inhibitable by protein kinase C but not protein kinase A, and the paradoxical lack of inhibition of adenylate cyclase activity.
Collapse
MESH Headings
- Adenylate Cyclase Toxin
- Adenylyl Cyclase Inhibitors
- Butyric Acid/pharmacology
- Calcium Signaling/immunology
- Cell Differentiation/immunology
- Cell Movement/immunology
- Clone Cells
- Down-Regulation/immunology
- Enzyme Activation/immunology
- Eosinophils/immunology
- Eosinophils/metabolism
- Eosinophils/pathology
- Humans
- Interleukin-5/physiology
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Ligands
- Pertussis Toxin
- Promoter Regions, Genetic/drug effects
- Promoter Regions, Genetic/immunology
- Protein Kinase C/metabolism
- Receptors, CCR3
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
- Receptors, Chemokine/physiology
- Transcriptional Activation/drug effects
- Transcriptional Activation/immunology
- Tumor Cells, Cultured
- Virulence Factors, Bordetella/pharmacology
Collapse
Affiliation(s)
- N Zimmermann
- Division of Pulmonary Medicine, Department of Pediatrics, Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | |
Collapse
|
77
|
Feniger-Barish R, Ran M, Zaslaver A, Ben-Baruch A. Differential modes of regulation of cxc chemokine-induced internalization and recycling of human CXCR1 and CXCR2. Cytokine 1999; 11:996-1009. [PMID: 10623425 DOI: 10.1006/cyto.1999.0510] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Studies of human neutrophil IL-8 receptors, CXCR1 and CXCR2, have shown that the two receptors are differentially regulated by ELR(+)-CXC chemokines, that they differ functionally and may have diverse roles in mediating the inflammatory process. To elucidate the role of CXCR1 and CXCR2 in inflammation and to delineate the basis for the divergent regulation of these receptors by IL-8 and NAP-2, we characterized the IL-8- and NAP-2-induced mechanisms regulating the expression of each receptor, focusing on receptor internalization and recycling. Using HEK 293 cell transfectants, IL-8 was shown to induce significantly higher levels of CXCR2 internalization than NAP-2. Moreover, although CXCR2 bound IL-8 and NAP-2 with similarly high affinity, IL-8 functionally competed with and displaced NAP-2, and prompted high levels of internalization, similar to those induced by IL-8 alone. In a system providing an identical cellular milieu for reliable comparisons between CXCR1 and CXCR2, we have shown that the mechanisms controlling the internalization of CXCR1 diverge from those regulating CXCR2 internalization. Whereas IL-8-induced internalization of CXCR1 was profoundly dependent on a region of the carboxyl terminus expressing six phosphorylation sites, internalization of CXCR2 was primarily regulated by a membrane proximal domain of the carboxyl terminus that does not express phosphorylation sites. Analysis of receptor re-expression on the plasma membrane indicated that at early time points following removal of free ligand and incubation of the cells at 37 degrees C, receptor recycling accounted for recovery of CXCR1 and CXCR2 expression, whereas at later time points other processes may be involved in receptor re-expression. Phosphorylation-independent mechanisms were shown to direct both receptors to the recycling pathway. The differential control of CXCR1 vs CXCR2 internalization by IL-8 and NAP-2, as well as by phosphorylation-mediated mechanisms, suggests that a chemokine- and receptor-specific mode of regulation of internalization may contribute to the divergent activities of these receptors.
Collapse
Affiliation(s)
- R Feniger-Barish
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | | | | | | |
Collapse
|
78
|
Foxman EF, Kunkel EJ, Butcher EC. Integrating conflicting chemotactic signals. The role of memory in leukocyte navigation. J Cell Biol 1999; 147:577-88. [PMID: 10545501 PMCID: PMC2151176 DOI: 10.1083/jcb.147.3.577] [Citation(s) in RCA: 185] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/1999] [Accepted: 09/27/1999] [Indexed: 11/23/2022] Open
Abstract
Leukocytes navigate through complex chemoattractant arrays, and in so doing, they must migrate from one chemoattractant source to another. By evaluating directional persistence and chemotaxis during neutrophil migration under agarose, we show that cells migrating away from a local chemoattractant, against a gradient, display true chemotaxis to distant agonists, often behaving as if the local gradient were without effect. We describe two interrelated properties of migrating cells that allow this to occur. First, migrating leukocytes can integrate competing chemoattractant signals, responding as if to the vector sum of the orienting signals present. Second, migrating cells display memory of their recent environment: cells' perception of the relative strength of orienting signals is influenced by their history, so that cells prioritize newly arising or newly encountered attractants. We propose that this cellular memory, by promoting sequential chemotaxis to one attractant after another, is in fact responsible for the integration of competitive orienting signals over time, and allows combinations of chemoattractants to guide leukocytes in a step-by-step fashion to their destinations within tissues.
Collapse
Affiliation(s)
- E F Foxman
- Laboratory of Immunology and Vascular Biology, Stanford University Medical School, Stanford, California 94305, USA.
| | | | | |
Collapse
|
79
|
Cyster JG, Ngo VN, Ekland EH, Gunn MD, Sedgwick JD, Ansel KM. Chemokines and B-cell homing to follicles. Curr Top Microbiol Immunol 1999; 246:87-92; discussion 93. [PMID: 10396043 DOI: 10.1007/978-3-642-60162-0_11] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- J G Cyster
- Department of Microbiology and Immunology, University of California at San Francisco 94143, USA
| | | | | | | | | | | |
Collapse
|
80
|
Barlic J, Khandaker MH, Mahon E, Andrews J, DeVries ME, Mitchell GB, Rahimpour R, Tan CM, Ferguson SS, Kelvin DJ. beta-arrestins regulate interleukin-8-induced CXCR1 internalization. J Biol Chem 1999; 274:16287-94. [PMID: 10347185 DOI: 10.1074/jbc.274.23.16287] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The functional role of neutrophils during acute inflammatory responses is regulated by two high affinity interleukin-8 receptors (CXCR1 and CXCR2) that are rapidly desensitized and internalized upon binding their cognate chemokine ligands. The efficient re-expression of CXCR1 on the surface of neutrophils following agonist-induced internalization suggests that CXCR1 surface receptor turnover may involve regulatory pathways and intracellular factors similar to those regulating beta2-adrenergic receptor internalization and re-expression. To examine the internalization pathway utilized by ligand-activated CXCR1, a CXCR1-GFP construct was transiently expressed in two different cell lines, HEK 293 and RBL-2H3 cells. While interleukin-8 stimulation promoted CXCR1 sequestration in RBL-2H3 cells, receptor internalization in HEK 293 cells required co-expression of G protein-coupled receptor kinase 2 and beta-arrestin proteins. The importance of beta-arrestins in CXCR1 internalization was confirmed by the ability of a dominant negative beta-arrestin 1-V53D mutant to block internalization of CXCR1 in RBL-2H3 cells. A role for dynamin was also demonstrated by the lack of CXCR1 internalization in dynamin I-K44A dominant negative mutant-transfected RBL-2H3 cells. Agonist-promoted co-localization of transferrin and CXCR1-GFP in endosomes of RBL-2H3 cells confirmed that receptor internalization occurs via clathrin-coated vesicles. Our data provides a direct link between agonist-induced internalization of CXCR1 and a requirement for G protein-coupled receptor kinase 2, beta-arrestins, and dynamin during this process.
Collapse
Affiliation(s)
- J Barlic
- Laboratory of Molecular Immunology and Inflammation, John P. Robarts Research Institute, London, Ontario N6G 2V4, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Mitchell GB, Khandaker MH, Rahimpour R, Xu L, Lazarovits AI, Pickering JG, Suria H, Madrenas J, Pomerantz DK, Feldman RD, Kelvin DJ. CD45 modulation of CXCR1 and CXCR2 in human polymorphonuclear leukocytes. Eur J Immunol 1999; 29:1467-76. [PMID: 10359100 DOI: 10.1002/(sici)1521-4141(199905)29:05<1467::aid-immu1467>3.0.co;2-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
All leukocytes express the cell surface glycoprotein CD45, which has intrinsic intracellular protein tyrosine phosphatase activity. CD45 is known to play a regulatory role in activation-induced signaling in lymphocytes; however, little is known of its role in non-lymphoid leukocytes. Therefore, we examined the potential effect of CD45 on chemokine-induced signaling in human neutrophils (polymorphonuclear cells, PMN). Treating isolated PMN for 2 h with an anti-CD45RB antibody (Bra11) down-modulated expression of the chemokine receptors CXCR1 and CXCR2 to 44 +/- 10% and 47 +/- 9% of their respective controls. The tyrosine kinase inhibitors genistein and herbimycin A significantly inhibited the Bra11-induced down-modulation of CXCR1 and CXCR2. Furthermore, Bra11-treated PMN were functionally inhibited in their capacity to exhibit IL-8-induced transient intracellular Ca2+ increases. Selected targeting of CXC receptors is indicated by the fact that N-formyl-Met-Leu-Phe (fMLP) receptor expression and function were not lost following Bra11 treatment. The effect of Bra11 on IL-8-mediated function and receptor expression was paralleled by decreased tyrosine phosphorylation of a 54- to 60-kDa protein. These findings indicate that CD45 can act to modulate PMN responses to chemokines; thus agents regulating CD45 can potentially modulate leukocyte traffic and may represent a novel therapeutic approach towards the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- G B Mitchell
- Department of Physiology, The University of Western Ontario, and John P. Robarts Research Institute, London, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Meddows-Taylor S, Martin DJ, Tiemessen CT. Impaired interleukin-8-induced degranulation of polymorphonuclear neutrophils from human immunodeficiency virus type 1-infected individuals. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 1999; 6:345-51. [PMID: 10225834 PMCID: PMC103721 DOI: 10.1128/cdli.6.3.345-351.1999] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Degranulation of peripheral blood polymorphonuclear leukocytes (PMNLs) was monitored in human immunodeficiency virus (HIV) type 1 (HIV-1)-infected individuals with or without pulmonary tuberculosis (HIV/TB and HIV groups, respectively) by measuring the release of beta-glucuronidase induced by interleukin-8 (IL-8). This was increased in a dose-dependent manner in the control groups consisting of healthy blood donors and patients with pulmonary tuberculosis. In contrast, PMNLs from the HIV and HIV/TB groups responded reciprocally in the same assay; that is, higher IL-8 input concentrations resulted in the release of less enzyme than lower IL-8 input concentrations. The degranulation response of PMNLs from HIV-1-infected individuals was similarly altered for another agonist, N-formyl-methionyl-leucyl-phenylalanine, suggesting that impairment of the nonoxidative armature of PMNL was a more generalized phenomenon. However, impaired IL-8-induced degranulation was found to be associated with the reduced expression of both IL-8 receptors, A and B, on whole-blood PMNLs from HIV-1-infected patients compared with that on whole-blood PMNLs from healthy persons. The density of IL-8RA, in particular, was most reduced on the surfaces of PMNLs from those patients with the poorest degranulation in response to IL-8. Inefficient agonist-induced degranulation may contribute to the increased susceptibility of HIV-1-infected persons to secondary microbial infections, this being further exacerbated in HIV/TB patients who, in addition, display defects in phagocytosis and oxidative burst.
Collapse
Affiliation(s)
- S Meddows-Taylor
- MRC AIDS Virus Research Unit, National Institute for Virology, and Department of Virology, University of the Witwatersrand, Johannesburg, South Africa
| | | | | |
Collapse
|
83
|
Zimmermann N, Conkright JJ, Rothenberg ME. CC chemokine receptor-3 undergoes prolonged ligand-induced internalization. J Biol Chem 1999; 274:12611-8. [PMID: 10212240 DOI: 10.1074/jbc.274.18.12611] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CC chemokine receptor-3 (CCR-3) is a major receptor involved in regulating eosinophil trafficking; therefore, elucidation of ligand-induced CCR-3 events has important implications in understanding the biological and pathological properties of eosinophils. Previous studies have demonstrated that unique receptor events occur in different cell types supporting investigation of CCR-3-mediated events in eosinophilic cells. We now report biochemical characterization of CCR-3 internalization following exposure of eosinophils to CCR-3 ligands. Treatment of freshly isolated human eosinophils with CCR-3 ligands resulted in marked and differential internalization of CCR-3 in a dose-dependent manner. Exposure to 100 ng/ml eotaxin reduced surface expression to 43, 43, and 76% at 15 min, 1 h, and 3 h, respectively. RANTES (reduced on activation T cell expressed and secreted) treatment induced more significant and prolonged internalization of CCR-3 than eotaxin; following 100 ng/ml of RANTES, 29, 24, and 47% of the receptor was expressed at 15 min, 3 h, and 18 h, respectively. Confocal microscopy demonstrated that receptor modulation involved receptor internalization by an endocytic pathway shared with the transferrin receptor. Receptor internalization was accompanied by partial degradation of CCR-3, and reexpression of CCR-3 was dependent in part upon de novo protein synthesis. Internalization was not blocked by pretreatment of eosinophils with pertussis toxin. Furthermore, staurosporine did not inhibit internalization although it blocked phorbol 12-myristate 13-acetate-induced CCR-3 down-modulation. These results demonstrate that CCR-3 ligands induce differential receptor internalization that is not dependent upon Gi-protein coupling, calcium transients, or protein kinase C.
Collapse
Affiliation(s)
- N Zimmermann
- Division of Pulmonary Medicine, Allergy, and Clinical Immunology, Department of Pediatrics, Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | | | | |
Collapse
|
84
|
Schütze S, Machleidt T, Adam D, Schwandner R, Wiegmann K, Kruse ML, Heinrich M, Wickel M, Krönke M. Inhibition of receptor internalization by monodansylcadaverine selectively blocks p55 tumor necrosis factor receptor death domain signaling. J Biol Chem 1999; 274:10203-12. [PMID: 10187805 DOI: 10.1074/jbc.274.15.10203] [Citation(s) in RCA: 162] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The 55-kDa receptor for tumor necrosis factor (TR55) triggers multiple signaling cascades initiated by adapter proteins like TRADD and FAN. By use of the primary amine monodansylcadaverine (MDC), we addressed the functional role of tumor necrosis factor (TNF) receptor internalization for intracellular signal distribution. We show that MDC does not prevent the interaction of the p55 TNF receptor (TR55) with FAN and TRADD. Furthermore, the activation of plasmamembrane-associated neutral sphingomyelinase activation as well as the stimulation of proline-directed protein kinases were not affected in MDC-treated cells. In contrast, activation of signaling enzymes that are linked to the "death domain" of TR55, like acid sphingomyelinase and c-Jun-N-terminal protein kinase as well as TNF signaling of apoptosis in U937 and L929 cells, are blocked in the presence of MDC. The results of our study suggest a role of TR55 internalization for the activation of select TR55 death domain signaling pathways including those leading to apoptosis.
Collapse
Affiliation(s)
- S Schütze
- Institute of Immunology, University of Kiel, 24105 Kiel, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Pelchen-Matthews A, Signoret N, Klasse PJ, Fraile-Ramos A, Marsh M. Chemokine receptor trafficking and viral replication. Immunol Rev 1999; 168:33-49. [PMID: 10399063 DOI: 10.1111/j.1600-065x.1999.tb01281.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Chemokines and chemokine receptors have emerged as crucial factors controlling the development and function of leukocytes. Recent studies have indicated that, in addition to these essential roles, both chemokines and chemokine receptors play critical roles in viral infection and replication. Not only are chemokine receptors key components of the receptor/fusion complexes of primate immunodeficiency viruses, but chemokines can also influence virus entry and infection. Many viruses, in particular herpesviruses, encode chemokines and chemokine receptors that influence the replication of both the parent virus and other unrelated viruses. The cell surface expression of the chemokine receptors is regulated through their interaction with membrane trafficking pathways. Ligands induce receptor internalization and downmodulation through endocytosis, and recycling is regulated within endosomes. Part of the mechanism through which chemokines protect cells from HIV infection is through ligand-induced internalization of the specific chemokine receptor co-receptors. In addition, mechanisms may exist to regulate the trafficking of newly synthesized receptors to the cell surface. Here we discuss aspects of the mechanisms through which chemokine receptors interact with membrane-trafficking pathways and the influence of these interactions on viral replication.
Collapse
Affiliation(s)
- A Pelchen-Matthews
- MRC Laboratory for Molecular Cell Biology, University College London, UK
| | | | | | | | | |
Collapse
|
86
|
Metalloproteinases Are Involved in Lipopolysaccharide– and Tumor Necrosis Factor-–Mediated Regulation of CXCR1 and CXCR2 Chemokine Receptor Expression. Blood 1999. [DOI: 10.1182/blood.v93.7.2173] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe neutrophil-specific G-protein–coupled chemokine receptors, CXCR1 and CXCR2, bind with high affinity to the potent chemoattractant interleukin-8 (IL-8). The mechanisms of IL-8 receptor regulation are not well defined, although previous studies have suggested a process of ligand-promoted internalization as a putative regulatory pathway. Herein, we provide evidence for two distinct processes of CXCR1 and CXCR2 regulation. Confocal microscopy data showed a redistribution of CXCR1 expression from the cell surface of neutrophils to internal compartments after stimulation with IL-8, whereas stimulation with bacterial lipopolysaccharide (LPS) or tumor necrosis factor- (TNF-) did not induce CXCR1 internalization but instead mediated a significant loss of membrane-proximal CXCR1 staining intensity. To investigate whether proteolytic cleavage was the mechanism responsible for LPS- and TNF-–induced downmodulation of IL-8 receptors, we tested a panel of proteinase inhibitors. The downmodulation of CXCR1 and CXCR2 by LPS and TNF- was most dramatically inhibited by metalloproteinase inhibitors; 1,10-phenanthroline and EDTA significantly attenuated LPS- and TNF-–induced loss of CXCR1 and CXCR2 cell surface expression. Metalloproteinase inhibitors also blocked the release of CXCR1 cleavage fragments into the cell supernatants of LPS- and TNF-–stimulated neutrophils. In addition, while treatment of neutrophils with LPS and TNF- inhibited IL-8 receptor–mediated calcium mobilization and IL-8–directed neutrophil chemotaxis, both 1,10-phenanthroline and EDTA blocked these inhibitory processes. In contrast, metalloproteinase inhibitors did not affect IL-8–mediated downmodulation of CXCR1 and CXCR2 cell surface expression or receptor signaling. Thus, these findings may provide further insight into the mechanisms of leukocyte regulation during immunologic and inflammatory responses.
Collapse
|
87
|
Metalloproteinases Are Involved in Lipopolysaccharide– and Tumor Necrosis Factor-–Mediated Regulation of CXCR1 and CXCR2 Chemokine Receptor Expression. Blood 1999. [DOI: 10.1182/blood.v93.7.2173.407a06_2173_2185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The neutrophil-specific G-protein–coupled chemokine receptors, CXCR1 and CXCR2, bind with high affinity to the potent chemoattractant interleukin-8 (IL-8). The mechanisms of IL-8 receptor regulation are not well defined, although previous studies have suggested a process of ligand-promoted internalization as a putative regulatory pathway. Herein, we provide evidence for two distinct processes of CXCR1 and CXCR2 regulation. Confocal microscopy data showed a redistribution of CXCR1 expression from the cell surface of neutrophils to internal compartments after stimulation with IL-8, whereas stimulation with bacterial lipopolysaccharide (LPS) or tumor necrosis factor- (TNF-) did not induce CXCR1 internalization but instead mediated a significant loss of membrane-proximal CXCR1 staining intensity. To investigate whether proteolytic cleavage was the mechanism responsible for LPS- and TNF-–induced downmodulation of IL-8 receptors, we tested a panel of proteinase inhibitors. The downmodulation of CXCR1 and CXCR2 by LPS and TNF- was most dramatically inhibited by metalloproteinase inhibitors; 1,10-phenanthroline and EDTA significantly attenuated LPS- and TNF-–induced loss of CXCR1 and CXCR2 cell surface expression. Metalloproteinase inhibitors also blocked the release of CXCR1 cleavage fragments into the cell supernatants of LPS- and TNF-–stimulated neutrophils. In addition, while treatment of neutrophils with LPS and TNF- inhibited IL-8 receptor–mediated calcium mobilization and IL-8–directed neutrophil chemotaxis, both 1,10-phenanthroline and EDTA blocked these inhibitory processes. In contrast, metalloproteinase inhibitors did not affect IL-8–mediated downmodulation of CXCR1 and CXCR2 cell surface expression or receptor signaling. Thus, these findings may provide further insight into the mechanisms of leukocyte regulation during immunologic and inflammatory responses.
Collapse
|
88
|
Meddows-Taylor S, Martin DJ, Tiemessen CT. Dysregulated production of interleukin-8 in individuals infected with human immunodeficiency virus type 1 and Mycobacterium tuberculosis. Infect Immun 1999; 67:1251-60. [PMID: 10024568 PMCID: PMC96454 DOI: 10.1128/iai.67.3.1251-1260.1999] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/1998] [Accepted: 11/20/1998] [Indexed: 11/20/2022] Open
Abstract
Interleukin-8 (IL-8) production in vivo was monitored in four study groups: normal blood donors, patients with pulmonary tuberculosis (TB), patients with human immunodeficiency virus type 1 (HIV-1) infection, and dually infected (HIV/TB) patients. We show that whereas there was evidence of detectable levels of cell-associated IL-8 (mRNA and protein) in peripheral cells of healthy individuals, this was largely lost in the disease states studied. Coupled with this finding was significantly increased circulating levels of IL-8 in HIV-1-infected individuals with or without concomitant pulmonary TB (P < 0.001). On the other hand, the capacity of peripheral mononuclear cells to produce IL-8 spontaneously ex vivo was enhanced in HIV-1 and TB patients (P < 0.05) and many of the HIV/TB group, but their corresponding capacities to respond to various stimuli, in particular phytohemagglutinin, were significantly diminished compared to those of normal donors (P < 0.05). Circulating levels of IL-8 in a group of HIV/TB patients were significantly positively correlated with the percentage of polymorphonuclear leukocytes (PMN) in the peripheral circulation (r = 0.65; P = 0.01), the proportions of IL-8 receptor A (IL-8RA)-expressing (r = 0.86; P < 0.01) and IL-8RB-expressing (r = 0.77; P < 0.01) PMN, and the capacity of PMN to migrate in response to IL-8 as chemoattractant (r = 0.68; P < 0. 01). IL-8RB fluorescence intensity, however, was negatively correlated with plasma IL-8 levels (r = -0.73; P < 0.01). Our results suggest that altered regulation of IL-8 in HIV-1 may have important implications for antimicrobial defenses and for normal immune processes.
Collapse
Affiliation(s)
- S Meddows-Taylor
- MRC AIDS Virus Research Unit, National Institute for Virology, and Department of Virology, University of the Witwatersrand, Johannesburg, South Africa
| | | | | |
Collapse
|
89
|
Cummings CJ, Martin TR, Frevert CW, Quan JM, Wong VA, Mongovin SM, Hagen TR, Steinberg KP, Goodman RB. Expression and Function of the Chemokine Receptors CXCR1 and CXCR2 in Sepsis. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.4.2341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Neutrophils (polymorphonuclear neutrophils; PMN) and a redundant system of chemotactic cytokines (chemokines) have been implicated in the pathogenesis of the acute respiratory distress syndrome in patients with sepsis. PMN express two cell surface receptors for the CXC chemokines, CXCR1 and CXCR2. We investigated the expression and function of these receptors in patients with severe sepsis. Compared with normal donors, CXCR2 surface expression was down-regulated by 50% on PMN from septic patients (p < 0.005), while CXCR1 expression persisted. In vitro migratory responses to the CXCR1 ligand, IL-8, were similar in PMN from septic patients and normal donors. By contrast, the migratory response to the CXCR2 ligands, epithelial cell-derived neutrophil activator (ENA-78) and the growth-related oncogene proteins, was markedly suppressed in PMN from septic patients (p < 0.05). Ab specific for CXCR1 blocked in vitro migration of PMN from septic patients to IL-8 (p < 0.05), but not to FMLP. Thus, functionally significant down-regulation of CXCR2 occurs on PMN in septic patients. We conclude that in a complex milieu of multiple CXC chemokines, CXCR1 functions as the single dominant CXC chemokine receptor in patients with sepsis. These observations offer a potential strategy for attenuating adverse inflammation in sepsis while preserving host defenses mediated by bacteria-derived peptides such as FMLP.
Collapse
Affiliation(s)
- C. James Cummings
- Medical Research Service, Seattle Veterans Affairs and Harborview Medical Centers, and Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195
| | - Thomas R. Martin
- Medical Research Service, Seattle Veterans Affairs and Harborview Medical Centers, and Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195
| | - Charles W. Frevert
- Medical Research Service, Seattle Veterans Affairs and Harborview Medical Centers, and Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195
| | - Joanne M. Quan
- Medical Research Service, Seattle Veterans Affairs and Harborview Medical Centers, and Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195
| | - Venus A. Wong
- Medical Research Service, Seattle Veterans Affairs and Harborview Medical Centers, and Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195
| | - Steven M. Mongovin
- Medical Research Service, Seattle Veterans Affairs and Harborview Medical Centers, and Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195
| | - Tonja R. Hagen
- Medical Research Service, Seattle Veterans Affairs and Harborview Medical Centers, and Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195
| | - Kenneth P. Steinberg
- Medical Research Service, Seattle Veterans Affairs and Harborview Medical Centers, and Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195
| | - Richard B. Goodman
- Medical Research Service, Seattle Veterans Affairs and Harborview Medical Centers, and Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195
| |
Collapse
|
90
|
The Biologic Role of Interleukin-8: Functional Analysis and Expression of CXCR1 and CXCR2 on Human Eosinophils. Blood 1999. [DOI: 10.1182/blood.v93.2.694.402k31_694_702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Chemokines play an important role in attracting granulocytes into sites of inflammation. Two chemokine subfamilies differ in their biologic activity for different granulocyte subsets. Whereas CXC chemokines such as interleukin-8 (IL-8) activate predominantly neutrophils, CC chemokines such as RANTES and eotaxin activate predominantly eosinophils. However, controversial results have been published in the past regarding the biologic role of IL-8 in eosinophil activation, particularly in allergic diseases. In this study, we investigated the functional evidence and expression of both IL-8 receptors, CXCR1 and CXCR2, on highly purified human eosinophils. In the first set of experiments, a chemotaxis assay was performed showing that IL-8 did not induce chemotaxis of eosinophils. In addition, and in contrast to neutrophils and lymphocytes, IL-8 did not induce a rapid and transient release of cytosolic free Ca2+([Ca2+]i) in eosinophils, even after preincubation with TH1- and TH2-like cytokines. To investigate whether neutrophil contamination might be responsible for the reported IL-8 effects on eosinophils, neutrophils were added to highly purified eosinophils from the same donor in different concentrations. Interestingly, as little as 5% of neutrophil contamination was sufficient to induce an increase of [Ca2+]iafter stimulation with IL-8. Flow cytometry experiments with monoclonal antibodies against both IL-8 receptors demonstrated no expression of CXCR1 and CXCR2 on eosinophils before or after cytokine activation. Reverse transcriptase-polymerase chain reaction experiments showed that eosinophils, in contrast to neutrophils and lymphocytes, did not express mRNA for CXCR1 and CXCR2. In summary, this study clearly demonstrates that CXCR1 and CXCR2 are not expressed on human eosinophils, even after priming with different bioactive cytokines. Because the CXC chemokine IL-8 did not induce in vitro effects on human eosinophils, IL-8 may also not contribute in vivo to the influx of eosinophil granulocytes into sites of allergic inflammation. Our results suggest that CC chemokines such as eotaxin, eotaxin-2, and MCP-4 are predominant for the activation of eosinophils.
Collapse
|
91
|
The Biologic Role of Interleukin-8: Functional Analysis and Expression of CXCR1 and CXCR2 on Human Eosinophils. Blood 1999. [DOI: 10.1182/blood.v93.2.694] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractChemokines play an important role in attracting granulocytes into sites of inflammation. Two chemokine subfamilies differ in their biologic activity for different granulocyte subsets. Whereas CXC chemokines such as interleukin-8 (IL-8) activate predominantly neutrophils, CC chemokines such as RANTES and eotaxin activate predominantly eosinophils. However, controversial results have been published in the past regarding the biologic role of IL-8 in eosinophil activation, particularly in allergic diseases. In this study, we investigated the functional evidence and expression of both IL-8 receptors, CXCR1 and CXCR2, on highly purified human eosinophils. In the first set of experiments, a chemotaxis assay was performed showing that IL-8 did not induce chemotaxis of eosinophils. In addition, and in contrast to neutrophils and lymphocytes, IL-8 did not induce a rapid and transient release of cytosolic free Ca2+([Ca2+]i) in eosinophils, even after preincubation with TH1- and TH2-like cytokines. To investigate whether neutrophil contamination might be responsible for the reported IL-8 effects on eosinophils, neutrophils were added to highly purified eosinophils from the same donor in different concentrations. Interestingly, as little as 5% of neutrophil contamination was sufficient to induce an increase of [Ca2+]iafter stimulation with IL-8. Flow cytometry experiments with monoclonal antibodies against both IL-8 receptors demonstrated no expression of CXCR1 and CXCR2 on eosinophils before or after cytokine activation. Reverse transcriptase-polymerase chain reaction experiments showed that eosinophils, in contrast to neutrophils and lymphocytes, did not express mRNA for CXCR1 and CXCR2. In summary, this study clearly demonstrates that CXCR1 and CXCR2 are not expressed on human eosinophils, even after priming with different bioactive cytokines. Because the CXC chemokine IL-8 did not induce in vitro effects on human eosinophils, IL-8 may also not contribute in vivo to the influx of eosinophil granulocytes into sites of allergic inflammation. Our results suggest that CC chemokines such as eotaxin, eotaxin-2, and MCP-4 are predominant for the activation of eosinophils.
Collapse
|
92
|
Jones SA, Horiuchi S, Novick D, Yamamoto N, Fuller GM. Shedding of the soluble IL-6 receptor is triggered by Ca2+ mobilization, while basal release is predominantly the product of differential mRNA splicing in THP-1 cells. Eur J Immunol 1998; 28:3514-22. [PMID: 9842894 DOI: 10.1002/(sici)1521-4141(199811)28:11<3514::aid-immu3514>3.0.co;2-t] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The soluble IL-6 receptor (sIL-6R) is generated through either proteolytic shedding of the cognate receptor (PC-sIL-6R), or released as the product of differential mRNA splicing (DS-sIL-6R). Using monocytic THP-1 cells, we demonstrate that both mechanisms are independently regulated, and that each process contributes to sIL-6R production. Shedding of the IL-6R was activated by the Ca2+ ionophore, ionomycin, and inhibited by the TNF-alpha protease inhibitor (TAPI). In contrast, basal sIL-6R release was unaffected by Ca2+ depletion and largely insensitive to TAPI. Moreover, although IL-6R shedding was inactivated by serum starvation, non-stimulated production remained intact. Basal sIL-6R production via differential mRNA splicing was shown through the inhibitory action of brefeldin A and an enzyme-linked immunosorbent assay specific for DS-sIL-6R. Release of this isoform was unaffected by ionomycin or TAPI, indicating that Ca2+ mobilization activates PC-sIL-6R generation, but not DS-sIL-6R. The divergent control of these sIL-6R isoforms indicates that they may independently influence the inflammatory response.
Collapse
Affiliation(s)
- S A Jones
- Department of Cell Biology, University of Alabama at Birmingham, 35233-0005, USA.
| | | | | | | | | |
Collapse
|
93
|
Bodaghi B, Jones TR, Zipeto D, Vita C, Sun L, Laurent L, Arenzana-Seisdedos F, Virelizier JL, Michelson S. Chemokine sequestration by viral chemoreceptors as a novel viral escape strategy: withdrawal of chemokines from the environment of cytomegalovirus-infected cells. J Exp Med 1998; 188:855-66. [PMID: 9730887 PMCID: PMC2213390 DOI: 10.1084/jem.188.5.855] [Citation(s) in RCA: 247] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Human cytomegalovirus (HCMV), a betaherpesvirus, has developed several ways to evade the immune system, notably downregulation of cell surface expression of major histocompatibility complex class I heavy chains. Here we report that HCMV has devised another means to compromise immune surveillance mechanisms. Extracellular accumulation of both constitutively produced monocyte chemoattractant protein (MCP)-1 and tumor necrosis factor-superinduced RANTES (regulated on activation, normal T cell expressed and secreted) was downregulated in HCMV-infected fibroblasts in the absence of transcriptional repression or the expression of polyadenylated RNA for the cellular chemokine receptors CCR-1, CCR-3, and CCR-5. Competitive binding experiments demonstrated that HCMV-infected cells bind RANTES, MCP-1, macrophage inflammatory protein (MIP)-1beta, and MCP-3, but not MCP-2, to the same receptor as does MIP-1alpha, which is not expressed in uninfected cells. HCMV encodes three proteins with homology to CC chemokine receptors: US27, US28, and UL33. Cells infected with HCMV mutants deleted of US28, or both US27 and US28 genes, failed to downregulate extracellular accumulation of either RANTES or MCP-1. In contrast, cells infected with a mutant deleted of US27 continues to bind and downregulate those chemokines. Depletion of chemokines from the culture medium was at least partially due to continuous internalization of extracellular chemokine, since exogenously added, biotinylated RANTES accumulated in HCMV-infected cells. Thus, HCMV can modify the chemokine environment of infected cells through intense sequestering of CC chemokines, mediated principally by expression of the US28-encoded chemokine receptor.
Collapse
Affiliation(s)
- B Bodaghi
- Unite d'Immunologie Virale, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Khandaker MH, Xu L, Rahimpour R, Mitchell G, DeVries ME, Pickering JG, Singhal SK, Feldman RD, Kelvin DJ. CXCR1 and CXCR2 Are Rapidly Down-Modulated by Bacterial Endotoxin Through a Unique Agonist-Independent, Tyrosine Kinase-Dependent Mechanism. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.4.1930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
The expression of the seven-transmembrane domain chemokine receptors CXCR1 and CXCR2 modulates neutrophil responsiveness to the chemoattractant IL-8 and a number of closely related CXC chemokines. In the present study, we investigated the mechanism by which bacterial LPS induces the down-modulation of IL-8 responsiveness and CXCR1 and CXCR2 expression on human neutrophils. Treating neutrophils with LPS reduced IL-8R expression to 55 ± 5% of the control within 30 min and to 23 ± 2% within 1 h of stimulation. Furthermore, this down-modulation could not be attributed to increased concentrations of IL-8, TNF-α, or IL-1β, since ELISA studies indicated that LPS-stimulated neutrophils did not release detectable amounts of these proteins before 2 h poststimulation. The tyrosine kinase (TK) inhibitors genistein and herbimycin A attenuated the LPS-mediated down-modulation of CXCR1 and CXCR2, indicating that the activation of a TK is required for LPS to mediate its effect. The effect of LPS on receptor expression paralleled the hyperphosphorylation of the protein TK p72syk. Although IL-8 induced a comparable down-modulation of CXCR1 and CXCR2, TK inhibitors did not attenuate this effect. These studies provide the first evidence of an agonist-independent, TK-dependent pathway of chemokine receptor regulation by endotoxin.
Collapse
Affiliation(s)
- Masud H. Khandaker
- *Departments of Microbiology and Immunology and
- ‡Laboratory of Molecular Immunology and Inflammation, John P. Robarts Research Institute, London, Ontario, Canada
| | - Luoling Xu
- ‡Laboratory of Molecular Immunology and Inflammation, John P. Robarts Research Institute, London, Ontario, Canada
| | - Rahbar Rahimpour
- *Departments of Microbiology and Immunology and
- ‡Laboratory of Molecular Immunology and Inflammation, John P. Robarts Research Institute, London, Ontario, Canada
| | - Gordon Mitchell
- ‡Laboratory of Molecular Immunology and Inflammation, John P. Robarts Research Institute, London, Ontario, Canada
| | - Mark E. DeVries
- *Departments of Microbiology and Immunology and
- ‡Laboratory of Molecular Immunology and Inflammation, John P. Robarts Research Institute, London, Ontario, Canada
| | - J. Geoffrey Pickering
- ‡Laboratory of Molecular Immunology and Inflammation, John P. Robarts Research Institute, London, Ontario, Canada
| | | | - Ross D. Feldman
- †Medicine, University of Western Ontario, London, Ontario, Canada; and
| | - David J. Kelvin
- *Departments of Microbiology and Immunology and
- ‡Laboratory of Molecular Immunology and Inflammation, John P. Robarts Research Institute, London, Ontario, Canada
| |
Collapse
|
95
|
Compton SJ, Cairns JA, Holgate ST, Walls AF. The Role of Mast Cell Tryptase in Regulating Endothelial Cell Proliferation, Cytokine Release, and Adhesion Molecule Expression: Tryptase Induces Expression of mRNA for IL-1β and IL-8 and Stimulates the Selective Release of IL-8 from Human Umbilical Vein Endothelial Cells. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.4.1939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Mast cells are found frequently in close proximity to blood vessels, and endothelial cells are likely to be exposed to high concentrations of their granule mediators. We have investigated the proinflammatory actions of the major mast cell product tryptase on HUVEC. Addition of purified tryptase was found to stimulate thymidine incorporation, but induced little alteration in cell numbers, suggesting it is not a growth factor for HUVEC. Expression of ICAM-1, VCAM-1, and E-selectin was not altered following incubation with tryptase, but the potent granulocyte chemoattractant IL-8 was released in a dose-dependent fashion in response to physiologically relevant concentrations, with maximal levels in supernatants after 24 h. The actions of tryptase on HUVEC were inhibited by heat inactivation of the enzyme, or by preincubating with the protease inhibitors leupeptin or benzamidine, suggesting a requirement for an intact catalytic site. Reverse-transcription PCR analysis indicated up-regulation of mRNA for IL-8 as well as for IL-1β in response to tryptase or TNF-α. However, tryptase was a more selective stimulus than TNF-α and did not induce increased expression of mRNA for granulocyte-macrophage CSF or stimulate the release of this cytokine. Leukocyte accumulation in response to tryptase may be mediated in part through the selective secretion of IL-8 from endothelial cells.
Collapse
Affiliation(s)
- Steven J. Compton
- Immunopharmacology Group, Southampton General Hospital, Southampton, United Kingdom
| | - Jennifer A. Cairns
- Immunopharmacology Group, Southampton General Hospital, Southampton, United Kingdom
| | - Stephen T. Holgate
- Immunopharmacology Group, Southampton General Hospital, Southampton, United Kingdom
| | - Andrew F. Walls
- Immunopharmacology Group, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
96
|
Ramdin L, Perks B, Sheron N, Shute JK. Regulation of interleukin-8 binding and function by heparin and alpha2-macroglobulin. Clin Exp Allergy 1998; 28:616-24. [PMID: 9645599 DOI: 10.1046/j.1365-2222.1998.00283.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Increased expression of interleukin-8 (IL-8), a potent neutrophil chemoattractant, is associated with a number of inflammatory diseases. Interleukin-8 binds to the glycosaminoglycan (GAG) heparin and the protease inhibitor alpha2-macroglobulin, molecules which regulate the function of a number of cytokines. Heparan sulphate was previously shown to enhance neutrophil chemotactic responses to IL-8. OBJECTIVE The purpose of this study was to investigate the effect of heparin, heparan sulphate and alpha2-macroglobulin on IL-8 binding to neutrophils and subsequent functional effects in vitro. METHODS The binding of 125I-IL-8 to normal neutrophils at 4 degrees C was studied and the IL-8 induced neutrophil chemotactic response was investigated using micro-Boyden chambers. Complexation of IL-8 with alpha2-macroglobulin was confirmed using gel filtration chromatography. RESULTS Heparin, but not heparan sulphate, inhibited the binding of 125I-IL-8 to neutrophils (IC50=26 microg/mL) and IL-8 induced neutrophil chemotactic responses (IC50=4 microg/mL). The specific inhibitory effect of heparin was apparently due to an interaction with IL-8 which was charge-dependent, since dextran sulphate had a greater inhibitory effect on chemotactic responses (IC50=2 microg/mL) and FITC-heparin did not bind to neutrophils. The heparin-induced inhibition of IL-8 binding and chemotactic responses was reversed in a dose-dependent manner in the presence of alpha2-macroglobulin. The binding of 125I-IL-8 to neutrophils in the presence of alpha2-macroglobulin appears to be, in part, through the specific IL-8 receptor. CONCLUSION These results point to an anti-inflammatory role for heparin and a novel, potentially, pro-inflammatory role for alpha2-macroglobulin which together indicate the importance of cytokine-binding macromolecules in determining net cytokine function.
Collapse
Affiliation(s)
- L Ramdin
- University Medicine, Southampton General Hospital, UK
| | | | | | | |
Collapse
|
97
|
Reduced Ex Vivo Interleukin-8 Production by Neutrophils in Septic and Nonseptic Systemic Inflammatory Response Syndrome. Blood 1998. [DOI: 10.1182/blood.v91.9.3439.3439_3439_3446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ex vivo cytokine production by circulating lymphocytes and monocytes is reduced in patients with infectious or noninfectious systemic inflammatory response syndrome. Very few studies have addressed the reactivity of polymorphonuclear cells (PMN). To analyze further the relative contribution of systemic inflammatory response syndrome alone or in combination with infection we studied the interleukin-8 (IL-8) production by PMN isolated from patients who had undergone cardiac surgery with cardiopulmonary bypass (CPB) and patients with sepsis. Cells were activated with either lipopolysaccharide (LPS) or heat-killed streptococci. Compared with healthy controls, the release of IL-8 by PMN in both groups of patients was significantly reduced whether activated by LPS, independently of its concentration and origin, or by heat-killed streptococci. These observations suggest that stressful conditions related to inflammation, independently of infection, rapidly dampened the reactivity of circulating PMN. We investigated whether the observed diminished reactivity of PMN might reflect an endotoxin tolerance phenomenon. Our in vitro experiments with PMN from healthy controls indicated that PMN could not be rendered tolerant stricto sensu. However, our data suggested that LPS-induced mediators such as IL-10 may be responsible for the observed anergy in patients.
Collapse
|
98
|
Asagoe K, Yamamoto K, Takahashi A, Suzuki K, Maeda A, Nohgawa M, Harakawa N, Takano K, Mukaida N, Matsushima K, Okuma M, Sasada M. Down-Regulation of CXCR2 Expression on Human Polymorphonuclear Leukocytes by TNF-α. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.9.4518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
TNF-α is implicated in the initiation of cytokine cascades in various inflammatory settings. To assess the interactions of multiple cytokines at the level of inflammatory effector cells, we examined the effects of TNF-α on the expression of two IL-8Rs (CXCR1 and CXCR2) on polymorphonuclear leukocytes (PMNs). TNF-α decreased the surface expression of CXCR2 in a dose- and time-dependent manner. In contrast, CXCR1 expression was not affected by TNF-α. The release of CXCR2 into the supernatant of TNF-α-treated PMNs was detected by immunoblotting and immuno-slot-blot analyses, suggesting that the down-regulation of CXCR2 was caused mainly by shedding from the cell surface. The CXCR2 down-regulation was inhibited by PMSF and aprotinin, supporting the hypothesis that the shedding was mediated by serine protease(s). The intracellular Ca2+ mobilization and chemotaxis in response to IL-8 were suppressed by the pretreatment of PMNs with TNF-α, indicating that the decrease in CXCR2 was reflected in the decreased functional responses to IL-8. In contrast, the O2− release, which is mediated by CXCR1, was not suppressed by TNF-α. The treatment of whole blood with TNF-α also caused a significant reduction in CXCR2 and markedly suppressed intracellular Ca2+ mobilization and chemotaxis in response to IL-8, while enhancing the O2− release. These findings suggest that TNF-α down-regulates CXCR2 expression on PMNs and modulates IL-8-induced biologic responses, leading to the intravascular retention of PMNs with an enhanced production of reactive oxygen metabolites.
Collapse
Affiliation(s)
- Kohsuke Asagoe
- *Department of Hematology and Oncology, Clinical Sciences for Pathological Organs, Graduate School of Medicine, and
| | - Kokichi Yamamoto
- *Department of Hematology and Oncology, Clinical Sciences for Pathological Organs, Graduate School of Medicine, and
| | - Atsushi Takahashi
- *Department of Hematology and Oncology, Clinical Sciences for Pathological Organs, Graduate School of Medicine, and
| | - Kazuo Suzuki
- ‡National Institute of Infectious Diseases, Tokyo; and
| | - Akinori Maeda
- *Department of Hematology and Oncology, Clinical Sciences for Pathological Organs, Graduate School of Medicine, and
| | - Masaharu Nohgawa
- *Department of Hematology and Oncology, Clinical Sciences for Pathological Organs, Graduate School of Medicine, and
| | - Nari Harakawa
- *Department of Hematology and Oncology, Clinical Sciences for Pathological Organs, Graduate School of Medicine, and
| | - Kuniko Takano
- *Department of Hematology and Oncology, Clinical Sciences for Pathological Organs, Graduate School of Medicine, and
| | - Naofumi Mukaida
- §Department of Pharmacology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Kouji Matsushima
- §Department of Pharmacology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Minoru Okuma
- *Department of Hematology and Oncology, Clinical Sciences for Pathological Organs, Graduate School of Medicine, and
| | - Masataka Sasada
- *Department of Hematology and Oncology, Clinical Sciences for Pathological Organs, Graduate School of Medicine, and
- †College of Medical Technology, Kyoto University, Sakyo-ku, Kyoto
| |
Collapse
|
99
|
Reduced Ex Vivo Interleukin-8 Production by Neutrophils in Septic and Nonseptic Systemic Inflammatory Response Syndrome. Blood 1998. [DOI: 10.1182/blood.v91.9.3439] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractEx vivo cytokine production by circulating lymphocytes and monocytes is reduced in patients with infectious or noninfectious systemic inflammatory response syndrome. Very few studies have addressed the reactivity of polymorphonuclear cells (PMN). To analyze further the relative contribution of systemic inflammatory response syndrome alone or in combination with infection we studied the interleukin-8 (IL-8) production by PMN isolated from patients who had undergone cardiac surgery with cardiopulmonary bypass (CPB) and patients with sepsis. Cells were activated with either lipopolysaccharide (LPS) or heat-killed streptococci. Compared with healthy controls, the release of IL-8 by PMN in both groups of patients was significantly reduced whether activated by LPS, independently of its concentration and origin, or by heat-killed streptococci. These observations suggest that stressful conditions related to inflammation, independently of infection, rapidly dampened the reactivity of circulating PMN. We investigated whether the observed diminished reactivity of PMN might reflect an endotoxin tolerance phenomenon. Our in vitro experiments with PMN from healthy controls indicated that PMN could not be rendered tolerant stricto sensu. However, our data suggested that LPS-induced mediators such as IL-10 may be responsible for the observed anergy in patients.
Collapse
|
100
|
Hosie MJ, Broere N, Hesselgesser J, Turner JD, Hoxie JA, Neil JC, Willett BJ. Modulation of feline immunodeficiency virus infection by stromal cell-derived factor. J Virol 1998; 72:2097-104. [PMID: 9499065 PMCID: PMC109504 DOI: 10.1128/jvi.72.3.2097-2104.1998] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/1997] [Accepted: 11/19/1997] [Indexed: 02/06/2023] Open
Abstract
The alpha-chemokine receptor CXCR4 has recently been shown to support syncytium formation mediated by strains of feline immunodeficiency virus (FIV) that have been selected for growth in the Crandell feline kidney cell line (CrFK-tropic virus). Given that both human and feline CXCR4 support syncytium formation mediated by FIV, we investigated whether human stromal cell-derived factor (SDF-1) would inhibit infection with FIV. Human SDF-1alpha and SDF-1beta bound with a high affinity (K(D)s of 12.0 and 10.4 nM, respectively) to human cells stably expressing feline CXCR4, and treatment of CrFK cells with human SDF-1alpha resulted in a dose-dependent inhibition of infection by FIV(PET). No inhibitory activity was detected when the interleukin-2 (IL-2)-dependent feline T-cell line Mya-1 was used in place of CrFK cells, suggesting the existence of a CXCR4-independent mechanism of infection. Furthermore, neither the human beta-chemokines RANTES, MIP-1alpha, MIP-1beta, and MCP-1 nor the alpha-chemokine IL-8 had an effect on infection of either CrFK or Mya-1 cells with CrFK-tropic virus. Envelope glycoprotein purified from CrFK-tropic virus competed specifically for binding of SDF-1alpha to feline CXCR4 and CXCR4 expression was reduced in FIV-infected cells, suggesting that the inhibitory activity of SDF-1alpha in CrFK cells may be the result of steric hindrance of the virus-receptor interaction following the interaction between SDF and CXCR4. Prolonged incubation of CrFK cells with SDF-1alpha led to an enhancement rather than an inhibition of infection. Flow cytometric analysis revealed that this effect may be due largely to up-regulation of CXCR4 expression by SDF-1alpha on CrFK cells, an effect mimicked by treatment of the cells with phorbol myristate acetate. The data suggest that infection of feline cells with FIV can be mediated by CXCR4 and that, depending on the assay conditions, infection can be either inhibited or enhanced by SDF-1alpha. Infection with FIV may therefore prove a valuable model in which to study the development of novel therapeutic interventions for the treatment of AIDS.
Collapse
Affiliation(s)
- M J Hosie
- Department of Veterinary Pathology, University of Glasgow Veterinary School, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|