Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Clin Exp Allergy ; 49(2): 190-198, 2019 02.
Article in English | MEDLINE | ID: mdl-30295352

ABSTRACT

BACKGROUND: Eosinophil-associated RNases (EARs) are stored preformed in eosinophil cytoplasmic secretory granules and have a key role in eosinophil effector functions in host defence and inflammatory disorders. However, the secretion mechanisms of EARs are poorly understood. OBJECTIVE: Our study aimed to understand the involvement of cytoskeleton machinery in EAR secretion. METHODS: Fresh human and mouse eosinophils were stimulated with CCL11, and the secretion of enzymatically active EARs was detected using an RNase activity assay. The involvement of cytoskeletal elements or microtubules was probed using specific inhibitors. RESULTS: We found that dynamic polymerization of microtubules and cytoskeletal elements, such as Rho and Rac, is required for chemokine-mediated EAR secretion from human and mouse eosinophils. However, inhibition of ROCK (Rho-associated protein kinase) increased EAR secretion in human and mouse eosinophils even in the absence of chemokine stimulation, suggesting ROCK negatively regulates EAR secretion. CONCLUSIONS: Collectively, these data suggest a cytoskeleton-dependent mechanism of EAR secretion from eosinophils, findings that are pertinent to host defence, allergy and other eosinophil-associated diseases.


Subject(s)
Eosinophil Cationic Protein/immunology , Eosinophils/immunology , rac GTP-Binding Proteins/immunology , rho-Associated Kinases/immunology , Animals , Chemokine CCL11/genetics , Chemokine CCL11/immunology , Eosinophil Cationic Protein/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , rac GTP-Binding Proteins/genetics , rho-Associated Kinases/genetics
3.
J Immunol ; 201(10): 2998-3005, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30315138

ABSTRACT

CD300a is an inhibitory receptor for mast cells and eosinophils in allergic inflammation (AI); however, the spatiotemporal expression of CD300a and its potential roles in the resolution of AI are still to be determined. In this study, employing a mouse model of allergic peritonitis, we demonstrate that CD300a expression on peritoneal cells is regulated from inflammation to resolution. Allergic peritonitis-induced CD300a-/- mice had a rapid increase in their inflammatory cell infiltrates and tryptase content in the peritoneal cavity compared with wild type, and their resolution process was significantly delayed. CD300a-/- mice expressed lower levels of ALX/FPR2 receptor on peritoneal cells and had higher levels of LXA4 in the peritoneal lavage. CD300a activation on mouse bone marrow-derived mast cells regulated ALX/FPR2 expression levels following IgE-mediated activation. Together, these findings indicate a role for CD300a in AI and its resolution, in part via the specialized proresolving mediator LXA4 and ALX/FPR2 receptor pathway activation.


Subject(s)
Hypersensitivity/immunology , Inflammation/immunology , Leukocytes/immunology , Receptors, Immunologic/immunology , Animals , Female , Mice , Mice, Inbred BALB C , Mice, Knockout , Peritonitis/immunology
4.
Exp Cell Res ; 337(2): 129-135, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26254897

ABSTRACT

BACKGROUND: SNARE members mediate membrane fusion during intracellular trafficking underlying innate and adaptive immune responses by different cells. However, little is known about the expression and function of these proteins in human eosinophils, cells involved in allergic, inflammatory and immunoregulatory responses. Here, we investigate the expression and distribution of the Qa-SNARE syntaxin17 (STX17) within human eosinophils isolated from the peripheral blood. METHODS: Flow cytometry and a pre-embedding immunonanogold electron microscopy (EM) technique that combines optimal epitope preservation and secondary Fab-fragments of antibodies linked to 1.4 nm gold particles for optimal access to microdomains, were used to investigate STX17. RESULTS: STX17 was detected within unstimulated eosinophils. Immunogold EM revealed STX17 on secretory granules and on granule-derived vesiculotubular transport carriers (Eosinophil Sombrero Vesicles-EoSVs). Quantitative EM analyses showed that 77.7% of the granules were positive for STX17 with a mean±SEM of 3.9±0.2 gold particles/granule. Labeling was present on both granule outer membranes and matrices while EoSVs showed clear membrane-associated labeling. STX17 was also present in secretory granules in eosinophils stimulated with the cytokine tumor necrosis factor alpha (TNF-α) or the CC-chemokine ligand 11 CCL11 (eotaxin-1), stimuli that induce eosinophil degranulation. The number of secretory granules labeled for STX17 was significantly higher in CCL11 compared with the unstimulated group. The level of cell labeling did not change when unstimulated cells were compared with TNF-α-stimulated eosinophils. CONCLUSIONS: The present study clearly shows by immunanonogold EM that STX17 is localized in eosinophil secretory granules and transport vesicles and might be involved in the transport of granule-derived cargos.


Subject(s)
Cytokines/metabolism , Eosinophils/metabolism , Qa-SNARE Proteins/metabolism , Secretory Vesicles/metabolism , Cells, Cultured , Eosinophils/cytology , Flow Cytometry , Humans , Microscopy, Immunoelectron , Secretory Vesicles/ultrastructure , Subcellular Fractions
5.
J Histochem Cytochem ; 62(6): 450-459, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24670795

ABSTRACT

Protein disulfide isomerase (PDI) has fundamental roles in the oxidative folding of proteins in the endoplasmic reticulum (ER) of eukaryotic cells. The study of this molecule has been attracting considerable attention due to its association with other cell functions and human diseases. In leukocytes, such as neutrophils, PDI is involved with cell adhesion, signaling and inflammation. However, the expression of PDI in other leukocytes, such as eosinophils, important cells in inflammatory, allergic and immunomodulatory responses, remains to be defined. Here we used different approaches to investigate PDI expression within human eosinophils. Western blotting and flow cytometry demonstrated high PDI expression in both unstimulated and CCL11/eotaxin-1-stimulated eosinophils, with similar levels in both conditions. By using an immunogold electron microscopy technique that combines better epitope preservation and secondary Fab-fragments of antibodies linked to 1.4-nm gold particles for optimal access to microdomains, we identified different intracellular sites for PDI. In addition to predictable strong PDI labeling at the nuclear envelope, other unanticipated sites, such as secretory granules, lipid bodies and vesicles, including large transport vesicles (eosinophil sombrero vesicles), were also labeled. Thus, we provide the first identification of PDI in human eosinophils, suggesting that this molecule may have additional/specific functions in these leukocytes.

6.
Curr Protoc Immunol ; Chapter 7: 7.31.1-7.31.8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22855360

ABSTRACT

Highly purified eosinophils can be isolated from peripheral blood by negative selection using an antibody-based magnetic negative selection protocol. The basic protocol describes a sequential fractionation of peripheral blood in which CD16+ granulocytes are enriched first from whole blood, followed by isolation of eosinophils. This technique is easy to use, fast, and highly reproducible. Support protocols describe a staining methods that can be used to evaluate the purity of eosinophils and differentiation from other leukocyte populations.


Subject(s)
Cell Separation/instrumentation , Cell Separation/methods , Eosinophils/cytology , Granulocytes/cytology , Eosinophils/immunology , Granulocytes/immunology , Humans , Magnetics , Receptors, IgG/immunology , Reproducibility of Results , Staining and Labeling/methods
7.
FASEB J ; 26(5): 2084-93, 2012 May.
Article in English | MEDLINE | ID: mdl-22294786

ABSTRACT

Rapid secretion of eosinophil-associated RNases (EARs), such as the human eosinophilic cationic protein (ECP), from intracellular granules is central to the role of eosinophils in allergic diseases and host immunity. Our knowledge regarding allergic inflammation has advanced based on mouse experimental models. However, unlike human eosinophils, capacities of mouse eosinophils to secrete granule proteins have been controversial. To study mechanisms of mouse eosinophil secretion and EAR release, we combined an RNase assay of mouse EARs with ultrastructural studies. In vitro, mouse eosinophils stimulated with the chemokine eotaxin-1 (CCL11) secreted enzymatically active EARs (EC(50) 5 nM) by piecemeal degranulation. In vivo, in a mouse model of allergic airway inflammation, increased airway eosinophil infiltration (24-fold) correlated with secretion of active RNases (3-fold). Moreover, we found that eosinophilic inflammation in mice can involve eosinophil cytolysis and release of cell-free granules. Cell-free mouse eosinophil granules expressed functional CCR3 receptors and secreted their granule proteins, including EAR and eosinophil peroxidase in response to CCL11. Collectively, these data demonstrate chemokine-dependent secretion of EARs from both intact mouse eosinophils and their cell-free granules, findings pertinent to understanding the pathogenesis of eosinophil-associated diseases, in which EARs are key factors.


Subject(s)
Chemokine CCL11/pharmacology , Eosinophils/drug effects , Ribonucleases/metabolism , Animals , Cell-Free System , Eosinophils/enzymology , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Transgenic , Microscopy, Electron, Transmission
8.
Cell Tissue Res ; 343(1): 57-83, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21042920

ABSTRACT

Eosinophils are innate immune leukocytes found in relatively low numbers within the blood. Terminal effector functions of eosinophils, deriving from their capacity to release their content of tissue-destructive cationic proteins, have historically been considered primary effector mechanisms against specific parasites, and are likewise implicated in tissue damage accompanying allergic responses such as asthma. However, the past decade has seen dramatic advancements in the field of eosinophil immunobiology, revealing eosinophils to also be key participants in many other facets of innate immunity, from bridging innate and adaptive immune responses to orchestrating tissue remodeling events. Here, we review the multifaceted functions of eosinophils in innate immunity that are currently known, and discuss new avenues in this evolving story.


Subject(s)
Eosinophils/immunology , Immunity, Innate/immunology , Animals , Disease , Health , Humans , Receptors, Immunologic/metabolism
9.
J Immunol ; 180(5): 2815-23, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18292502

ABSTRACT

Chemokines presented on endothelial tissues instantaneously trigger LFA-1-mediated arrest on ICAM-1 via rapid inside-out and outside-in (ligand-driven) LFA-1 activation. The GTPase RhoA was previously implicated in CCL21-triggered LFA-1 affinity triggering in murine T lymphocytes and in LFA-1-dependent adhesion strengthening to ICAM-1 on Peyer's patch high endothelial venules stabilized over periods of at least 10 s. In this study, we show that a specific RhoA 23/40 effector region is vital for the initial LFA-1-dependent adhesions of lymphocytes on high endothelial venules lasting 1-3 s. Blocking the RhoA 23/40 region in human T lymphocytes in vitro also impaired the subsecond CXCL12-triggered LFA-1-mediated T cell arrest on ICAM-1 by eliminating the rapid induction of an extended LFA-1 conformational state. However, the inflammatory chemokine CXCL9 triggered robust LFA-1-mediated T lymphocyte adhesion to ICAM-1 at subsecond contacts independently of the RhoA 23/40 region. CXCL9 did not induce conformational changes in the LFA-1 ectodomain, suggesting that particular chemokines can activate LFA-1 through outside-in post ligand binding stabilization changes. Like CXCL9, the potent diacylglycerol-dependent protein kinase C agonist PMA was found to trigger LFA-1 adhesiveness to ICAM-1 also without inducing integrin extension or an a priori clustering and independently of the RhoA 23/40 region. Our results collectively suggest that the 23/40 region of RhoA regulates chemokine-induced inside-out LFA-1 extension before ligand binding, but is not required for a variety of chemokine and non-chemokine signals that rapidly strengthen LFA-1-ICAM-1 bonds without an a priori induction of high-affinity extended LFA-1 conformations.


Subject(s)
Chemokine CXCL12/physiology , Chemokine CXCL9/physiology , Lymphocyte Function-Associated Antigen-1/metabolism , rhoA GTP-Binding Protein/physiology , Animals , Cell Adhesion/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Homeostasis/immunology , Humans , Intercellular Adhesion Molecule-1/metabolism , Leukocyte Rolling/immunology , Lymphocyte Activation/immunology , Lymphocyte Function-Associated Antigen-1/physiology , Mice , Mice, Inbred BALB C , Peyer's Patches/blood supply , Peyer's Patches/cytology , Peyer's Patches/immunology , Protein Structure, Tertiary
10.
J Immunol ; 179(2): 1030-8, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17617595

ABSTRACT

Cholesterol-enriched lipid microdomains regulate L-selectin signaling, but the role of membrane cholesterol in L-selectin adhesion is unclear. Arrest chemokines are a subset of endothelial chemokines that rapidly activate leukocyte integrin adhesiveness under shear flow. In the absence of integrin ligands, these chemokines destabilize L-selectin-mediated leukocyte rolling. In the present study, we investigated how cholesterol extraction from the plasma membrane of peripheral blood T or B cells affects L-selectin adhesions and their destabilization by arrest chemokines. Unlike the Jurkat T cell line, whose L-selectin-mediated adhesion is cholesterol dependent, in primary human PBLs and in murine B cells and B cell lines, cholesterol depletion did not impair any intrinsic adhesiveness of L-selectin, consistent with low selectin partitioning into lipid rafts in these cells. However, cholesterol raft disruption impaired the ability of two arrest chemokines, CXCL12 and CXCL13, but not of a third arrest chemokine, CCL21, to destabilize L-selectin-mediated rolling of T lymphocytes. Actin capping by brief incubation with cytochalasin D impaired the ability of all three chemokines to destabilize L-selectin rolling. Blocking of the actin regulatory phosphatidylinositol lipid, phosphatidylinositol 4,5-bisphosphate, did not affect chemokine-mediated destabilization of L-selectin adhesions. Collectively, our results suggest that L-selectin adhesions are inhibited by actin-associated, cholesterol-stabilized assemblies of CXCL12- and CXCL13-binding receptors on both T and B lymphocytes. Thus, the regulation of L-selectin by cholesterol-enriched microdomains varies with the cell type as well as with the identity of the destabilizing chemokine.


Subject(s)
Cell Adhesion/physiology , Chemokines/metabolism , Cholesterol/metabolism , L-Selectin/metabolism , Leukocyte Rolling/physiology , Membrane Microdomains/metabolism , Actins/metabolism , Chemokine CXCL12 , Chemokine CXCL13 , Chemokines/immunology , Chemokines, CXC/immunology , Chemokines, CXC/metabolism , Cholesterol/immunology , Flow Cytometry , Fluorescent Antibody Technique , Humans , L-Selectin/immunology , Lymphocytes/chemistry , Lymphocytes/immunology , Lymphocytes/metabolism , Membrane Microdomains/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Microscopy, Fluorescence
11.
Nat Immunol ; 6(5): 497-506, 2005 May.
Article in English | MEDLINE | ID: mdl-15834409

ABSTRACT

It is widely believed that rolling lymphocytes require successive chemokine-induced signaling for lymphocyte function-associated antigen 1 (LFA-1) to achieve a threshold avidity that will mediate lymphocyte arrest. Using an in vivo model of lymphocyte arrest, we show here that LFA-1-mediated arrest of lymphocytes rolling on high endothelial venules bearing LFA-1 ligands and chemokines was abrupt. In vitro flow chamber models showed that endothelium-presented but not soluble chemokines triggered instantaneous extension of bent LFA-1 in the absence of LFA-1 ligand engagement. To support lymphocyte adhesion, this extended LFA-1 conformation required immediate activation by its ligand, intercellular adhesion molecule 1. These data show that chemokine-triggered lymphocyte adhesiveness involves a previously unrecognized extension step that primes LFA-1 for ligand binding and firm adhesion.


Subject(s)
Chemokines/metabolism , Endothelium/metabolism , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , Allosteric Regulation , Cell Adhesion , Cells, Cultured , Chemokines/pharmacology , Cytoskeleton/metabolism , Epitopes/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocytes/drug effects , Lymphocytes/immunology , Protein Conformation/drug effects , Protein Subunits/immunology , Protein Subunits/metabolism , Solubility , Talin/metabolism
12.
J Immunol ; 173(12): 7282-91, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15585851

ABSTRACT

Lymphocyte transendothelial migration (TEM) is promoted by fluid shear signals and apical endothelial chemokines. Studying the role of these signals in neutrophil migration across differently activated HUVEC in a flow chamber apparatus, we gained new insights into how neutrophils integrate multiple endothelial signals to promote TEM. Neutrophils crossed highly activated HUVEC in a beta(2) integrin-dependent manner but independently of shear. In contrast, neutrophil migration across resting or moderately activated endothelium with low-level beta(2) integrin ligand activity was dramatically augmented by endothelial-presented chemoattractants, conditional to application of physiological shear stresses and intact beta(2) integrins. Shear stress signals were found to stimulate extensive neutrophil invaginations into the apical endothelial interface both before and during TEM. A subset of invaginating neutrophils completed transcellular diapedesis through individual endothelial cells within <1 min. Our results suggest that low-level occupancy of beta(2) integrins by adherent neutrophils can mediate TEM only if properly coupled to stimulatory shear stress and chemoattractant signals transduced at the apical neutrophil-endothelial interface.


Subject(s)
CD18 Antigens/metabolism , Chemotactic Factors/physiology , Chemotaxis, Leukocyte/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Neutrophils/ultrastructure , Signal Transduction/immunology , Cells, Cultured , Endothelium, Vascular/ultrastructure , Humans , Neutrophils/immunology , Neutrophils/metabolism , Platelet Activating Factor/physiology , Receptors, G-Protein-Coupled/physiology , Rheology , Stress, Mechanical , Tumor Necrosis Factor-alpha/pharmacology
13.
Blood ; 103(3): 1033-6, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14551137

ABSTRACT

Recently, we reported a rare leukocyte adhesion deficiency (LAD) associated with severe defects in integrin activation by chemokine signals, despite normal ligand binding of leukocyte integrins.(1) We now report that the small GTPase, Rap1, a key regulator of inside-out integrin activation is abnormally regulated in LAD Epstein-Barr virus (EBV) lymphocyte cells. Both constitutive and chemokine-triggered activation of Rap1 were abolished in LAD lymphocytes despite normal chemokine signaling. Nevertheless, Rap1 expression and activation by phorbol esters were intact, ruling out an LAD defect in Rap1 guanosine triphosphate (GTP) loading. The very late antigen 4 (VLA-4) integrin abnormally tethered LAD EBV lymphocytes to its ligand vascular cell adhesion molecule 1 (VCAM-1) under shear flow due to impaired generation of high-avidity contacts despite normal ligand binding and intact avidity to surface-bound anti-VLA-4 monoclonal antibody (mAb). Thus, a defect in constitutive Rap1 activation results in an inability of ligand-occupied integrins to generate high-avidity binding to ligand under shear flow. This is a first report of an inherited Rap1 activation defect associated with a pathologic disorder in leukocyte integrin function, we herein term it "LAD-III."


Subject(s)
Integrins/metabolism , Leukocyte-Adhesion Deficiency Syndrome/enzymology , Leukocyte-Adhesion Deficiency Syndrome/immunology , rap1 GTP-Binding Proteins/metabolism , Case-Control Studies , Cell Line, Transformed , Drug Stability , Enzyme Activation/genetics , Herpesvirus 4, Human , Humans , Integrin alpha4beta1/metabolism , Integrins/chemistry , Leukocyte-Adhesion Deficiency Syndrome/genetics , Vascular Cell Adhesion Molecule-1/metabolism
14.
J Biol Chem ; 278(51): 51203-12, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14532283

ABSTRACT

Leukocyte integrins must rapidly strengthen their binding to target endothelial sites to arrest rolling adhesions under physiological shear flow. We demonstrate that the integrin-associated tetraspanin, CD81, regulates VLA-4 and VLA-5 adhesion strengthening in monocytes and primary murine B cells. CD81 strengthens multivalent VLA-4 contacts within subsecond integrin occupancy without altering intrinsic adhesive properties to low density ligand. CD81 facilitates both VLA-4-mediated leukocyte rolling and arrest on VCAM-1 under shear flow as well as VLA-5-dependent adhesion to fibronectin during short stationary contacts. CD81 also augments VLA-4 avidity enhancement induced by either chemokine-stimulated Gi proteins or by protein kinase C activation, although it is not required for Gi protein or protein kinase C signaling activities. In contrast to other proadhesive integrin-associated proteins, CD81-promoted integrin adhesiveness does not require its own ligand occupancy or ligation. These results provide the first demonstration of an integrin-associated transmembranal protein that facilitates instantaneous multivalent integrin occupancy events that promote leukocyte adhesion to an endothelial ligand under shear flow.


Subject(s)
Antigens, CD/physiology , Integrin alpha4beta1/metabolism , Leukocytes/physiology , Membrane Proteins/physiology , Vascular Cell Adhesion Molecule-1/metabolism , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/physiology , Cell Adhesion , Cells, Cultured , Fibronectins/metabolism , Humans , Leukocyte Rolling , Leukocytes/metabolism , Mice , Monocytes/metabolism , Monocytes/physiology , Protein Binding , Rheology , Tetraspanin 28
15.
Blood ; 101(11): 4437-45, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12595312

ABSTRACT

Leukocyte arrest on vascular endothelium under disruptive shear flow is a multistep process that requires in situ integrin activation on the leukocyte surface by endothelium-displayed chemoattractants, primarily chemokines. A genetic deficiency of leukocyte adhesion to endothelium associated with defective beta2 integrin expression or function (LAD-1) has been described. We now report a novel severe genetic disorder in this multistep process associated with functional defects in multiple leukocyte integrins, reflected in recurrent infections, profound leukocytosis, and a bleeding tendency. This syndrome is associated with an impaired ability of neutrophil and lymphocyte beta1 and beta2 integrins to generate high avidity to their endothelial ligands and arrest cells on vascular endothelium in response to endothelial chemoattractant signals. Patient leukocytes roll normally on endothelial selectins, express intact integrins and G protein-coupled chemokine receptors (GPCR), spread on integrin ligands, and migrate normally along a chemotactic gradient. Activation of beta2 integrins in response to GPCR signals and intrinsic soluble ligand binding properties of the very late activation antigen-4 (VLA-4) integrin are also retained in patient leukocytes. Nevertheless, all integrins fail to generate firm adhesion to immobilized ligands in response to in situ GPCR-mediated activation by chemokines or chemoattractants, a result of a primary defect in integrin rearrangement at ligand-bearing contacts. This syndrome is the first example of a human integrin-activation deficiency associated with defective GPCR stimulation of integrin avidity at subsecond contacts, a key step in leukocyte arrest on vascular endothelium under shear flow.


Subject(s)
Chemokines/physiology , Endothelium, Vascular/cytology , Integrins/metabolism , Leukocyte-Adhesion Deficiency Syndrome/pathology , Cell Adhesion , Chemotaxis, Leukocyte , Child , Endothelium, Vascular/chemistry , Endothelium, Vascular/physiology , Humans , Leukocyte Rolling , Leukocyte-Adhesion Deficiency Syndrome/blood , Leukocyte-Adhesion Deficiency Syndrome/etiology , Leukocytes/chemistry , Leukocytes/pathology , Male , Perfusion , Receptors, Chemokine/metabolism , Stress, Mechanical , Umbilical Veins/cytology
16.
J Biol Chem ; 277(42): 40027-35, 2002 Oct 18.
Article in English | MEDLINE | ID: mdl-12163503

ABSTRACT

VLA-4 and LFA-1 are the major vascular integrins expressed on circulating lymphocytes. Previous studies suggested that intact cholesterol rafts are required for integrin adhesiveness in different leukocytes. We found the alpha(4) integrins VLA-4 and alpha(4)beta(7) as well as the LFA-1 integrin to be excluded from rafts of human peripheral blood lymphocytes. Disruption of cholesterol rafts with the chelator methyl-beta-cyclodextrin did not affect the ability of these lymphocyte integrins to generate high avidity to their respective endothelial ligands and to promote lymphocyte rolling and arrest on inflamed endothelium under shear flow. In contrast, cholesterol extraction abrogated rapid chemokine triggering of alpha(4)-integrin-dependent peripheral blood lymphocytes adhesion, a process tightly regulated by G(i)-protein activation of G protein-coupled chemokine receptors (GPCR). Strikingly, stimulation of LFA-1 avidity to intercellular adhesion molecule 1 (ICAM-1) by the same chemokines, although G(i)-dependent, was insensitive to raft disruption. Our results suggest that alpha(4) but not LFA-1 integrin avidity stimulation by chemokines involves rapid chemokine-induced GPCR rearrangement that takes place at cholesterol raft platforms upstream to G(i) signaling. Our results provide the first evidence that a particular chemokine/GPCR pair can activate different integrins on the same cell using distinct G(i) protein-associated machineries segregated within defined membrane compartments.


Subject(s)
Cholesterol/metabolism , Integrin alpha4beta1/chemistry , Integrins/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Actins/metabolism , Blotting, Western , Cell Adhesion , Cell Line , Cell Membrane/metabolism , Cell Separation , Cells, Cultured , Chemokines/metabolism , Cytoskeleton/metabolism , Endothelium, Vascular/cytology , Flow Cytometry , Humans , Inflammation , Integrin alpha4beta1/metabolism , Integrins/metabolism , Intercellular Adhesion Molecule-1/metabolism , Jurkat Cells , Leukocytes/metabolism , Ligands , Membrane Microdomains/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Protein Binding , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Receptors, Cell Surface/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Umbilical Veins/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...