Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Immunity ; 38(5): 1025-37, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23623383

ABSTRACT

Endothelial-dependent mechanisms of mononuclear cell influx are not well understood. We showed that acute stimulation of murine microvascular endothelial cells expressing the tumor necrosis factor receptors TNFR1 and TNFR2 with the soluble cytokine TNF led to CXCR3 chemokine generation. The TNF receptors signaled through interferon regulatory factor-1 (IRF1) to induce interferon-ß (IFN-ß) and subsequent autocrine signaling via the type I IFN receptor and the transcription factor STAT1. Both TNFR2 and TNFR1 were required for IRF1-IFNß signaling and, in human endothelial cells TNFR2 expression alone induced IFN-ß signaling and monocyte recruitment. In vivo, TNFR1 was required for acute renal neutrophil and monocyte influx after systemic TNF treatment, whereas the TNFR2-IRF1-IFN-ß autocrine loop was essential only for macrophage accumulation. In a chronic model of proliferative nephritis, IRF1 and renal-expressed TNFR2 were essential for sustained macrophage accumulation. Thus, our data identify a pathway in endothelial cells that selectively recruits monocytes during a TNF-induced inflammatory response.


Subject(s)
Interferon Regulatory Factor-1/metabolism , Interferon-beta/metabolism , Monocytes/immunology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Tumor Necrosis Factor-alpha/immunology , Animals , Autocrine Communication/immunology , Cells, Cultured , Endothelial Cells/metabolism , Humans , Inflammation/immunology , Interferon Regulatory Factor-1/genetics , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Nephritis/metabolism , Neutrophils/metabolism , Receptor, Interferon alpha-beta/metabolism , Receptors, CXCR3/biosynthesis , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , STAT1 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Mol Cell ; 56(2): 219-231, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25263595

ABSTRACT

Proinflammatory stimuli elicit rapid transcriptional responses via transduced signals to master regulatory transcription factors. To explore the role of chromatin-dependent signal transduction in the atherogenic inflammatory response, we characterized the dynamics, structure, and function of regulatory elements in the activated endothelial cell epigenome. Stimulation with tumor necrosis factor alpha prompted a dramatic and rapid global redistribution of chromatin activators to massive de novo clustered enhancer domains. Inflammatory super enhancers formed by nuclear factor-kappa B accumulate at the expense of immediately decommissioned, basal endothelial super enhancers, despite persistent histone hyperacetylation. Mass action of enhancer factor redistribution causes momentous swings in transcriptional initiation and elongation. A chemical genetic approach reveals a requirement for BET bromodomains in communicating enhancer remodeling to RNA Polymerase II and orchestrating the transition to the inflammatory cell state, demonstrated in activated endothelium and macrophages. BET bromodomain inhibition abrogates super enhancer-mediated inflammatory transcription, atherogenic endothelial responses, and atherosclerosis in vivo.


Subject(s)
Atherosclerosis/genetics , Inflammation/genetics , NF-kappa B p50 Subunit/immunology , Nuclear Proteins/antagonists & inhibitors , Transcription Factor RelA/immunology , Transcription Factors/antagonists & inhibitors , Acetylation , Animals , Atherosclerosis/immunology , Azepines/pharmacology , Cell Adhesion/immunology , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Chromatin/genetics , E-Selectin/biosynthesis , Endothelial Cells , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Enhancer Elements, Genetic , Histones/metabolism , Humans , Inflammation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B p50 Subunit/genetics , Nuclear Proteins/immunology , Protein Binding , RNA Polymerase II/genetics , Regulatory Sequences, Nucleic Acid , SOXF Transcription Factors/genetics , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factors/immunology , Transcription Initiation, Genetic , Transcription, Genetic/drug effects , Triazoles/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis
4.
J Autoimmun ; 117: 102575, 2021 02.
Article in English | MEDLINE | ID: mdl-33285511

ABSTRACT

Humoral immunity is reliant on efficient recruitment of circulating naïve B cells from blood into peripheral lymph nodes (LN) and timely transition of naive B cells to high affinity antibody (Ab)-producing cells. Current understanding of factor(s) coordinating B cell adhesion, activation and differentiation within LN, however, is incomplete. Prior studies on naïve B cells reveal remarkably strong binding to putative immunoregulator, galectin (Gal)-9, that attenuates BCR activation and signaling, implicating Gal-9 as a negative regulator in B cell biology. Here, we investigated Gal-9 localization in human tonsils and LNs and unearthed conspicuously high expression of Gal-9 on high endothelial and post-capillary venules. Adhesion analyses showed that Gal-9 can bridge human circulating and naïve B cells to vascular endothelial cells (EC), while decelerating transendothelial migration. Moreover, Gal-9 interactions with naïve B cells induced global transcription of gene families related to regulation of cell signaling and membrane/cytoskeletal dynamics. Signaling lymphocytic activation molecule F7 (SLAMF7) was among key immunoregulators elevated by Gal-9-binding, while SLAMF7's cytosolic adapter EAT-2, which is required for cell activation, was eliminated. Gal-9 also activated phosphorylation of pro-survival factor, ERK. Together, these data suggest that Gal-9 promotes B cell - EC interactions while delivering anergic signals to control B cell reactivity.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Endothelium, Vascular/metabolism , Galectins/metabolism , Immunomodulation , Signal Transduction , B-Lymphocytes/cytology , Biomarkers , Cell Adhesion , Cell Communication/immunology , Cell Differentiation/immunology , Cell Movement , Humans , Immunohistochemistry , Immunophenotyping , Lymphocyte Activation , Protein Transport
5.
Blood ; 134(17): 1430-1440, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31383641

ABSTRACT

Antibodies that bind CD47 on tumor cells and prevent interaction with SIRPα on phagocytes are active against multiple cancer types including T-cell lymphoma (TCL). Here we demonstrate that surface CD47 is heterogeneously expressed across primary TCLs, whereas major histocompatibility complex (MHC) class I, which can also suppress phagocytosis, is ubiquitous. Multiple monoclonal antibodies (mAbs) that block CD47-SIRPα interaction promoted phagocytosis of TCL cells, which was enhanced by cotreatment with antibodies targeting MHC class I. Expression levels of surface CD47 and genes that modulate CD47 pyroglutamation did not correlate with the extent of phagocytosis induced by CD47 blockade in TCL lines. In vivo treatment of multiple human TCL patient-derived xenografts or an immunocompetent murine TCL model with a short course of anti-CD47 mAb markedly reduced lymphoma burden and extended survival. Depletion of macrophages reduced efficacy in vivo, whereas depletion of neutrophils had no effect. F(ab')2-only fragments of anti-CD47 antibodies failed to induce phagocytosis by human macrophages, indicating a requirement for Fc-Fcγ receptor interactions. In contrast, F(ab')2-only fragments increased phagocytosis by murine macrophages independent of SLAMF7-Mac-1 interaction. Full-length anti-CD47 mAbs also induced phagocytosis by Fcγ receptor-deficient murine macrophages. An immunoglobulin G1 anti-CD47 mAb induced phagocytosis and natural killer cell-mediated cytotoxicity of TCL cells that was augmented by cotreatment with mogamulizumab, an anti-CCR4 mAb, or a mAb blocking MHC class I. These studies help explain the disparate activity of monotherapy with agents that block CD47 in murine models compared with patients. They also have direct translational implications for the deployment of anti-CD47 mAbs alone or in combination.


Subject(s)
Antigens, Differentiation/immunology , Antineoplastic Agents, Immunological/pharmacology , CD47 Antigen/immunology , Lymphoma, T-Cell/drug therapy , Receptors, IgG/immunology , Receptors, Immunologic/immunology , Animals , Antineoplastic Agents, Immunological/therapeutic use , CD47 Antigen/antagonists & inhibitors , Cell Line, Tumor , Humans , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/pathology , Mice , Receptors, Fc/immunology
6.
Arterioscler Thromb Vasc Biol ; 38(8): 1901-1912, 2018 08.
Article in English | MEDLINE | ID: mdl-29976772

ABSTRACT

Objective- Coronary artery thrombosis can occur in the absence of plaque rupture because of superficial erosion. Erosion-prone atheromata associate with more neutrophil extracellular traps (NETs) than lesions with stable or rupture-prone characteristics. The effects of NETs on endothelial cell (EC) inflammatory and thrombogenic properties remain unknown. We hypothesized that NETs alter EC functions related to erosion-associated thrombosis. Approach and Results- Exposure of human ECs to NETs increased VCAM-1 (vascular cell adhesion molecule 1) and ICAM-1 (intercellular adhesion molecule 1) mRNA and protein expression in a time- and concentration-dependent manner. THP-1 monocytoid cells and primary human monocytes bound more avidly to NET-treated human umbilical vein ECs than to unstimulated cells under flow. Treatment of human ECs with NETs augmented the expression of TF (tissue factor) mRNA, increased EC TF activity, and hastened clotting of recalcified plasma. Anti-TF-neutralizing antibody blocked NET-induced acceleration of clotting by ECs. NETs alone did not exhibit TF activity or acceleration of clotting in cell-free assays. Pretreatment of NETs with anti-interleukin (IL)-1α-neutralizing antibody or IL-1Ra (IL-1 receptor antagonist)-but not with anti-IL-1ß-neutralizing antibody or control IgG-blocked NET-induced VCAM-1, ICAM-1, and TF expression. Inhibition of cathepsin G, a serine protease abundant in NETs, also limited the effect of NETs on EC activation. Cathepsin G potentiated the effect of IL-1α on ECs by cleaving the pro-IL-1α precursor and releasing the more potent mature IL-1α form. Conclusions- NETs promote EC activation and increased thrombogenicity through concerted action of IL-1α and cathepsin G. Thus, NETs may amplify and propagate EC dysfunction related to thrombosis because of superficial erosion.


Subject(s)
Blood Coagulation , Cathepsin G/metabolism , Extracellular Traps/enzymology , Human Umbilical Vein Endothelial Cells/enzymology , Interleukin-1alpha/metabolism , Neutrophils/enzymology , Paracrine Communication , Thromboplastin/metabolism , Cell Adhesion , Coculture Techniques , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Signal Transduction , THP-1 Cells , Thromboplastin/genetics , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
7.
Cell Tissue Res ; 355(3): 647-56, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24562377

ABSTRACT

The immune cell system is a critical component of host defense. Recruitment of immune cells to sites of infection, immune reaction, or injury is complex and involves coordinated adhesive interactions between the leukocyte and the endothelial cell monolayer that lines blood vessels. This article reviews basic mechanisms in the recruitment of leukocytes to tissues and then selectively reviews new concepts that are emerging based on advances in live cell imaging microscopy and mouse strains. These emerging concepts are altering the conventional paradigms of inflammatory leukocyte recruitment established in the early 1990s. Indeed, recent publications have identified previously unrecognized contributions from pericytes and interstitial leukocytes and their secreted products that guide leukocytes to their targets. Investigators have also begun to design organs on a chip. Recent reports indicate that this avenue of research holds much promise.


Subject(s)
Image Processing, Computer-Assisted/methods , Inflammation/immunology , Leukocytes/immunology , Microscopy, Fluorescence/methods , Animals , Humans
8.
Trends Immunol ; 32(10): 461-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21839681

ABSTRACT

Neutrophils are the all-terrain vehicle of the innate immune system because of their ability to gain entry into tissues and organs, and thus, play an essential role in host defense. Exactly how this marvel of nature works is still incompletely understood. In the past 2-3 years, new players and processes have been identified in the endothelial-leukocyte adhesion cascade. Novel signaling pathways have been discovered in both the endothelium and the neutrophils that regulate various steps in the recruitment process. This review focuses on these emerging pathways and the mechanisms that regulate neutrophil recruitment across endothelium.


Subject(s)
Cell Adhesion/immunology , Chemokines/immunology , Immunity, Innate , Inflammation/immunology , Neutrophils , Receptors, Chemokine/immunology , Signal Transduction/immunology , Transendothelial and Transepithelial Migration/immunology , Animals , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Chemotaxis, Leukocyte/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Mice , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Chemokine/metabolism
9.
Arterioscler Thromb Vasc Biol ; 33(11): 2566-76, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23990210

ABSTRACT

OBJECTIVE: Although endothelial CD47, a member of the immunoglobulin superfamily, has been implicated in leukocyte diapedesis, its capacity for intracellular signaling and physical localization during this process has not been addressed in detail. This study examined endothelial CD47 spatiotemporal behavior and signaling pathways involved in regulating T-cell transendothelial migration. APPROACH AND RESULTS: By biochemical methods, transmigration assays, and live-cell microscopy techniques, we show that endothelial CD47 engagement results in intracellular calcium mobilization, increased permeability, and activation of Src and AKT1/phosphoinositide 3-kinase in brain microvascular endothelial cells. These signaling pathways converge to induce cytoskeleton remodeling and vascular endothelial cadherin phosphorylation, which are necessary steps during T-cell transendothelial migration. In addition, during T-cell migration, transmigratory cups and podo-prints enriched in CD47 appear on the surface of the endothelium, indicating that the spatial distribution of CD47 changes after its engagement. Consistent with previous findings of intercellular adhesion molecule 1, blockade of CD47 results in decreased T-cell transmigration across microvascular endothelium. The overlapping effect of intercellular adhesion molecule 1 and CD47 suggests their involvement in different steps of the diapedesis process. CONCLUSIONS: These data reveal a novel role for CD47-mediated signaling in the control of the molecular network governing endothelial-dependent T-cell diapedesis.


Subject(s)
CD47 Antigen/immunology , CD47 Antigen/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , Transendothelial and Transepithelial Migration/immunology , Actins/metabolism , Animals , Brain/blood supply , Calcium/metabolism , Cells, Cultured , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Humans , Intercellular Junctions/immunology , Intercellular Junctions/metabolism , Microvessels/immunology , Microvessels/metabolism , Rats , T-Lymphocytes/metabolism
10.
J Immunol ; 188(3): 1421-30, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22219321

ABSTRACT

T cell subset-specific migration to inflammatory sites is tightly regulated and involves interaction of the T cells with the endothelium. Th17 cells often appear at different inflammatory sites than Th1 cells, or both subsets appear at the same sites but at different times. Differences in T cell subset adhesion to endothelium may contribute to subset-specific migratory behavior, but this possibility has not been well studied. We examined the adhesion of mouse Th17 cells to endothelial adhesion molecules and endothelium under flow in vitro and to microvessels in vivo and we characterized their migratory phenotype by flow cytometry and quantitative RT-PCR. More Th17 than Th1 cells interacted with E-selectin. Fewer Th17 than Th1 cells bound to TNF-α-activated E-selectin-deficient endothelial cells, and intravital microscopy studies demonstrated that Th17 cells engage in more rolling interactions with TNF-α-treated microvessels than Th1 cells in wild-type mice but not in E-selectin-deficient mice. Th17 adhesion to ICAM-1 was dependent on integrin activation by CCL20, the ligand for CCR6, which is highly expressed by Th17 cells. In an air pouch model of inflammation, CCL20 triggered recruitment of Th17 but not Th1 cells. These data provide evidence that E-selectin- and ICAM-1-dependent adhesion of Th17 and Th1 cells with endothelium are quantitatively different.


Subject(s)
Cell Adhesion/immunology , Endothelium, Vascular/immunology , Th1 Cells/physiology , Animals , Chemokine CCL20/metabolism , E-Selectin/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Intercellular Adhesion Molecule-1/immunology , Mice , Th17 Cells
11.
J Immunol ; 189(5): 2553-62, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22815286

ABSTRACT

At sites of inflammation, endothelial adhesion molecules bind leukocytes and transmit signals required for transendothelial migration (TEM). We previously reported that adhesive interactions between endothelial cell CD47 and leukocyte signal regulatory protein γ (SIRPγ) regulate human T cell TEM. The role of endothelial CD47 in T cell TEM in vivo, however, has not been explored. In this study, CD47⁻/⁻ mice showed reduced recruitment of blood T cells as well as neutrophils and monocytes in a dermal air pouch model of TNF-α-induced inflammation. Reconstitution of CD47⁻/⁻ mice with wild-type bone marrow cells did not restore leukocyte recruitment to the air pouch, indicating a role for endothelial CD47. The defect in leukocyte TEM in the CD47⁻/⁻ endothelium was corroborated by intravital microscopy of inflamed cremaster muscle microcirculation in bone marrow chimera mice. In an in vitro human system, CD47 on both HUVEC and T cells was required for TEM. Although previous studies showed CD47-dependent signaling required G(αi)-coupled pathways, this was not the case for endothelial CD47 because pertussis toxin, which inactivates G(αi), had no inhibitory effect, whereas G(αi) was required by the T cell for TEM. We next investigated the endothelial CD47-dependent signaling events that accompany leukocyte TEM. Ab-induced cross-linking of CD47 revealed robust actin cytoskeleton reorganization and Src- and Pyk-2-kinase dependent tyrosine phosphorylation of the vascular endothelial-cadherin cytoplasmic tail. This signaling was pertussis toxin insensitive, suggesting that endothelial CD47 signaling is independent of G(αi). These findings suggest that engagement of endothelial CD47 by its ligands triggers outside-in signals in endothelium that facilitate leukocyte TEM.


Subject(s)
CD47 Antigen/physiology , Cadherins/blood , Chemotaxis, Leukocyte/immunology , Endothelium, Vascular/metabolism , Inflammation/immunology , Inflammation/pathology , T-Lymphocyte Subsets/immunology , Tyrosine/blood , Animals , CD47 Antigen/genetics , CD47 Antigen/metabolism , Disease Models, Animal , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/blood , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/immunology , Recombinant Proteins/toxicity , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Tumor Necrosis Factor-alpha/toxicity
12.
J Immunol ; 188(12): 6287-99, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22566565

ABSTRACT

IL-17A (IL-17) is the signature cytokine produced by Th17 cells and has been implicated in host defense against infection and the pathophysiology of autoimmunity and cardiovascular disease. Little is known, however, about the influence of IL-17 on endothelial activation and leukocyte influx to sites of inflammation. We hypothesized that IL-17 would induce a distinct pattern of endothelial activation and leukocyte recruitment when compared with the Th1 cytokine IFN-γ. We found that IL-17 alone had minimal activating effects on cultured endothelium, whereas the combination of TNF-α and IL-17 produced a synergistic increase in the expression of both P-selectin and E-selectin. Using intravital microscopy of the mouse cremaster muscle, we found that TNF-α and IL-17 also led to a synergistic increase in E-selectin-dependent leukocyte rolling on microvascular endothelium in vivo. In addition, TNF-α and IL-17 enhanced endothelial expression of the neutrophilic chemokines CXCL1, CXCL2, and CXCL5 and led to a functional increase in leukocyte transmigration in vivo and CXCR2-dependent neutrophil but not T cell transmigration in a parallel-plate flow chamber system. By contrast, endothelial activation with TNF-α and IFN-γ preferentially induced the expression of the integrin ligands ICAM-1 and VCAM-1, as well as the T cell chemokines CXCL9, CXCL10, and CCL5. These effects were further associated with a functional increase in T cell but not neutrophil transmigration under laminar shear flow. Overall, these data show that IL-17 and TNF-α act in a synergistic manner to induce a distinct pattern of endothelial activation that sustains and enhances neutrophil influx to sites of inflammation.


Subject(s)
Endothelial Cells/metabolism , Inflammation/metabolism , Interleukin-17/metabolism , Neutrophil Infiltration/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Chemokines/biosynthesis , Endothelial Cells/immunology , Flow Cytometry , Inflammation/immunology , Interleukin-17/immunology , Leukocyte Rolling/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/immunology
13.
Eur Heart J ; 34(8): 615-24, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22927557

ABSTRACT

AIMS: Activation of vascular endothelial cells (ECs) contributes importantly to inflammation and atherogenesis. We previously reported that apolipoprotein CIII (apoCIII), found abundantly on circulating triglyceride-rich lipoproteins, enhances adhesion of human monocytes to ECs in vitro. Statins may exert lipid-independent anti-inflammatory effects. The present study examined whether statins suppress apoCIII-induced EC activation in vitro and in vivo. METHODS AND RESULTS: Physiologically relevant concentrations of purified human apoCIII enhanced attachment of the monocyte-like cell line THP-1 to human saphenous vein ECs (HSVECs) or human coronary artery ECs (HCAECs) under both static and laminar shear stress conditions. This process mainly depends on vascular cell adhesion molecule-1 (VCAM-1), as a blocking VCAM-1 antibody abolished apoCIII-induced monocyte adhesion. ApoCIII significantly increased VCAM-1 expression in HSVECs and HCAECs. Pre-treatment with statins suppressed apoCIII-induced VCAM-1 expression and monocyte adhesion, with two lipophilic statins (pitavastatin and atorvastatin) exhibiting inhibitory effects at lower concentration than those of hydrophilic pravastatin. Nuclear factor κB (NF-κB) mediated apoCIII-induced VCAM-1 expression, as demonstrated via loss-of-function experiments, and pitavastatin treatment suppressed NF-κB activation. Furthermore, in the aorta of hypercholesterolaemic Ldlr(-/-) mice, pitavastatin administration in vivo suppressed VCAM-1 mRNA and protein, induced by apoCIII bolus injection. Similarly, in a subcutaneous dorsal air pouch mouse model of leucocyte recruitment, apoCIII injection induced F4/80+ monocyte and macrophage accumulation, whereas pitavastatin administration reduced this effect. CONCLUSIONS: These findings further establish the direct role of apoCIII in atherogenesis and suggest that anti-inflammatory effects of statins could improve vascular disease in the population with elevated plasma apoCIII.


Subject(s)
Apolipoprotein C-III/antagonists & inhibitors , Endothelial Cells/physiology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Leukocytes, Mononuclear/drug effects , Quinolines/pharmacology , Animals , Aorta , Cell Adhesion/physiology , Cells, Cultured , Humans , Leukocytes, Mononuclear/physiology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Saphenous Vein , Vascular Cell Adhesion Molecule-1/metabolism
14.
J Exp Med ; 204(2): 431-9, 2007 Feb 19.
Article in English | MEDLINE | ID: mdl-17296784

ABSTRACT

The transcription factor T-bet was identified in CD4(+) T cells, and it controls interferon gamma production and T helper type 1 cell differentiation. T-bet is expressed in certain other leukocytes, and we recently showed (Lord, G.M., R.M. Rao, H. Choe, B.M. Sullivan, A.H. Lichtman, F.W. Luscinskas, and L.H. Glimcher. 2005. Blood. 106:3432-3439) that it regulates T cell trafficking. We examined whether T-bet influences homing of mast cell progenitors (MCp) to peripheral tissues. Surprisingly, we found that MCp homing to the lung or small intestine in T-bet(-/-) mice is reduced. This is reproduced in adhesion studies using bone marrow-derived MCs (BMMCs) from T-bet(-/-) mice, which showed diminished adhesion to mucosal addresin cellular adhesion molecule-1 (MAdCAM-1) and vascular cell adhesion molecule-1 (VCAM-1), endothelial ligands required for MCp intestinal homing. MCp, their precursors, and BMMCs do not express T-bet, suggesting that T-bet plays an indirect role in homing. However, adoptive transfer experiments revealed that T-bet expression by BM cells is required for MCp homing to the intestine. Furthermore, transfer of WT BM-derived dendritic cells (DCs) to T-bet(-/-) mice restores normal MCp intestinal homing in vivo and MCp adhesion to MAdCAM-1 and VCAM-1 in vitro. Nonetheless, T-bet(-/-) mice respond vigorously to intestinal infection with Trichinella spiralis, eliminating a role for T-bet in MC recruitment to sites of infection and their activation and function. Therefore, remarkably, T-bet expression by DCs indirectly controls MCp homing to mucosal tissues.


Subject(s)
Bone Marrow Cells/metabolism , Cell Adhesion/physiology , Cell Movement/physiology , Dendritic Cells/metabolism , Mast Cells/cytology , Stem Cells/metabolism , T-Box Domain Proteins/metabolism , Animals , Blotting, Western , Cell Adhesion Molecules/metabolism , DNA Primers , Gastric Mucosa/metabolism , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mucoproteins , Respiratory Mucosa/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/physiology , T-Box Domain Proteins/genetics , Vascular Cell Adhesion Molecule-1/metabolism
15.
Gastroenterology ; 143(6): 1544-1554.e7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22960654

ABSTRACT

BACKGROUND & AIMS: Signaling lymphocyte activation molecule (Slamf)1 is a co-stimulatory receptor on T cells and regulates cytokine production by macrophages and dendritic cells. Slamf1 regulates microbicidal mechanisms in macrophages, therefore we investigated whether the receptor affects development of colitis in mice. METHODS: We transferred CD45RB(hi) CD4(+) T cells into Rag(-/-) or Slamf1(-/-)Rag(-/-) mice to induce colitis. We also induced colitis by injecting mice with an antibody that activates CD40. We determined the severity of enterocolitis based on disease activity index, histology scores, and levels of cytokine production, and assessed the effects of antibodies against Slamf1 on colitis induction. We quantified migration of monocytes and macrophage to inflamed tissues upon induction of colitis or thioglycollate-induced peritonitis and in response to tumor necrosis factor-α in an air-pouch model of leukocyte migration. RESULTS: Colitis was reduced in Slamf1(-/-)Rag(-/-) mice, compared with Rag(-/-) mice, after transfer of CD45RB(hi) CD4(+) T cells or administration of the CD40 agonist. The numbers of monocytes and macrophages were reduced in inflamed tissues of Slamf1(-/-)Rag(-/-) mice, compared with Rag(-/-) mice, after induction of colitis and other inflammatory disorders. An antibody that inhibited Slamf1 reduced the level of enterocolitis in Rag(-/-) mice. CONCLUSIONS: Slamf1 contributes to the development of colitis in mice. It appears to indirectly regulate the appearance of monocytes and macrophages in inflamed intestinal tissues. Antibodies that inhibit Slamf1 reduce colitis in mice, so human SLAMF1 might be a therapeutic target for inflammatory bowel disease.


Subject(s)
Antigens, CD/physiology , Colitis/physiopathology , Receptors, Cell Surface/physiology , Animals , Antigens, CD/genetics , CD40 Antigens/adverse effects , Cell Movement , Chemokine CCL2/blood , Chemokine CCL7/blood , Colitis/blood , Colitis/chemically induced , Disease Models, Animal , Intestines/pathology , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Signaling Lymphocytic Activation Molecule Family Member 1
16.
Eur J Immunol ; 42(2): 296-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22266717

ABSTRACT

A major focus of researchers studying leukocyte recruitment has been to identify and understand how cell surface endothelial adhesion molecules, cell-to-cell junctional protein complexes, secreted chemokines and chemoattractants, and the vessel basement membrane structure organization coordinate the process of leukocyte recruitment. As research expands beyond the components initially identified as being necessary for leukocyte recruitment, attention has turned to the structures that regulate endothelial cell-to-matrix adhesion. In this issue of the European Journal of Immunology, Parsons et al. [Eur. J. Immunol. 2012. 42: 436-446] identify new players in the regulation of neutrophil diapedesis (transendothelial migration), namely the focal adhesion proteins, paxillin and focal adhesion kinase (FAK). While understudied, and indeed previously underappreciated, in leukocyte diapedesis, this Commentary discusses how the work by Parsons et al. implicates FAK and paxillin in the proximal (leukocyte rolling) and distal (diapedesis) steps of the multistep adhesion cascade of leukocyte recruitment.


Subject(s)
Endothelium/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Neutrophils/metabolism , Paxillin/metabolism , Transendothelial and Transepithelial Migration/immunology , Humans
17.
J Immunol ; 187(7): 3521-9, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21873519

ABSTRACT

The ability of regulatory T cells (Treg) to traffic to sites of inflammation supports their role in controlling immune responses. This feature supports the idea that adoptive transfer of in vitro expanded human Treg could be used for treatment of immune/inflammatory diseases. However, the migratory behavior of Treg, as well as their direct influence at the site of inflammation, remains poorly understood. To explore the possibility that Treg may have direct anti-inflammatory influences on tissues, independent of their well-established suppressive effects on lymphocytes, we studied the adhesive interactions between mouse Treg and endothelial cells, as well as their influence on endothelial function during acute inflammation. We show that Foxp3(+) adaptive/inducible Treg (iTreg), but not naturally occurring Treg, efficiently interact with endothelial selectins and transmigrate through endothelial monolayers in vitro. In response to activation by endothelial Ag presentation or immobilized anti-CD3ε, Foxp3(+) iTreg suppressed TNF-α- and IL-1ß-mediated endothelial selectin expression and adhesiveness to effector T cells. This suppression was contact independent, rapid acting, and mediated by TGF-ß-induced activin receptor-like kinase 5 signaling in endothelial cells. In addition, Foxp3(+) iTreg adhered to inflamed endothelium in vivo, and their secretion products blocked acute inflammation in a model of peritonitis. These data support the concept that Foxp3(+) iTreg help to regulate inflammation independently of their influence on effector T cells by direct suppression of endothelial activation and leukocyte recruitment.


Subject(s)
Chemotaxis, Leukocyte/immunology , Endothelium, Vascular/immunology , Inflammation/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Adhesion/immunology , Cell Separation , Endothelium, Vascular/metabolism , Flow Cytometry , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Knock-In Techniques , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/metabolism
18.
Proc Natl Acad Sci U S A ; 107(37): 16252-6, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20805498

ABSTRACT

The F-BAR domain containing protein CIP4 (Cdc42 interacting protein 4) interacts with Cdc42 and WASP/N-WASP and is thought to participate in the assembly of filamentous actin. CIP4(-/-) mice had normal T- and B-lymphocyte development but impaired T cell-dependent antibody production, IgG antibody affinity maturation, and germinal center (GC) formation, despite an intact CD40L-CD40 axis. CIP4(-/-) mice also had impaired contact hypersensitivity (CHS) to haptens, and their T cells failed to adoptively transfer CHS. Ovalbumin-activated CD4(+) effector T cells from CIP4(-/-)/OT-II mice migrated poorly to antigen-challenged skin. Activated CIP4(-/-) T cells exhibited impaired adhesion and polarization on immobilized VCAM-1 and ICAM-1 and defective arrest and transmigration across murine endothelial cell monolayers under shear flow conditions. These results demonstrate an important role for CIP4 in integrin-dependent T cell-dependent antibody responses and GC formation and in integrin-mediated recruitment of effector T cells to cutaneous sites of antigen-driven immune reactions.


Subject(s)
Cell Movement , Integrins/immunology , Microtubule-Associated Proteins/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/immunology , Cell Adhesion , Cell Polarity , Dermatitis, Allergic Contact/genetics , Dermatitis, Allergic Contact/immunology , Intercellular Adhesion Molecule-1/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/deficiency , Minor Histocompatibility Antigens , Vascular Cell Adhesion Molecule-1/immunology
19.
Am J Physiol Cell Physiol ; 303(4): C385-95, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22648953

ABSTRACT

Leukocyte transendothelial migration (TEM) is regulated by several signaling pathways including Src family kinases (SFK) and the small RhoGTPases. Previous studies have shown that vascular endothelial-cadherin (VE-cad) forms a complex with ß-,γ-, and p120-catenins and this complex disassociates to form a transient gap during leukocyte TEM. Additionally, p120-catenin (p120-1A) overexpression in human umbilical vein endothelial cells (HUVEC) stabilizes VE-cad surface expression, prevents tyrosine phosphorylation of VE-cad, and inhibits leukocyte TEM. Based on reports showing that p120 overexpression in fibroblasts or epithelial cells inhibits RhoA and activates Rac and Cdc42 GTPases, and on other reports showing that RhoA activation in endothelial cells is necessary for leukocyte TEM, we reasoned that p120 overexpression inhibited TEM through inhibition of RhoA. To test this idea, we overexpressed a mutant p120 isoform, p120-4A, which does not interact with RhoA. p120-4A colocalized with VE-cad in HUVEC junctions and enhanced VE-cad surface expression, similar to overexpression of p120-1A. Interestingly, overexpression of either p120-4A or p120-1A dramatically blocked TEM, and overexpression of p120-1A in HUVEC did not affect RhoA basal activity or activation of RhoA and Rac induced by thrombin or ICAM-1 crosslinking. In contrast, biochemical studies revealed that overexpression of p120-1A reduced activated pY416-Src association with VE-cad. In summary, p120 overexpression inhibits neutrophil TEM independently of an effect on RhoA or Rac and instead blocks TEM by preventing VE-cad tyrosine phosphorylation and association of active Src with the VE-cad complex.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Catenins/metabolism , Neutrophils/drug effects , Neutrophils/physiology , Proto-Oncogene Proteins pp60(c-src)/metabolism , rhoA GTP-Binding Protein/metabolism , Antigens, CD/genetics , Cadherins/genetics , Catenins/genetics , Cell Movement/physiology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Phosphorylation , Proto-Oncogene Proteins pp60(c-src)/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , rhoA GTP-Binding Protein/genetics , Delta Catenin
20.
J Exp Med ; 203(4): 985-94, 2006 Apr 17.
Article in English | MEDLINE | ID: mdl-16585266

ABSTRACT

Multiple sclerosis (MS) is a devastating inflammatory disorder of the central nervous system (CNS). A major hallmark of MS is the infiltration of T cells reactive against myelin components. T cell infiltration is mediated by the interaction of integrins of the beta1 and beta2 family expressed by lymphocytes with their endothelial counter-receptors, vascular cell adhesion molecule 1 and intercellular adhesion molecule (ICAM)-1, respectively. We have reported previously that extracellular adherence protein (Eap) of Staphylococcus aureus exerts antiinflammatory activities by interacting with ICAM-1 and blocking beta2-integrin-dependent neutrophil recruitment. Here, we report that Eap inhibits experimental autoimmune encephalomyelitis (EAE) in mice. In vitro, Eap reduced adhesion of peripheral blood T cells to immobilized ICAM-1 as well as their adhesion and transmigration of TNF-activated human endothelium under static and shear flow conditions. These inhibitory effects were corroborated in two mouse models of inflammation. In a delayed-type hypersensitivity model, both T cell infiltration and the corresponding tissue edema were significantly reduced by Eap. In addition, Eap administration prevented the development of EAE and markedly decreased infiltration of inflammatory cells into the CNS. Strikingly, intervention with Eap after the onset of EAE suppressed the disease. Collectively, our findings indicate that Eap represents an attractive treatment for autoimmune neuroinflammatory disorders such as MS.


Subject(s)
Bacterial Proteins/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/microbiology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , RNA-Binding Proteins/therapeutic use , Staphylococcus aureus/immunology , Amino Acid Sequence , Animals , Cell Adhesion/immunology , Cell Communication/immunology , Cell Migration Inhibition , Cell Movement/immunology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/microbiology , Hypersensitivity, Delayed/prevention & control , Intercellular Adhesion Molecule-1/physiology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , T-Lymphocytes/cytology , T-Lymphocytes/microbiology , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL