Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(4): e0266566, 2022.
Article in English | MEDLINE | ID: mdl-35413056

ABSTRACT

The SIRPα-CD47 axis plays an important role in T cell recruitment to sites of immune reaction and inflammation but its role in T cell antigen priming is incompletely understood. Employing OTII TCR transgenic mice bred to Cd47-/- (Cd47KO) or SKI mice, a knock-in transgenic animal expressing non-signaling cytoplasmic-truncated SIRPα, we investigated how the SIRPα-CD47 axis contributes to antigen priming. Here we show that adoptive transfer of Cd47KO or SKI Ova-specific CD4+ T cells (OTII) into Cd47KO and SKI recipients, followed by Ova immunization, elicited reduced T cell division and proliferation indices, increased apoptosis, and reduced expansion compared to transfer into WT mice. We confirmed prior reports that splenic T cell zone, CD4+ conventional dendritic cells (cDCs) and CD4+ T cell numbers were reduced in Cd47KO and SKI mice. We report that in vitro derived DCs from Cd47KO and SKI mice exhibited impaired migration in vivo and exhibited reduced CD11c+ DC proximity to OTII T cells in T cell zones after Ag immunization, which correlates with reduced TCR activation in transferred OTII T cells. These findings suggest that reduced numbers of CD4+ cDCs and their impaired migration contributes to reduced T cell-DC proximity in splenic T cell zone and reduced T cell TCR activation, cell division and proliferation, and indirectly increased T cell apoptosis.


Subject(s)
CD47 Antigen , Receptors, Immunologic , Spleen , Animals , Antigens , CD47 Antigen/genetics , CD47 Antigen/metabolism , Cell Communication , Dendritic Cells , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/metabolism
2.
JCI Insight ; 6(21)2021 11 08.
Article in English | MEDLINE | ID: mdl-34591795

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a well-characterized animal model of multiple sclerosis. During the early phase of EAE, infiltrating monocytes and monocyte-derived macrophages contribute to T cell recruitment, especially CD4+ T cells, into the CNS, resulting in neuronal demyelination; however, in later stages, they promote remyelination and recovery by removal of myelin debris by phagocytosis. Signal regulatory protein α and CD47 are abundantly expressed in the CNS, and deletion of either molecule is protective in myelin oligodendrocyte glycoprotein-induced EAE because of failed effector T cell expansion and trafficking. Here we report that treatment with the function blocking CD47 Ab Miap410 substantially reduced the infiltration of pathogenic immune cells but impaired recovery from paresis. The underlying mechanism was by blocking the emergence of CD11chiMHCIIhi microglia at peak disease that expressed receptors for phagocytosis, scavenging, and lipid catabolism, which mediated clearance of myelin debris and the transition of monocytes to macrophages in the CNS. In the recovery phase of EAE, Miap410 Ab-treated mice had worsening paresis with sustained inflammation and limited remyelination as compared with control Ab-treated mice. In summary, Ab blockade of CD47 impaired resolution of CNS inflammation, thus worsening EAE.


Subject(s)
CD47 Antigen/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Macrophages/metabolism , Microglia/metabolism , Monocytes/metabolism , Phagocytosis/genetics , Animals , Disease Models, Animal , Female , Mice , Mice, Knockout
3.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34156982

ABSTRACT

The stimulator of IFN genes (STING) protein senses cyclic dinucleotides released in response to double-stranded DNA and functions as an adaptor molecule for type I IFN (IFNI) signaling by activating IFNI-stimulated genes (ISG). We found impaired T cell infiltration into the peritoneum in response to TNF-α in global and EC-specific STING-/- mice and discovered that T cell transendothelial migration (TEM) across mouse and human endothelial cells (EC) deficient in STING was strikingly reduced compared with control EC, whereas T cell adhesion was not impaired. STING-/- T cells showed no defect in TEM or adhesion to EC, or immobilized endothelial cell-expressed molecules ICAM1 and VCAM1, compared with WT T cells. Mechanistically, CXCL10, an ISG and a chemoattractant for T cells, was dramatically reduced in TNF-α-stimulated STING-/- EC, and genetic loss or pharmacologic antagonisms of IFNI receptor (IFNAR) pathway reduced T cell TEM. Our data demonstrate a central role for EC-STING during T cell TEM that is dependent on the ISG CXCL10 and on IFNI/IFNAR signaling.


Subject(s)
Interferon Type I , Membrane Proteins/immunology , Receptor, Interferon alpha-beta , T-Lymphocytes , Transendothelial and Transepithelial Migration/immunology , Animals , Immunity, Innate , Intercellular Adhesion Molecule-1/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Mice , Receptor, Interferon alpha-beta/immunology , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/immunology
4.
Mucosal Immunol ; 14(2): 331-341, 2021 03.
Article in English | MEDLINE | ID: mdl-32561828

ABSTRACT

Dysregulated neutrophil (PMN) transmigration across epithelial surfaces (TEpM) significantly contributes to chronic inflammatory diseases, yet mechanisms defining this process remain poorly understood. In the intestine, uncontrolled PMN TEpM is a hallmark of disease flares in ulcerative colitis. Previous in vitro studies directed at identifying molecular determinants that mediate TEpM have shown that plasma membrane proteins including CD47 and CD11b/CD18 play key roles in regulating PMN TEpM across monolayers of intestinal epithelial cells. Here, we show that CD47 modulates PMN TEpM in vivo using an ileal loop assay. Importantly, using novel tissue-specific CD47 knockout mice and in vitro approaches, we report that PMN-expressed, but not epithelial-expressed CD47 plays a major role in regulating PMN TEpM. We show that CD47 associates with CD11b/CD18 in the plasma membrane of PMN, and that loss of CD47 results in impaired CD11b/CD18 activation. In addition, in vitro and in vivo studies using function blocking antibodies support a role of CD47 in regulating CD11b-dependent PMN TEpM and chemotaxis. Taken together, these findings provide new insights for developing approaches to target dysregulated PMN infiltration in the intestine. Moreover, tissue-specific CD47 knockout mice constitute an important new tool to study contributions of cells expressing CD47 to inflammation in vivo.


Subject(s)
CD47 Antigen/metabolism , Inflammation/immunology , Intestines/immunology , Neutrophils/immunology , Animals , CD11b Antigen/metabolism , CD18 Antigens/metabolism , CD47 Antigen/genetics , Cells, Cultured , Chemotaxis , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Activation , Organ Specificity , Transendothelial and Transepithelial Migration
5.
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
6.
Nat Commun ; 11(1): 6396, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33328477

ABSTRACT

Clinical studies reveal changes in blood eosinophil counts and eosinophil cationic proteins that may serve as risk factors for human coronary heart diseases. Here we report an increase of blood or heart eosinophil counts in humans and mice after myocardial infarction (MI), mostly in the infarct region. Genetic or inducible depletion of eosinophils exacerbates cardiac dysfunction, cell death, and fibrosis post-MI, with concurrent acute increase of heart and chronic increase of splenic neutrophils and monocytes. Mechanistic studies reveal roles of eosinophil IL4 and cationic protein mEar1 in blocking H2O2- and hypoxia-induced mouse and human cardiomyocyte death, TGF-ß-induced cardiac fibroblast Smad2/3 activation, and TNF-α-induced neutrophil adhesion on the heart endothelial cell monolayer. In vitro-cultured eosinophils from WT mice or recombinant mEar1 protein, but not eosinophils from IL4-deficient mice, effectively correct exacerbated cardiac dysfunctions in eosinophil-deficient ∆dblGATA mice. This study establishes a cardioprotective role of eosinophils in post-MI hearts.


Subject(s)
Eosinophils/physiology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Aged , Animals , Cell Death , Diphtheria Toxin/toxicity , Electrocardiography , Eosinophils/drug effects , Eosinophils/pathology , Female , Fibroblasts/pathology , Fibroblasts/physiology , Humans , Interleukin-4/genetics , Interleukin-4/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/pathology , Ribonucleases/genetics , Ribonucleases/metabolism
7.
JCI Insight ; 5(15)2020 08 06.
Article in English | MEDLINE | ID: mdl-32573493

ABSTRACT

T follicular helper (Tfh) cell migration into germinal centers (GCs) is essential for the generation of GC B cells and antibody responses to T cell-dependent (TD) antigens. This process requires interactions between lymphocyte function-associated antigen 1 (LFA-1) on Tfh cells and ICAMs on B cells. The mechanisms underlying defective antibody responses to TD antigens in DOCK8 deficiency are incompletely understood. We show that mice selectively lacking DOCK8 in T cells had impaired IgG antibody responses to TD antigens, decreased GC size, and reduced numbers of GC B cells. However, they developed normal numbers of Tfh cells with intact capacity for driving B cell differentiation into a GC phenotype in vitro. Notably, migration of DOCK8-deficient T cells into GCs was defective. Following T cell receptor (TCR)/CD3 ligation, DOCK8-deficient T cells had impaired LFA-1 activation and reduced binding to ICAM-1. Our results therefore indicate that DOCK8 is important for LFA-1-dependent positioning of Tfh cells in GCs, and thereby the generation of GC B cells and IgG antibody responses to TD antigen.


Subject(s)
Antibody Formation/immunology , B-Lymphocytes/immunology , Germinal Center/immunology , Guanine Nucleotide Exchange Factors/physiology , Lymphocyte Activation/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , T Follicular Helper Cells/immunology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Differentiation , Child , Child, Preschool , Female , Germinal Center/metabolism , Germinal Center/pathology , Humans , Immunity, Humoral , Lymphocyte Function-Associated Antigen-1/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/metabolism , T Follicular Helper Cells/metabolism , T Follicular Helper Cells/pathology
8.
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
9.
Sci Rep ; 9(1): 10608, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31337788

ABSTRACT

CD47, also known as integrin-associated protein (IAP), is a transmembrane protein with multiple biological functions including regulation of efferocytosis and leukocyte trafficking. In this study we investigated the effect of CD47-deficiency on atherosclerosis using a model of adeno-associated virus (AAV)-induced hypercholesterolemia. We observed increased plaque formation in CD47 null mice compared to wild-type controls. Loss of CD47 caused activation of dendritic cells, T cells and natural killer (NK) cells, indicating an important role for CD47 in regulating immunity. In particular, Cd47 deficiency increased the proportion of IFN-γ producing CD90+ NK cells. Treatment with depleting anti-NK1.1 monoclonal antibody (mAb), but not depleting anti-CD4/CD8 mAbs, equalized atherosclerotic burden, suggesting NK cells were involved in the enhanced disease in Cd47 deficient mice. Additional studies revealed that levels of CD90+ and IFN-γ+ NK cells were expanded in atherosclerotic aorta and that CD90+ NK cells produce more IFN-γ than CD90- NK cells. Finally, we demonstrate that anti-CD47 (MIAP410) causes splenomegaly and activation of DCs and T cells, without affecting NK cell activation. In summary, we demonstrate that loss of CD47 causes increased lymphocyte activation that results in increased atherosclerosis.


Subject(s)
Atherosclerosis/etiology , CD47 Antigen/deficiency , Lymphocyte Activation , Animals , Dendritic Cells/metabolism , Disease Models, Animal , Female , Flow Cytometry , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/metabolism
10.
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
11.
Sci Immunol ; 2(7)2017 Jan.
Article in English | MEDLINE | ID: mdl-28529998

ABSTRACT

The deposition of immune complexes (IC) in tissues induces a "type III hypersensitivity" that results in tissue damage and underlies the pathogenesis of many autoimmune diseases. The neutrophil is the first immune cell recruited into sites of IC deposition and plays a critical role in shaping the overall tissue response. However, the mechanism by which IC initiate and propagate neutrophil infiltration into tissue is not known. Here, using intravital multiphoton joint imaging of IC-induced arthritis in live mice, we found that the complement C5a receptor (C5aR) was the key initiator of neutrophil adhesion on joint endothelium. C5a presented on joint endothelium induced ß2 integrin-dependent neutrophil arrest, facilitating neutrophil spreading and transition to crawling, and subsequent leukotriene B4 receptor (BLT1)-mediated extravasation of the first neutrophils. The chemokine receptor CCR1 promoted neutrophil crawling on the joint endothelium while CXCR2 amplified late neutrophil recruitment and survival once in the joint. Thus, imaging arthritis has defined a new paradigm for type III hypersensitivity where C5a directly initiates neutrophil adhesion on the joint endothelium igniting inflammation.

12.
J Leukoc Biol ; 101(2): 493-505, 2017 02.
Article in English | MEDLINE | ID: mdl-27965383

ABSTRACT

CD47 is known to play an important role in CD4+ T cell homeostasis. We recently reported a reduction in mice deficient in the Cd47 gene (Cd47-/-) CD4+ T cell adhesion and transendothelial migration (TEM) in vivo and in vitro as a result of impaired expression of high-affinity forms of LFA-1 and VLA-4 integrins. A prior study concluded that Cd47-/- mice were resistant to experimental autoimmune encephalomyelitis (EAE) as a result of complete failure in CD4+ T cell activation after myelin oligodendrocyte glycoprotein peptide 35-55 aa (MOG35-55) immunization. As the prior EAE study was published before our report, authors could not have accounted for defects in T cell integrin function as a mechanism to protect Cd47-/- in EAE. Thus, we hypothesized that failure of T cell activation involved defects in LFA-1 and VLA-4 integrins. We confirmed that Cd47-/- mice were resistant to MOG35-55-induced EAE. Our data, however, supported a different mechanism that was not a result of failure of CD4+ T cell activation. Instead, we found that CD4+ T cells in MOG35-55-immunized Cd47-/- mice were activated, but clonal expansion contracted within 72 h after immunization. We used TCR crosslinking and mitogen activation in vitro to investigate the underlying mechanism. We found that naïve Cd47-/- CD4+ T cells exhibited a premature block in proliferation and survival because of impaired activation of LFA-1, despite effective TCR-induced activation. These results identify CD47 as an important regulator of LFA-1 and VLA-4 integrin-adhesive functions in T cell proliferation, as well as recruitment, and clarify the roles played by CD47 in MOG35-55-induced EAE.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD47 Antigen/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Lymphocyte Function-Associated Antigen-1/metabolism , Adoptive Transfer , Animals , Antibodies, Monoclonal/pharmacology , Antigen Presentation/drug effects , Antigen Presentation/immunology , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/drug effects , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Chemokines/pharmacology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Immunization , Integrin alpha4beta1/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lymph Nodes/drug effects , Lymph Nodes/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/immunology , Receptors, Antigen, T-Cell/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
13.
J Clin Invest ; 126(10): 3837-3851, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27599296

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is associated with mutations in the WAS protein (WASp), which plays a critical role in the initiation of T cell receptor-driven (TCR-driven) actin polymerization. The clinical phenotype of WAS includes susceptibility to infection, allergy, autoimmunity, and malignancy and overlaps with the symptoms of dedicator of cytokinesis 8 (DOCK8) deficiency, suggesting that the 2 syndromes share common pathogenic mechanisms. Here, we demonstrated that the WASp-interacting protein (WIP) bridges DOCK8 to WASp and actin in T cells. We determined that the guanine nucleotide exchange factor activity of DOCK8 is essential for the integrity of the subcortical actin cytoskeleton as well as for TCR-driven WASp activation, F-actin assembly, immune synapse formation, actin foci formation, mechanotransduction, T cell transendothelial migration, and homing to lymph nodes, all of which also depend on WASp. These results indicate that DOCK8 and WASp are in the same signaling pathway that links TCRs to the actin cytoskeleton in TCR-driven actin assembly. Further, they provide an explanation for similarities in the clinical phenotypes of WAS and DOCK8 deficiency.


Subject(s)
Actin Cytoskeleton/metabolism , Carrier Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Receptors, Antigen, T-Cell/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Movement , Cytoskeletal Proteins , HEK293 Cells , Humans , Immunological Synapses/metabolism , Lymph Nodes/cytology , Mechanotransduction, Cellular , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Protein Interaction Maps , Protein Multimerization , Protein Transport , T-Lymphocytes/physiology
14.
Nat Commun ; 7: 10363, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26831939

ABSTRACT

Endomucin is a membrane-bound glycoprotein expressed luminally by endothelial cells that line postcapillary venules, a primary site of leukocyte recruitment during inflammation. Here we show that endomucin abrogation on quiescent endothelial cells enables neutrophils to adhere firmly, via LFA-1-mediated binding to ICAM-1 constitutively expressed by endothelial cells. Moreover, TNF-α stimulation downregulates cell surface expression of endomucin concurrent with increased expression of adhesion molecules. Adenovirus-mediated expression of endomucin under inflammatory conditions prevents neutrophil adhesion in vitro and reduces the infiltration of CD45(+) and NIMP-R14(+) cells in vivo. These results indicate that endomucin prevents leukocyte contact with adhesion molecules in non-inflamed tissues and that downregulation of endomucin is critical to facilitate adhesion of leukocytes into inflamed tissues.


Subject(s)
Cell Adhesion/physiology , Endothelial Cells/physiology , Inflammation/metabolism , Leukocytes/physiology , Sialomucins/metabolism , Aged , Animals , Female , Gene Expression Regulation/physiology , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Inbred C57BL , Neutrophils , RNA, Small Interfering , Sialomucins/genetics , Skin/cytology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
Nat Commun ; 6: 10182, 2015 Dec 18.
Article in English | MEDLINE | ID: mdl-26680259

ABSTRACT

The mechanisms driving T cell homing to lymph nodes and migration to tissue are well described but little is known about factors that affect T cell egress from tissues. Here, we generate mice with a T cell-specific deletion of the scaffold protein A kinase anchoring protein 9 (AKAP9) and use models of inflammatory disease to demonstrate that AKAP9 is dispensable for T cell priming and migration into tissues and lymph nodes, but is required for T cell retention in tissues. AKAP9 deficiency results in increased T cell egress to draining lymph nodes, which is associated with impaired T cell re-activation in tissues and protection from organ damage. AKAP9-deficient T cells exhibit reduced microtubule-dependent recycling of TCRs back to the cell surface and this affects antigen-dependent activation, primarily by non-classical antigen-presenting cells. Thus, AKAP9-dependent TCR trafficking drives efficient T cell re-activation and extends their retention at sites of inflammation with implications for disease pathogenesis.


Subject(s)
A Kinase Anchor Proteins/genetics , Cell Movement/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Microtubule-Associated Proteins/genetics , Nephritis/immunology , Reperfusion Injury/immunology , T-Lymphocytes/immunology , A Kinase Anchor Proteins/immunology , Animals , Antigen-Presenting Cells/immunology , Cell Adhesion/immunology , Cell Migration Assays, Leukocyte , Cells, Cultured , Endosomes , Gene Knockout Techniques , Glomerular Basement Membrane/immunology , In Vitro Techniques , Inflammation , Kidney/blood supply , Kidney/immunology , Mice , Microtubule-Associated Proteins/immunology , Receptors, Antigen, T-Cell , Transendothelial and Transepithelial Migration/immunology
16.
Tissue Barriers ; 3(1-2): e1015825, 2015.
Article in English | MEDLINE | ID: mdl-25927017

ABSTRACT

This issue of Tissue Barriers contains the inaugural special issue devoted to recent advances in barrier function of endothelial and epithelial cells. We used this opportunity to invite experts in vascular endothelial cell biology and epithelial cell biology to comment on critical questions and problems in permeability of organ and tissue barriers, and to provide insight into common areas in these fields, namely how these cells maintain homeostasis and response to injury and infection. To complement these reviews, this issue also contains four research articles that explore specific questions related respiratory and intestinal epithelial cell function.

17.
PLoS One ; 10(3): e0121968, 2015.
Article in English | MEDLINE | ID: mdl-25799045

ABSTRACT

Previous studies have demonstrated that the cell surface receptor Slamf1 (CD150) is requisite for optimal NADPH-oxidase (Nox2) dependent reactive oxygen species (ROS) production by phagocytes in response to Gram- bacteria. By contrast, Slamf8 (CD353) is a negative regulator of ROS in response to Gram+ and Gram- bacteria. Employing in vivo migration after skin sensitization, induction of peritonitis, and repopulation of the small intestine demonstrates that in vivo migration of Slamf1-/- dendritic cells and macrophages is reduced, as compared to wt mice. By contrast, in vivo migration of Slamf8-/- dendritic cells, macrophages and neutrophils is accelerated. These opposing effects of Slamf1 and Slamf8 are cell-intrinsic as judged by in vitro migration in transwell chambers in response to CCL19, CCL21 or CSF-1. Importantly, inhibiting ROS production of Slamf8-/- macrophages by diphenyleneiodonium chloride blocks this in vitro migration. We conclude that Slamf1 and Slamf8 govern ROS-dependent innate immune responses of myeloid cells, thus modulating migration of these cells during inflammation in an opposing manner.


Subject(s)
Antigens, CD/physiology , Myeloid Cells/metabolism , Receptors, Cell Surface/physiology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Movement/genetics , Cell Movement/physiology , Chemotaxis/drug effects , Dendritic Cells/cytology , Dendritic Cells/metabolism , Macrophages/cytology , Macrophages/metabolism , Membrane Proteins , Mice , Mice, Inbred BALB C , Myeloid Cells/cytology , Neutrophils/cytology , Neutrophils/metabolism , Peritonitis/metabolism , Peritonitis/pathology , Reactive Oxygen Species/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1
18.
Mol Cell Biol ; 34(23): 4343-54, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25246631

ABSTRACT

The Wiskott-Aldrich syndrome protein (WASp) is important for actin polymerization in T cells and for their migration. WASp-interacting protein (WIP) binds to and stabilizes WASp and also interacts with actin. Cytoskeletal and functional defects are more severe in WIP(-/-) T cells, which lack WASp, than in WASp(-/-) T cells, suggesting that WIP interaction with actin may be important for T cell cytoskeletal integrity and function. We constructed mice that lack the actin-binding domain of WIP (WIPΔABD mice). WIPΔABD associated normally with WASp but not F-actin. T cells from WIPΔABD mice had normal WASp levels but decreased cellular F-actin content, a disorganized actin cytoskeleton, impaired chemotaxis, and defective homing to lymph nodes. WIPΔABD mice exhibited a T cell intrinsic defect in contact hypersensitivity and impaired responses to cutaneous challenge with protein antigen. Adoptively transferred antigen-specific CD4(+) T cells from WIPΔABD mice had decreased homing to antigen-challenged skin of wild-type recipients. These findings show that WIP binding to actin, independently of its binding to WASp, is critical for the integrity of the actin cytoskeleton in T cells and for their migration into tissues. Disruption of WIP binding to actin could be of therapeutic value in T cell-driven inflammatory diseases.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , CD4-Positive T-Lymphocytes/immunology , Carrier Proteins/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/transplantation , Carrier Proteins/genetics , Cell Movement/immunology , Chemokine CCL19/immunology , Chemokine CXCL12/immunology , Cytoskeletal Proteins , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Gene Knock-In Techniques , Hemocyanins/immunology , Inflammation/genetics , Inflammation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Polymerization , Protein Structure, Tertiary/genetics , Receptors, CCR7/immunology , Receptors, CXCR4/immunology , Wiskott-Aldrich Syndrome Protein/genetics
19.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...