Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 878
Filter
1.
Eur J Immunol ; 49(12): 2172-2183, 2019 12.
Article in English | MEDLINE | ID: mdl-31339552

ABSTRACT

Aggregation of IgE bound to high affinity IgE receptor (FcεRI) by multivalent antigen induces mast cell activation. Reportedly, disaggregation of aggregated FcεRI immediately terminated degranulation, and formation of co-ligated FcεRI and low affinity IgG receptor FcγRIIB blocked degranulation by inhibitory signal via SH2-containing inositol 5'-phosphatase 1 (SHIP1) phosphorylation. However, their molecular mechanisms to inhibit mast cell activation have been unclear in detail. Herein, we found that addition of excess monomeric hapten (TNP-alanine) to multivalent antigen (TNP-OVA)-activated rat basophilic leukemia cells and mouse bone marrow-derived mast cells induced immediate and transient Syk dephosphorylation, which was previously phosphorylated by TNP-OVA addition. Syk dephosphorylation correlated to rapidly decreased intracellular Ca2+ concentrations ([Ca2+ ]i ), terminated degranulation, and suppressed cytokine production through inhibition of Akt and ERK phosphorylation. Addition of hapten-specific IgG monoclonal antibody (anti-TNP IgG1) to activated mast cells induced translocation of SHIP1 to the plasma membrane and its phosphorylation, indicating that co-ligation of FcεRI and FcγRIIB after FcεRI aggregation can lead to SHIP1 activation. SHIP1 phosphorylation led to gradually decreased [Ca2+ ]i , weak inhibition of degranulation, and strong inhibition of cytokine production. Our findings clearly show the inhibitory mechanism of cell function in activated mast cells by operating Fc receptor crosslinking.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Haptens/immunology , Immunoglobulin G/immunology , Mast Cells/immunology , Animals , Cell Line, Tumor , Immunologic Capping/immunology , Mast Cells/cytology , Mice , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/immunology , Rats , Receptors, IgE/immunology , Receptors, IgG/immunology
2.
MAbs ; 11(6): 996-1011, 2019.
Article in English | MEDLINE | ID: mdl-31156033

ABSTRACT

Agonism of members of the tumor necrosis factor receptor superfamily (TNFRSF) with monoclonal antibodies is of high therapeutic interest due to their role in immune regulation and cell proliferation. A major hurdle for pharmacologic activation of this receptor class is the requirement for high-order clustering, a mechanism that imposes a reliance in vivo on Fc receptor-mediated crosslinking. This extrinsic dependence represents a potential limitation of virtually the entire pipeline of agonist TNFRSF antibody drugs, of which none have thus far been approved or reached late-stage clinical trials. We show that tetravalent biepitopic targeting enables robust intrinsic antibody agonism for two members of this family, OX40 and DR5, that is superior to extrinsically crosslinked native parental antibodies. Tetravalent biepitopic anti-OX40 engagement co-stimulated OX40low cells, obviated the requirement for CD28 co-signal for T cell activation, and enabled superior pharmacodynamic activity relative to native IgG in a murine vaccination model. This work establishes a proof of concept for an engineering approach that addresses a major gap for the therapeutic activation of this important receptor class.


Subject(s)
Antibodies, Monoclonal/immunology , Immunologic Capping , OX40 Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , CD28 Antigens/immunology , CHO Cells , Cricetulus , Humans , Jurkat Cells , Mice , Mice, SCID , Mice, Transgenic , OX40 Ligand/immunology , Receptors, Fc/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , T-Lymphocytes/cytology
3.
Allergol Int ; 68(3): 342-351, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30803853

ABSTRACT

BACKGROUND: The reported prevalences of IgG autoantibodies (AAbs) to FcεRIα and IgE in sera from patients with chronic spontaneous urticaria (CSU) have varied, and these AAbs are also often observed in healthy control subjects. Regarding the histamine release activity of purified IgG from patients with CSU, the number of examined patients has been small. Thus, we sought to determine the prevalence and FcεRI crosslinking ability of these AAbs in a large number of patients with CSU and non-atopic control (NC) subjects. METHODS: We compared the concentrations of anti-IgE and anti-FcεRIα AAbs and the abilities of these AAbs to cause FcεRI aggregation in patients with CSU (n = 134) and NC subjects (n = 55) using ELISA and an in vitro elicitation test, respectively. RESULTS: The concentration of anti-IgE AAbs was significantly different between the NC subjects and the CSU patients (P < 0.0001, cutoff value: 0.558 µg/mL), whereas the concentration of anti-FcεRIα AAbs was not. A significant difference in the duration of illness was noted between patients with lower and those with higher concentrations of anti-IgE AAbs relative to the cutoff value. The abilities of anti-IgE AAbs, but not anti-FcεRIα AAbs, to induce FcεRI crosslinking were significantly higher in CSU patients than in NC subjects (P = 0.0106). CONCLUSIONS: In the Japanese population of CSU patients studied, the ability of the anti-IgE AAbs to induce FcεRI crosslinking differed significantly between NC subjects and CSU patients, suggesting the involvement of anti-IgE AAbs in the pathogenesis of CSU in the Japanese population.


Subject(s)
Autoantibodies/immunology , Immunoglobulin E/immunology , Receptors, IgE/immunology , Urticaria/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Autoantibodies/blood , Basophils/immunology , Cells, Cultured , Chronic Disease , Female , Histamine Release , Humans , Immunoglobulin G/blood , Immunologic Capping , Male , Mast Cells/immunology , Middle Aged , Young Adult
4.
Front Immunol ; 9: 2814, 2018.
Article in English | MEDLINE | ID: mdl-30555482

ABSTRACT

Low affinity receptors for the Fc portion of IgG (FcγRs) represent a critical link between innate and adaptive immunity. Immune complexes (ICs) are the natural ligands for low affinity FcγRs, and high levels of ICs are usually detected in both, chronic viral infections and autoimmune diseases. The expression and function of FcγRs in myeloid cells, NK cells and B cells have been well characterized. By contrast, there are controversial reports about the expression and function of FcγRs in T cells. Here, we demonstrated that ~2% of resting CD4+ T cells express cell surface FcγRII (CD32). Analysis of CD32 expression in permeabilized cells revealed an increased proportion of CD4+CD32+ T cells (~9%), indicating that CD4+ T cells store a CD32 cytoplasmic pool. Activation of CD4+ T cells markedly increased the expression of CD32 either at the cell surface or intracellularly. Analysis of CD32 mRNA transcripts in activated CD4+ T cells revealed the presence of both, the stimulatory FcγRIIa (CD32a) and the inhibitory FcγRIIb (CD32b) isoforms of CD32, being the CD32a:CD32b mRNA ratio ~5:1. Consistent with this finding, we found not only that CD4+ T cells bind aggregated IgG, used as an IC model, but also that CD32 ligation by specific mAb induced a strong calcium transient in CD4+ T cells. Moreover, we found that pretreatment of CD4+ T cells with immobilized IgG as well as cross-linking of CD32 by specific antibodies increased both, the proliferative response of CD4+ T cells and the release of a wide pattern of cytokines (IL-2, IL-5, IL-10, IL-17, IFN-γ, and TNF-α) triggered by either PHA or anti-CD3 mAb. Collectively, our results indicate that ligation of CD32 promotes the activation of CD4+ T cells. These findings suggest that ICs might contribute to the perpetuation of chronic inflammatory responses by virtue of its ability to directly interact with CD4+ T cells through CD32a, promoting the activation of T cells into different inflammatory profiles.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Capping , Receptors, IgG/immunology , Adult , CD4-Positive T-Lymphocytes/cytology , Female , Humans , Lymphocyte Activation , Male
5.
Front Immunol ; 9: 1937, 2018.
Article in English | MEDLINE | ID: mdl-30210494

ABSTRACT

Activation of the high-affinity receptor for IgE (FcεRI) follows a bell-shaped dose-response curve. Upon supra-optimal stimulation, mast cell effector responses are down-regulated by inhibitory molecules like the SH2-containing inositol-5'-phosphatase SHIP1 and the SRC-family-kinase LYN. To identify further molecules involved in a negative regulatory signalosome, we screened for proteins showing the same pattern of tyrosine phosphorylation as SHIP1, which is tyrosine-phosphorylated strongest upon supra-optimal antigen (Ag) stimulation. The low-affinity IgG receptor, FcγRIIB, was found to be most strongly phosphorylated under supra-optimal conditions. This phosphorylation is the consequence of passive, Ag/IgE-dependent and progressive co-localization of FcεRI and FcγRIIB, which is not dependent on IgG. Upon supra-optimal FcεRI cross-linking, FcγRIIB phosphorylation is executed by LYN and protected from dephosphorylation by SHIP1. Analysis of FcγRIIB-deficient bone marrow-derived mast cells revealed an ambiguous phenotype upon FcεRI cross-linking. Absence of FcγRIIB significantly diminished the level of SHIP1 phosphorylation and resulted in augmented Ca2+ mobilization. Though, degranulation and IL-6 production were only weakly altered. Altogether our data establish the LYN/FcγRIIB/SHIP1 signalosome in the context of FcεRI activation, particularly at supra-optimal Ag concentrations. The fact that SHIP1 tyrosine phosphorylation/activation not only depends on FcγRIIB, highlights the necessity for its tight backup control.


Subject(s)
Bone Marrow Cells/immunology , Immunoglobulin G/immunology , Immunologic Capping , Mast Cells/immunology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/immunology , Receptors, IgE/immunology , Receptors, IgG/immunology , src-Family Kinases/immunology , Animals , Bone Marrow Cells/cytology , Calcium Signaling/genetics , Calcium Signaling/immunology , Immunoglobulin G/genetics , Mast Cells/cytology , Mice , Mice, Knockout , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphorylation/genetics , Phosphorylation/immunology , Receptors, IgE/genetics , Receptors, IgG/genetics , src-Family Kinases/genetics
6.
Macromol Biosci ; 18(11): e1800224, 2018 11.
Article in English | MEDLINE | ID: mdl-30259654

ABSTRACT

A therapeutic platform-drug-free macromolecular therapeutics (DFMT)-that induces apoptosis in B cells by cross-linking of CD20 receptors, without the need for low molecular weight cytotoxic drug, is developed. In this report, a DFMT system is synthesized and evaluated based on human serum albumin (HSA) and two complementary coiled-coil forming peptides, CCE and CCK. Fab' fragment of anti-CD20 monoclonal antibody rituximab is attached to CCE (Fab'-CCE); multiple grafts of CCK are conjugated to HSA (HSA-(CCK)7 ). The colocalization of both nanoconjugates at the surface of non-Hodgkin's lymphoma (NHL) Raji cells is demonstrated by confocal fluorescence microscopy. The colocalization leads to coiled-coil formation, CD20 cross-linking, and apoptosis induction. The apoptotic levels are evaluated by Annexin V, Caspase 3, and terminal deoxynucleotidyl transferase dUTP nick end labeling assays. Selective surface binding of DFMT to CD20+ cells is validated in experiments on a coculture of CD20+ (Raji) and CD20-(DG-75) cells. It is found that DFMT can trigger calcium influx only in Raji cells, but not in DG-75 cells. A highly specific treatment for NHL and other B cell malignancies with considerable translational potential is presented by HSA-based DFMT system.


Subject(s)
Apoptosis/drug effects , Immunoglobulin Fab Fragments , Immunologic Capping/drug effects , Lymphoma, B-Cell/drug therapy , Peptides , Rituximab , Serum Albumin, Human , Cell Line, Tumor , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/pharmacology , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Peptides/chemistry , Peptides/pharmacology , Rituximab/chemistry , Rituximab/pharmacology , Serum Albumin, Human/chemistry , Serum Albumin, Human/pharmacology
7.
Cell Death Dis ; 9(5): 544, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29748606

ABSTRACT

CD47 is a cell-surface marker well recognized for its anti-phagocytic functions. As such, an emerging avenue for targeted cancer therapies involves neutralizing the anti-phagocytic function using monoclonal antibodies (mAbs) to enhance tumour cell immunogenicity. A lesser known consequence of CD47 receptor ligation is the direct induction of tumour cell death. While several mAbs and their derivatives with this property have been studied, the best characterized is the commercially available mAb B6H12, which requires immobilization for induction of cell death. Here, we describe a commercially available mAb, CC2C6, which induces T-cell acute lymphoblastic leukemia (ALL) cell death in soluble form. Soluble CC2C6 induces CD47-dependent cell death in a manner consistent with immobilized B6H12, which is characterized by mitochondrial deficiencies but is independent of caspase activation. Titration studies indicated that CC2C6 shares a common CD47-epitope with B6H12. Importantly, CC2C6 retains the anti-phagocytic neutralizing function, thus possessing dual anti-tumour properties. Although CD47-ligation induced cell death occurs in a caspase-independent manner, CC2C6 was found to stimulate increases in Mcl-1 and NOXA levels, two Bcl-2 family proteins that govern the intrinsic apoptosis pathway. Further analysis revealed that the ratio of Mcl-1:NOXA were minimally altered for cells treated with CC2C6, in comparison to cells treated with agents that induced caspase-dependent apoptosis which alter this ratio in favour of NOXA. Finally, we found that CC2C6 can synergize with low dose chemotherapeutic agents that induce classical apoptosis, giving rise to the possibility of an effective combination treatment with reduced long-term sequelae associated with high-dose chemotherapies in childhood ALL.


Subject(s)
Antineoplastic Agents, Immunological/immunology , CD47 Antigen/immunology , Immunologic Capping , Neoplasm Proteins/immunology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Animals , Antineoplastic Agents, Immunological/pharmacology , CD47 Antigen/antagonists & inhibitors , Cell Death/drug effects , Cell Death/immunology , Epitopes/immunology , Humans , Jurkat Cells , Mice , Neoplasm Proteins/antagonists & inhibitors , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology
8.
Methods Mol Biol ; 1707: 171-181, 2018.
Article in English | MEDLINE | ID: mdl-29388107

ABSTRACT

The B cell antigen receptor (BCR) is found to be non-randomly organized at nano-scale distances on the B cell surface. Studying the organization and relocalization of the BCR is thus likely to provide new clues to understand the activation of the BCR. Indeed, with the in situ Fab proximity ligation assay (Fab-PLA), we now obtain proofs for the dissociation activation of BCRs and start to gain insight into how the relocalization of B cell surface signaling molecules could activate the cells. This chapter describes our methods to study the nano-scale organization of B cell surface receptors and co-receptors with Fab-PLA.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin Fab Fragments/chemistry , Immunologic Capping , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , Animals , B-Lymphocytes/cytology , Humans
9.
Discov Med ; 20(108): 79-89, 2015.
Article in English | MEDLINE | ID: mdl-26321090

ABSTRACT

Ankylosing spondylitis (AS) is a chronic, inflammatory arthritis of the spine and peripheral joints linked to the antigen presenting molecule HLA-B27. The risk of AS is increased in patients possessing endoplasmic reticulum aminopeptidase-1 (ERAP1) polymorphisms rs30187 and rs27044 encoding amino acid changes K528R and Q730E, respectively. Dysfunction of ERAP1 is hypothesized to cause changes in expression of HLA-B27 classical (pHLA) and non-classical (FHC) conformers on antigen presenting cells (APCs), which interact with the natural killer (NK) cell receptor KIR3DL1. Dysregulation of this pathway may be pathogenic in AS. APC cell lines expressing HLA-B27 were found to inhibit cytokine production in KIR3DL1+ NK cells due to decreased APC-NK cell adhesion, and possibly activation of receptor down-regulation. Blocking pHLA and FHC reveals that both conformers inhibit cytokine production through KIR3DL1. KIR3DL1 affinity and HLA-B27 surface expression studies suggest that ERAP1 R528 and E730 expression protects from AS by generating sub-optimal pHLA, causing reduced KIR3DL1 affinity and weaker cytokine inhibition. Secondarily we observed that KIR3DL1 binding to C1R-B27 APCs is enhanced by blocking pHLA, but not FHC, raising the possibility that antibody mediated HLA-B27 cross-linking may be important in enhancing KIR3DL1+ NK cell function. This study establishes the role of both FHC and pHLA in modulating NK cell cytokine secretion and adhesion functions by interacting with KIR3DL1. This interaction varies depending on the AS association status of the ERAP1 variant expressed in APCs. Additionally antibody cross-linking of HLA-B27 enhances KIR3DL1 binding and as such could be an important pathogenic mechanism in AS.


Subject(s)
Aminopeptidases/immunology , HLA-B27 Antigen/immunology , Immunologic Capping , Receptors, KIR3DL1/immunology , Spondylitis, Ankylosing/immunology , Aminopeptidases/genetics , Cell Line , Gene Expression Regulation/immunology , HLA-B27 Antigen/genetics , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Minor Histocompatibility Antigens , Polymorphism, Genetic , Receptors, KIR3DL1/genetics , Spondylitis, Ankylosing/genetics , Spondylitis, Ankylosing/pathology
10.
Eur J Immunol ; 45(8): 2286-98, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26046663

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are characterized by an invariant TCRVα7.2 chain recognizing microbial vitamin B metabolites presented by the MHC-Ib molecule MR1. They are mainly detectable in the CD8(+) and CD8(-) CD4(-) "double negative" T-cell compartments of mammals and exhibit both Th1- and Th17-associated features. As MAIT cells show a tissue-homing phenotype and operate at mucosal surfaces with myriads of pathogenic encounters, we wondered how IL-15, a multifaceted cytokine being part of the intestinal mucosal barrier, impacts on their functions. We demonstrate that in the absence of TCR cross-linking, human MAIT cells secrete IFN-γ, increase perforin expression and switch on granzyme B production in response to IL-15. As this mechanism was dependent on the presence of CD14(+) cells and sensitive to IL-18 blockade, we identified IL-15 induced IL-18 production by monocytes as an inflammatory, STAT5-dependent feedback mechanism predominantly activating the MAIT-cell population. IL-15 equally affects TCR-mediated MAIT-cell functions since it dramatically amplifies bacteria-induced IFN-γ secretion, granzyme production, and cytolytic activity at early time points, an effect being most pronounced under suboptimal TCR stimulation conditions. Our data reveal a new quality of IL-15 as player in an inflammatory cytokine network impacting on multiple MAIT-cell functions.


Subject(s)
Interleukin-15/immunology , Interleukin-18/immunology , Lymphocyte Activation/physiology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Female , Humans , Immunologic Capping , Interferon-gamma/immunology , Male , Mucous Membrane/cytology , Mucous Membrane/immunology , T-Lymphocytes/cytology
11.
Eur J Immunol ; 45(3): 705-15, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25471597

ABSTRACT

Depletion of B cells with the anti-CD20 antibody rituximab is an established therapy for rheumatoid arthritis. However, rituximab has only moderate efficacy, most likely due to insufficient depletion of B cells in lymphoid organs and expansion of pathogenic B cells. We found that an antibody against mouse CD79b profoundly blocks B-cell proliferation induced via the B-cell receptor, CD40, CD180, and chondroitin sulfate, but not via TLR4 or TLR9. Treatment with anti-CD79b also induces death in resting and activated B cells. B-cell inhibition is mediated by cross-linkage of CD79b, but independent of Fc-receptor engagement. In the model of collagen-induced arthritis, an antibody against mouse CD20 depletes B cells very efficiently but fails to suppress the humoral immune response against collagen and the development of arthritis. In contrast, the antibody against CD79b, and a deglycosylated variant of this antibody, almost completely inhibits the increase in anti-collagen antibodies and the development of arthritis. In mice with established arthritis only the fully glycosylated antibody against CD79b is effective. Our data show that targeting B cells via CD79b is much more effective than B-cell depletion with anti-CD20 antibodies for therapy of arthritis. These findings may have important implications for treatment of B-cell-mediated autoimmune diseases.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antirheumatic Agents/pharmacology , Arthritis, Experimental/drug therapy , B-Lymphocytes/immunology , CD79 Antigens/antagonists & inhibitors , Lymphocyte Depletion , Animals , Antigens, CD/immunology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , B-Lymphocytes/pathology , CD40 Antigens/immunology , CD79 Antigens/immunology , Cell Proliferation/drug effects , Immunity, Humoral/drug effects , Immunologic Capping/drug effects , Male , Mice , Mice, Inbred DBA , Receptors, Antigen, B-Cell/immunology , Rituximab
12.
MAbs ; 7(1): 199-211, 2015.
Article in English | MEDLINE | ID: mdl-25484043

ABSTRACT

Epratuzumab has demonstrated therapeutic activity in patients with non-Hodgkin lymphoma, acute lymphoblastic leukemia, systemic lupus erythematosus, and Sjögren's syndrome, but its mechanism of affecting normal and malignant B cells remains incompletely understood. We reported previously that epratuzumab displayed in vitro cytotoxicity to CD22-expressing Burkitt lymphoma cell lines (Daudi and Ramos) only when immobilized on plates or combined with a crosslinking antibody plus a suboptimal amount of anti-IgM (1 µg/mL). Herein, we show that, in the absence of additional anti-IgM ligation, extensive crosslinking of CD22 by plate-immobilized epratuzumab induced intracellular changes in Daudi cells similar to ligating B-cell antigen receptor with a sufficiently high amount of anti-IgM (10 µg/mL). Specifically, either treatment led to phosphorylation of CD22, CD79a and CD79b, along with their translocation to lipid rafts, both of which were essential for effecting caspase-dependent apoptosis. Moreover, such immobilization induced stabilization of F-actin, phosphorylation of Lyn, ERKs and JNKs, generation of reactive oxygen species (ROS), decrease in mitochondria membrane potential (Δψm), upregulation of pro-apoptotic Bax, and downregulation of anti-apoptotic Bcl-xl and Mcl-1. The physiological relevance of immobilized epratuzumab was implicated by noting that several of its in vitro effects, including apoptosis, drop in Δψm, and generation of ROS, could be observed with soluble epratuzumab in Daudi cells co-cultivated with human umbilical vein endothelial cells. These results suggest that the in vivo mechanism of non-ligand-blocking epratuzumab may, in part, involve the unmasking of CD22 to facilitate the trans-interaction of B cells with vascular endothelium.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Apoptosis/drug effects , Burkitt Lymphoma/immunology , Caspases/immunology , Immunologic Capping/drug effects , Neoplasm Proteins/immunology , Receptors, Antigen, B-Cell/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 2/antagonists & inhibitors , Signal Transduction/drug effects , Apoptosis/immunology , Burkitt Lymphoma/drug therapy , Cell Line, Tumor , Humans , Membrane Microdomains/immunology , Neoplasm Proteins/antagonists & inhibitors , Protein Transport/drug effects , Protein Transport/immunology , Receptors, Antigen, B-Cell/immunology , Sialic Acid Binding Ig-like Lectin 2/immunology
13.
J Biol Chem ; 289(25): 17647-57, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24817116

ABSTRACT

Natural killer (NK) cell activation is well orchestrated by a wide array of NK cell receptor repertoire. T-cell immunoglobulin and ITIM domain (TIGIT) receptor was recently defined as an inhibitory receptor that is expressed on NK cells and T cells. TIGIT receptor/poliovirus receptor (PVR) ligand engagement signaling inhibits cytotoxicity mediated by NK and CD8(+) T cells. However, it is unclear how TIGIT/PVR signaling regulates cytokine secretion in NK cells. Here we show that TIGIT/PVR engagement suppresses interferon-γ (IFN-γ) production of NK cells. TIGIT transgenic NK cells generate less IFN-γ undergoing TIGIT/PVR ligation. Moreover, TIGIT knock-out NK cells produce much more IFN-γ. TIGIT/PVR ligation signaling mediates suppression of IFN-γ production via the NF-κB pathway. We identified a novel adaptor ß-arrestin 2 that associates with phosphorylated TIGIT for further recruitment of SHIP1 (SH2-containing inositol phosphatase 1) through the ITT-like motif. Importantly, SHIP1, but not other phosphatases, impairs the TNF receptor-associated factor 6 (TRAF6) autoubiquitination to abolish NF-κB activation, leading to suppression of IFN-γ production in NK cells.


Subject(s)
Arrestins/metabolism , Interferon-gamma/biosynthesis , Killer Cells, Natural/metabolism , Receptors, Immunologic/metabolism , Receptors, Virus/metabolism , Signal Transduction/physiology , Animals , Arrestins/genetics , Arrestins/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Humans , Immunologic Capping/physiology , Inositol Polyphosphate 5-Phosphatases , Interferon-gamma/genetics , Interferon-gamma/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Mice , Mice, Knockout , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/immunology , Phosphoric Monoester Hydrolases/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Virus/genetics , Receptors, Virus/immunology , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , beta-Arrestin 2 , beta-Arrestins
14.
J Autoimmun ; 53: 78-84, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24768065

ABSTRACT

The Mertk receptor tyrosine kinase facilitates macrophage and DC apoptotic-cell clearance and regulates immune tolerance. Mertk may also contribute to B-cell activation, because Mertk-KO mice fail to develop autoantibodies when allo-activated by T cells. We investigated this possibility with a well-characterized model in which injection of mice with goat anti-IgD antibody causes membrane IgD cross-linking that induces T-independent B cell activation and antigen presentation to T cells. Goat anti-mouse IgD antibody-injected C57BL/6 Mertk-KO mice had normal initial B cell activation and proliferation, but significantly lower T cell activation and proliferation, as well as lower IgE and IgG anti-goat IgG responses, as compared to C57BL/6 WT controls. B cell antigen processing, analyzed by evaluating B cell fluorescence following injection of monoclonal anti-IgD antibody labeled with biotin or FITC, was comparable between Mertk-KO mice and WT mice. IgD Ab primed B cells from Mertk-KO mice exhibited significantly lower ability in activating memory T cells isolated from WT mice injected with the same antigen 10 days before. These observations suggest that Mertk expression is required for optimal B-cell antigen presentation, which is, in turn, required in this model for optimal T cell activation and subsequent T cell-dependent B cell differentiation.


Subject(s)
B-Lymphocytes/immunology , Cell Communication/immunology , Immunoglobulin D/immunology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Antigen, B-Cell/immunology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/cytology , Cell Communication/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/immunology , Immunoglobulin D/genetics , Immunologic Capping/genetics , Immunologic Capping/immunology , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Antigen, B-Cell/genetics , T-Lymphocytes/cytology , c-Mer Tyrosine Kinase
15.
J Leukoc Biol ; 96(2): 275-82, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24752483

ABSTRACT

Monocyte-derived, fibroblast-like cells, called fibrocytes, participate in wound-healing and the formation of fibrotic lesions. Aggregated or cross-linked IgG are key effectors in infections, autoimmune diseases, anaphylaxis, and immunotherapy. Cells, including monocytes and fibrocytes, bind IgG using FcγRs, and aggregated or cross-linked IgG inhibits fibrocyte differentiation. Mice have four different FcγRs, and which of these, if any, mediate the cross-linked IgG effect on fibrocyte differentiation is unknown. We find that in mice, deletion of FcγRI or the common signaling protein FcRγ significantly reduces the ability of cross-linked IgG or IgG2a to inhibit fibrocyte differentiation. Cells from FcγRIIb/III/IV KO mice are still sensitive to cross-linked IgG, whereas cells from FcγRI/IIb/III/IV KO mice are insensitive to cross-linked IgG. These observations suggest that IgG-mediated inhibition of fibrocyte differentiation is mediated by FcγRs, with FcγRI mediating most of the signaling.


Subject(s)
Cell Differentiation/immunology , Fibroblasts/immunology , Immunoglobulin G/immunology , Immunologic Capping , Receptors, IgG/immunology , Signal Transduction/immunology , Animals , Cell Differentiation/genetics , Fibroblasts/cytology , Mice , Mice, Knockout , Receptors, IgG/genetics , Signal Transduction/genetics
16.
Eur J Immunol ; 44(6): 1802-13, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24610736

ABSTRACT

Along with MHC class I (MHCI), 2B4 provides nonredundant NK-cell inhibition in mice. The immunoregulatory role of 2B4 has been increasingly appreciated in models of tumor and viral infection, however, the interactions among 2B4, MHCI, and other activating NK-cell receptors remain uncertain. Here, we dissect the influence of two distinct inhibitory pathways in modulating NK-cell-mediated control of tumors expressing strong activating ligands, including RAE-1γ. In vitro cytotoxicity and in vivo peritoneal clearance assays using MHCI(+) CD48(+) (RMA-neo), MHCI(+) CD48(+) RAE-1γ (RMA-RAE-1γ), MHCI(-) CD48(+) (RMA-S-neo), and MHCI(-) CD48(+) RAE-1γ (RMA-S-RAE-1γ) tumor lines demonstrated that NKG2D activation supersedes the inhibitory effect of both 2B4- and MHCI-mediated immune-tolerance systems. Furthermore, 2B4KO mice subcutaneously challenged with RMA-neo and RMA-S-neo exhibited reduced tumor growth and significantly prolonged survival compared with WT mice, implying that 2B4 is constitutively engaged in the NK-cell tolerance mechanism in vivo. Nevertheless, the inhibitory effect of 2B4 is significantly attenuated when NK cells encountered highly stressed tumor cells expressing RAE-1γ, resulting in an immune response shift toward NK-cell activation and tumor regression. Therefore, our data highlight the importance of the 2B4-mediated inhibitory system as an alternate self-tolerance mechanism, whose role can be modulated by the strength of activating receptor signaling within the tumor microenvironment.


Subject(s)
Antigens, CD/immunology , Immunologic Capping/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Receptors, Immunologic/immunology , Self Tolerance , Animals , Antigens, CD/genetics , Cell Line, Tumor , Female , Immunologic Capping/genetics , Killer Cells, Natural/pathology , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Receptors, Immunologic/genetics , Signaling Lymphocytic Activation Molecule Family , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
17.
Immunology ; 142(4): 551-61, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24405267

ABSTRACT

Programmed death-ligand 1 (PD-L1) blockade is accepted as a novel strategy for the reactivation of exhausted T cells that express programmed death-1 (PD-1). However, the mechanism of PD-L1-mediated inhibitory signalling after PD-L1 cross-linking by anti-PD-L1 monoclonal antibody (mAb) or PD-1-immunogloblin fusion protein (PD-1-Ig) is still unknown, although it may induce cell death of PD-L1(+) cells required for regular immune reactions. In this study, PD-1-Ig or anti-PD-L1 mAb treatment was tested in cell lines that expressed PD-L1 and bovine lymphocytes to investigate whether the treatment induces immune reactivation or PD-L1-mediated cell death. PD-L1 cross-linking by PD-1-Ig or anti-PD-L1 mAb primarily increased the number of dead cells in PD-L1(high) cells, but not in PD-L1(low) cells; these cells were prepared from Cos-7 cells in which bovine PD-L1 expression was induced by transfection. The PD-L1-mediated cell death also occurred in Cos-7 and HeLa cells transfected with vectors only encoding the extracellular region of PD-L1. In bovine lymphocytes, the anti-PD-L1 mAb treatment up-regulated interferon-γ (IFN-γ) production, whereas PD-1-Ig treatment decreased this cytokine production and cell proliferation. The IFN-γ production in B-cell-depleted peripheral blood mononuclear cells was not reduced by PD-1-Ig treatment and the percentages of dead cells in PD-L1(+) B cells were increased by PD-1-Ig treatment, indicating that PD-1-Ig-induced immunosuppression in bovine lymphocytes could be caused by PD-L1-mediated B-cell death. This study provides novel information for the understanding of signalling through PD-L1.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , B-Lymphocytes/immunology , B7-H1 Antigen/immunology , Immune Tolerance/drug effects , Immunologic Capping/drug effects , Animals , B7-H1 Antigen/genetics , CHO Cells , COS Cells , Cattle , Cell Death/drug effects , Cell Death/immunology , Chlorocebus aethiops , Cricetinae , Cricetulus , HeLa Cells , Humans , Interferon-gamma/immunology
18.
Eur J Immunol ; 43(12): 3175-82, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24019170

ABSTRACT

Soluble TRAIL (sTRAIL) can be produced by myeloid-derived cells to kill cancer cells. Whether this mechanism is used by T cells, and if so, how sTRAIL production is regulated, remains unclear. Our previous studies showed that ex vivo expanded human γδ T cells express TRAIL and NK receptor group 2 (R2), member D (NKG2D), and possess potent anticancer activities both in vitro and in vivo. Here, we investigated in greater detail the mechanisms by which γδ T cells utilize TRAIL and NKG2D to kill lung cancer cells. We demonstrate that human lung cancer cells express TRAIL R2 and NKG2D ligands. Blocking TRAIL or NKG2D during γδ T-cell-lung cancer cell co-cultures significantly reduced γδ T-cell-mediated cytotoxicity. Cross-linking NKG2D with anti-NKG2D antibody to mimic ligand binding promoted γδ T cells to produce sTRAIL, which induced apoptosis in lung cancer cells through TRAIL R2. Either neutralizing sTRAIL or blocking lung cancer cell TRAIL R2 significantly reduced γδ T-cell-mediated cytotoxicity to lung cancer cells. This study demonstrates that γδ T cells can mediate anticancer immunity via NKG2D-regulated production of sTRAIL.


Subject(s)
Immunity, Cellular , Lung Neoplasms/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , TNF-Related Apoptosis-Inducing Ligand/immunology , Antibodies/pharmacology , Apoptosis/drug effects , Apoptosis/immunology , Cell Line, Tumor , Coculture Techniques , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Immunologic Capping/drug effects , Lung Neoplasms/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , T-Lymphocytes/pathology
19.
J Biol Chem ; 288(43): 31139-53, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24022490

ABSTRACT

Humans and other higher primates are unique among mammals in using complement receptor 1 (CR1, CD35) on red blood cells (RBC) to ligate complement-tagged inflammatory particles (immune complexes, apoptotic/necrotic debris, and microbes) in the circulation for quiet transport to the sinusoids of spleen and liver where resident macrophages remove the particles, but allow the RBC to return unharmed to the circulation. This process is called immune-adherence clearance. In this study we found using luminometric- and fluorescence-based methods that ligation of CR1 on human RBC promotes ATP release. Our data show that CR1-mediated ATP release does not depend on Ca(2+) or enzymes previously shown to mediate an increase in membrane deformability promoted by CR1 ligation. Furthermore, ATP release following CR1 ligation increases the mobility of the lipid fraction of RBC membranes, which in turn facilitates CR1 clustering, and thereby enhances the binding avidity of complement-opsonized particles to the RBC CR1. Finally, we have found that RBC-derived ATP has a stimulatory effect on phagocytosis of immune-adherent immune complexes.


Subject(s)
Adenosine Triphosphate/metabolism , Erythrocytes/metabolism , Immunologic Capping , Receptors, Complement 3b/metabolism , Adenosine Triphosphate/immunology , Complement System Proteins/immunology , Complement System Proteins/metabolism , Erythrocytes/cytology , Erythrocytes/immunology , Female , Humans , Male , Membrane Lipids/immunology , Membrane Lipids/metabolism , Phagocytosis/immunology , Receptors, Complement 3b/immunology
20.
J R Soc Interface ; 10(82): 20121048, 2013 May 06.
Article in English | MEDLINE | ID: mdl-23466558

ABSTRACT

The internalization of matter by phagocytosis is of key importance in the defence against bacterial pathogens and in the control of cancerous tumour growth. Despite the fact that phagocytosis is an inherently mechanical process, little is known about the forces and energies that a cell requires for internalization. Here, we use functionalized magnetic particles as phagocytic targets and track their motion while actuating them in an oscillating magnetic field, in order to measure the translational and rotational stiffnesses of the phagocytic cup as a function of time. The measured evolution of stiffness reveals a characteristic pattern with a pronounced peak preceding the finalization of uptake. The measured stiffness values and their time dependence can be interpreted with a model that describes the phagocytic cup as a prestressed membrane connected to an elastically deformable actin cortex. In the context of this model, the stiffness peak is a direct manifestation of a previously described mechanical bottleneck, and a comparison of model and data suggests that the membrane advances around the particle at a speed of about 20 nm s(-1). This approach is a novel way of measuring the progression of emerging phagocytic cups and their mechanical properties in situ and in real time.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Membrane/metabolism , Immunologic Capping/physiology , Magnetite Nanoparticles , Phagocytosis/physiology , Cell Line, Tumor , Elasticity , Humans , Immunologic Capping/drug effects , Phagocytosis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL