Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nat Immunol ; 24(12): 2032-2041, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37945822

ABSTRACT

Cancer cells often overexpress CD47, which triggers the inhibitory receptor SIRPα expressed on macrophages, to elude phagocytosis and antitumor immunity. Pharmacological blockade of CD47 or SIRPα is showing promise as anticancer therapy, although CD47 blockade has been associated with hematological toxicities that may reflect its broad expression pattern on normal cells. Here we found that, in addition to triggering SIRPα, CD47 suppressed phagocytosis by a SIRPα-independent mechanism. This mechanism prevented phagocytosis initiated by the pro-phagocytic ligand, SLAMF7, on tumor cells, due to a cis interaction between CD47 and SLAMF7. The CD47-SLAMF7 interaction was disrupted by CD47 blockade and by a first-in-class agonist SLAMF7 antibody, but not by SIRPα blockade, thereby promoting antitumor immunity. Hence, CD47 suppresses phagocytosis not only by engaging SIRPα, but also by masking cell-intrinsic pro-phagocytic ligands on tumor cells and knowledge of this mechanism may influence the decision between CD47 blockade or SIRPα blockade for therapeutic purposes.


Subject(s)
CD47 Antigen , Neoplasms , Tumor Escape , Humans , Antigens, Differentiation/metabolism , Antigens, Differentiation/therapeutic use , Ligands , Macrophages , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Phagocytosis , Animals , Mice
2.
Nat Immunol ; 20(4): 447-457, 2019 04.
Article in English | MEDLINE | ID: mdl-30833791

ABSTRACT

Invariant natural killer T cells (iNKT cells) develop through an incompletely understood process that requires positive selection by CD4+CD8+ double-positive thymocytes and SLAM family receptors (SFRs). Here we found that SFRs promoted the development of iNKT cells by reducing the strength of the T cell antigen receptor (TCR) signal after positive selection. This effect improved the survival of iNKT cells and their responses to antigen. Loss of SFRs upregulated the expression of inhibitory receptors, including PD-1, on iNKT cells to mitigate the deleterious effect of SFR deficiency. The role of SFRs could be mimicked by expression of SLAMF6 alone in SFR-deficient mice, and this involved the adaptor SAP-kinase Fyn complex and the phosphatase SHP-1. Thus, SFRs foster iNKT cell development by attenuating TCR signal strength after positive selection.


Subject(s)
Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/metabolism , Signaling Lymphocytic Activation Molecule Family/physiology , Animals , Cell Proliferation , Cell Survival , Costimulatory and Inhibitory T-Cell Receptors/metabolism , Humans , Mice , Mice, Knockout , Natural Killer T-Cells/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, Antigen, T-Cell/immunology , Signal Transduction , Signaling Lymphocytic Activation Molecule Associated Protein/genetics , Signaling Lymphocytic Activation Molecule Family/genetics , Signaling Lymphocytic Activation Molecule Family/metabolism
3.
Nat Immunol ; 17(4): 387-96, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26878112

ABSTRACT

Activation of natural killer (NK) cells by hematopoietic target cells is controlled by the SLAM family of receptors and by the associated SAP family of adaptors. Here we found that SLAM receptors also enhanced NK cell activation by nonhematopoietic target cells, which lack ligands for SLAM receptors. This function was mediated by SLAMF6, a homotypic SLAM receptor found on NK cells and other hematopoietic cells, and was regulated by SAP adaptors, which uncoupled SLAM receptors from phosphatase SHP-1 and diminished the effect of SLAMF6 on NK cell responsiveness toward nonhematopoietic cells. Thus, in addition to their role in NK cell activation by hematopoietic cells, the SLAM-SAP pathways influence responsiveness toward nonhematopoietic targets by a process akin to NK cell 'education'.


Subject(s)
Antigens, CD/immunology , Intracellular Signaling Peptides and Proteins/immunology , Killer Cells, Natural/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Receptors, Cell Surface/immunology , Adoptive Transfer , Animals , Cell Line, Tumor , HeLa Cells , Humans , Immunity, Innate , Lymphocyte Activation , Melanoma, Experimental , Mice , Signal Transduction , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family , Signaling Lymphocytic Activation Molecule Family Member 1
4.
Nature ; 544(7651): 493-497, 2017 04 27.
Article in English | MEDLINE | ID: mdl-28424516

ABSTRACT

Cancer cells elude anti-tumour immunity through multiple mechanisms, including upregulated expression of ligands for inhibitory immune checkpoint receptors. Phagocytosis by macrophages plays a critical role in cancer control. Therapeutic blockade of signal regulatory protein (SIRP)-α, an inhibitory receptor on macrophages, or of its ligand CD47 expressed on tumour cells, improves tumour cell elimination in vitro and in vivo, suggesting that blockade of the SIRPα-CD47 checkpoint could be useful in treating human cancer. However, the pro-phagocytic receptor(s) responsible for tumour cell phagocytosis is(are) largely unknown. Here we find that macrophages are much more efficient at phagocytosis of haematopoietic tumour cells, compared with non-haematopoietic tumour cells, in response to SIRPα-CD47 blockade. Using a mouse lacking the signalling lymphocytic activation molecule (SLAM) family of homotypic haematopoietic cell-specific receptors, we determined that phagocytosis of haematopoietic tumour cells during SIRPα-CD47 blockade was strictly dependent on SLAM family receptors in vitro and in vivo. In both mouse and human cells, this function required a single SLAM family member, SLAMF7 (also known as CRACC, CS1, CD319), expressed on macrophages and tumour cell targets. In contrast to most SLAM receptor functions, SLAMF7-mediated phagocytosis was independent of signalling lymphocyte activation molecule-associated protein (SAP) adaptors. Instead, it depended on the ability of SLAMF7 to interact with integrin Mac-1 (refs 18, 19, 20) and utilize signals involving immunoreceptor tyrosine-based activation motifs. These findings elucidate the mechanism by which macrophages engulf and destroy haematopoietic tumour cells. They also reveal a novel SAP adaptor-independent function for a SLAM receptor. Lastly, they suggest that patients with tumours expressing SLAMF7 are more likely to respond to SIRPα-CD47 blockade therapy.


Subject(s)
Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Macrophage-1 Antigen/metabolism , Macrophages/immunology , Phagocytosis/immunology , Signaling Lymphocytic Activation Molecule Family/metabolism , Actins/metabolism , Animals , Antigens, Differentiation/immunology , Antigens, Differentiation/metabolism , CD47 Antigen/immunology , CD47 Antigen/metabolism , Female , Hematologic Neoplasms/drug therapy , Humans , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signaling Lymphocytic Activation Molecule Family/deficiency
5.
Nat Immunol ; 10(9): 973-80, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19648922

ABSTRACT

The adaptors SAP, EAT-2 and ERT are specific to cells of the immune system and belong to the SAP family. All three are expressed in natural killer (NK) cells. Here we examined the global function of the SAP family using mice lacking SAP, EAT-2 and ERT. These adaptors acted together in a mechanism that was essential for the elimination of hematopoietic but not nonhematopoietic cells by NK cells. This function was mediated by many receptors of the SLAM family on NK cells that were engaged by ligands found solely on hematopoietic cells. In the absence of SAP-related adaptors, SLAM receptors lost their activating function and became inhibitory receptors that repressed other activating receptors, such as NKG2D. Hence, the SAP family is essential for the elimination of unwanted hematopoietic cells by NK cells.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Hematopoietic System/cytology , Intracellular Signaling Peptides and Proteins/physiology , Killer Cells, Natural/physiology , Animals , Antigens, CD/physiology , Antigens, Ly/physiology , CD48 Antigen , CHO Cells , Cricetinae , Cricetulus , Histocompatibility Antigens Class I/physiology , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/physiology , Rats , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family , Transcription Factors/physiology
6.
Immunity ; 33(2): 167-80, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20727793

ABSTRACT

PTP-PEST (encoded by Ptpn12) is an intracellular protein tyrosine phosphatase belonging to the same family as LYP. LYP inhibits secondary T cell responses by suppressing Src family protein tyrosine kinases and is implicated in human autoimmunity. To determine the function of PTP-PEST in T cells, we generated mice with a conditionally deleted allele of Ptpn12. By removing PTP-PEST in T cells, we determined that PTP-PEST was not necessary for T cell development or primary responses. However, PTP-PEST was required for secondary T cell responses, anergy prevention, and autoimmunity induction. PTP-PEST specifically regulated the phosphorylation of Pyk2, a substrate of the Src family kinase Fyn. It also promoted the formation of T cell homoaggregates, which are known to enhance T cell activation. Thus, PTP-PEST controls Pyk2 activity and is a positive regulator of secondary T cell activation. These data illustrate the critical role of protein tyrosine phosphatases in T cell regulation.


Subject(s)
Focal Adhesion Kinase 2/immunology , Focal Adhesion Kinase 2/metabolism , Lymphocyte Activation , Protein Tyrosine Phosphatase, Non-Receptor Type 12/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , T-Lymphocytes/immunology , Animals , Cell Aggregation , Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 12/deficiency , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
7.
Blood ; 121(17): 3386-95, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23430111

ABSTRACT

The adaptor molecule signaling lymphocytic activation molecule-associated protein (SAP) plays critical roles during invariant natural killer T (iNKT) cell ontogeny. As a result, SAP-deficient humans and mice lack iNKT cells. The strict developmental requirement for SAP has made it difficult to discern its possible involvement in mature iNKT cell functions. By using temporal Cre recombinase-mediated gene deletion to ablate SAP expression after completion of iNKT cell development, we demonstrate that SAP is essential for T-cell receptor (TCR)-induced iNKT cell cytotoxicity against T-cell and B-cell leukemia targets in vitro and iNKT-cell-mediated control of T-cell leukemia growth in vivo. These findings are not restricted to the murine system: silencing RNA-mediated suppression of SAP expression in human iNKT cells also significantly impairs TCR-induced cytolysis. Mechanistic studies reveal that iNKT cell killing requires the tyrosine kinase Fyn, a known SAP-binding protein. Furthermore, SAP expression is required within iNKT cells to facilitate their interaction with T-cell targets and induce reorientation of the microtubule-organizing center to the immunologic synapse (IS). Collectively, these studies highlight a novel and essential role for SAP during iNKT cell cytotoxicity and formation of a functional IS.


Subject(s)
Cytotoxicity, Immunologic/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Natural Killer T-Cells/pathology , Neoplasms, Experimental/immunology , Synapses/pathology , Animals , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, Antigen, T-Cell/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein , Synapses/immunology , Synapses/metabolism
8.
J Biol Chem ; 288(44): 31423-36, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24045941

ABSTRACT

Signaling lymphocytic activation molecule-associated protein (SAP) is an Src homology 2 domain-only adaptor involved in multiple immune cell functions. It has also been linked to immunodeficiencies and autoimmune diseases, such as systemic lupus erythematosus. Here, we examined the role and mechanism of action of SAP in autoimmunity using a mouse model of autoimmune arthritis, collagen-induced arthritis (CIA). We found that SAP was essential for development of CIA in response to collagen immunization. It was also required for production of collagen-specific antibodies, which play a key role in disease pathogenesis. These effects required SAP expression in T cells, not in B cells. In mice immunized with a high dose of collagen, the activity of SAP was nearly independent of its ability to bind the protein tyrosine kinase Fyn and correlated with the capacity of SAP to promote full differentiation of follicular T helper (TFH) cells. However, with a lower dose of collagen, the role of SAP was more dependent on Fyn binding, suggesting that additional mechanisms other than TFH cell differentiation were involved. Further studies suggested that this might be due to a role of the SAP-Fyn interaction in natural killer T cell development through the ability of SAP-Fyn to promote Vav-1 activation. We also found that removal of SAP expression during progression of CIA attenuated disease severity. However, it had no effect on disease when CIA was clinically established. Together, these results indicate that SAP plays an essential role in CIA because of Fyn-independent and Fyn-dependent effects on TFH cells and, possibly, other T cell types.


Subject(s)
Arthritis, Experimental/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Proto-Oncogene Proteins c-fyn/metabolism , T-Lymphocytes/metabolism , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , B-Lymphocytes , Cell Line , Collagen/toxicity , Gene Expression Regulation/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Mice , Mice, Knockout , Protein Binding , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/immunology , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/immunology , Proto-Oncogene Proteins c-vav/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein , T-Lymphocytes/immunology , T-Lymphocytes/pathology
9.
Cell Rep ; 43(3): 113800, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38386559

ABSTRACT

Infection of mice by mouse cytomegalovirus (MCMV) triggers activation and expansion of Ly49H+ natural killer (NK) cells, which are virus specific and considered to be "adaptive" or "memory" NK cells. Here, we find that signaling lymphocytic activation molecule family receptors (SFRs), a group of hematopoietic cell-restricted receptors, are essential for the expansion of Ly49H+ NK cells after MCMV infection. This activity is largely mediated by CD48, an SFR broadly expressed on NK cells and displaying augmented expression after MCMV infection. It is also dependent on the CD48 counter-receptor, 2B4, expressed on host macrophages. The 2B4-CD48 axis promotes expansion of Ly49H+ NK cells by repressing their phagocytosis by virus-activated macrophages through inhibition of the pro-phagocytic integrin lymphocyte function-associated antigen-1 (LFA-1) on macrophages. These data identify key roles of macrophages and the 2B4-CD48 pathway in controlling the expansion of adaptive NK cells following MCMV infection. Stimulation of the 2B4-CD48 axis may be helpful in enhancing adaptive NK cell responses for therapeutic purposes.


Subject(s)
Cytomegalovirus Infections , Receptors, Immunologic , Animals , Mice , Receptors, Immunologic/metabolism , CD48 Antigen/metabolism , Antigens, CD/metabolism , Lymphocyte Activation , Killer Cells, Natural , Receptors, Cell Surface/metabolism , Carrier Proteins/metabolism , Macrophages/metabolism , Phagocytosis
11.
Immunol Rev ; 232(1): 229-39, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19909367

ABSTRACT

The signaling lymphocytic activation molecule (SLAM)-associated protein (SAP) family of adapters includes SAP, Ewing's sarcoma-associated transcript-2 (EAT-2), and EAT-2-related transducer (ERT). These Src homology-2 (SH2) domain-only molecules play critical roles in immune regulation. The prototype of the SAP family, SAP, is mutated in X-linked lymphoproliferative disease in humans. Moreover, genetically engineered mice lacking one or more SAP family members have defects in multiple immune cell types including T cells, natural killer (NK) cells, NKT cells, and B cells. Accumulating data show that SAP family adapters regulate immunity by influencing the functions of SLAM family receptors, through two distinct but cooperative mechanisms. First, SAP family adapters couple SLAM family receptors to active biochemical signals, which promote immune cell functions. Second, SAP family adapters interfere with the intrinsic ability of SLAM family receptors to trigger inhibitory signals, which could be mediated via molecules such as SH2 domain-containing 5'-inositol phosphatase-1. The latter effect of SAP family adapters does not seem to be because of direct blocking of inhibitory effector binding to SLAM family receptors. Rather, it appears to implicate alternative mechanisms such as functional competition, trans-regulation, or steric hindrance. In the absence of SAP family adapters, the inhibitory signals mediated by SLAM family receptors suppress critical activating receptors, explaining in part the pronounced phenotypes seen in SAP family adapter-deficient humans and mice. Thus, SAP family adapters are molecular switches that regulate immunity as a result of their capacity to control the type of signals and functions emanating from SLAM family receptors.


Subject(s)
Antigens, CD/metabolism , Lymphocytes/metabolism , Lymphoproliferative Disorders/immunology , Mitogen-Activated Protein Kinase 8/metabolism , Receptors, Cell Surface/metabolism , Transcription Factors/metabolism , Allosteric Regulation , Animals , Antigens, CD/immunology , Binding, Competitive , Humans , Lymphocytes/immunology , Lymphocytes/pathology , Lymphoproliferative Disorders/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/immunology , Protein Multimerization/immunology , Receptors, Cell Surface/immunology , Signal Transduction/immunology , Signaling Lymphocytic Activation Molecule Family Member 1 , Transcription Factors/immunology , src Homology Domains/immunology
12.
Sci Immunol ; 7(74): eabn6373, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35930657

ABSTRACT

CD2 is largely described to promote T cell activation when engaged by its ligands, CD48 in mice and CD58 in humans, that are present on antigen-presenting cells (APCs). However, both CD48 and CD58 are also expressed on T cells. By generating new knockout mouse strains lacking CD2 or CD48 in the C57BL/6 background, we determined that whereas CD2 was necessary on T cells for T cell activation, its ligand CD48 was not required on APCs. Rather, CD48 was also needed on T cells. One exception was during cytotoxicity, which required CD48 on T cells and APCs. Fluorescence resonance energy transfer (FRET) studies in nonimmune cells provided evidence that cis interactions between CD2 and CD48 existed within individual cells. CD2-CD48 interactions on T cells enabled more robust T cell receptor (TCR) signals, including protein tyrosine phosphorylation. Using T cells from a CD2 knock-in mouse in which a tag was inserted at the carboxyl terminus of CD2, mass spectrometry analyses revealed that the role of CD2 in T cell activation correlated with its ability to interact with components of the TCR complex and the protein tyrosine kinase Lck. CD2-CD58 provided a similar function in human T cells. Thus, our data imply that T cell-intrinsic cis interactions of CD2 with its ligands are required for TCR signaling and T cell activation. Interactions with ligands on APCs contribute during cytotoxicity.


Subject(s)
Antigens, CD , Lymphocyte Activation , T-Lymphocytes , Animals , CD2 Antigens/chemistry , CD2 Antigens/metabolism , CD48 Antigen/metabolism , CD58 Antigens/metabolism , Humans , Ligands , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/metabolism
13.
Proc Natl Acad Sci U S A ; 105(4): 1273-8, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18212118

ABSTRACT

SAP (also named SH2D1A) is an intracellular adaptor molecule expressed in T cells, natural killer (NK) cells, and some B cells. The SAP gene is mutated in X-linked lymphoproliferative (XLP) disease, a human immunodeficiency characterized by a faulty immune response to Epstein-Barr virus infection. Previous reports documented severe defects in antibody production and germinal center (GC) formation in SAP-deficient humans and mice genetically engineered to lack SAP expression. However, in vitro studies and adoptive transfer experiments provided conflicting data as to whether this phenotype is caused by a functional defect resulting from SAP deficiency in T cells, B cells, or both. Here, we ascertained which cell types are responsible for this humoral immunity defect by using a conditional gene targeting approach. We also thoroughly examined the expression pattern of SAP in normal immune cells by using intracellular flow cytometry. The results showed that expression of SAP in T cells, but not in B cells or NK cells, is required and sufficient for SAP-dependent antibody production and GC formation. These data provide a critical insight into the mechanism by which SAP regulates humoral immunity. They also help elucidate the basis of a severe human immunodeficiency.


Subject(s)
Antibody Formation , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Intracellular Signaling Peptides and Proteins/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antibody Formation/genetics , B-Lymphocytes/pathology , Chickens , Female , Gene Expression Regulation/immunology , Gene Targeting , Haptens/immunology , Haptens/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signaling Lymphocytic Activation Molecule Associated Protein , T-Lymphocytes/pathology , Trinitrobenzenes/immunology , Trinitrobenzenes/metabolism , gamma-Globulins/immunology
14.
Cell Rep ; 37(11): 110111, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34910922

ABSTRACT

Blockade of the inhibitory checkpoint SIRPα-CD47 promotes phagocytosis of cancer cells by macrophages and is a promising avenue in anti-cancer therapy. Productive phagocytosis is strictly predicated on co-engagement of pro-phagocytic receptors-namely, Fc receptors (FcRs), integrin CD11b, or SLAMF7-by their ligands on cancer cells. Here, we examine whether additional pro-phagocytic receptors could be harnessed to broaden the scope of phagocytosis. Inflammatory stimuli, including multiple cytokines and Toll-like receptor (TLR) ligands, augment phagocytosis efficiency and fully alleviate the requirement of FcRs, CD11b, and SLAMF7 for phagocytosis. These effects are mediated by the unconventional pro-phagocytic integrins CD11a and CD11c, which act with CD18 to initiate actin polarization, leading to phagocytosis. Some inflammatory stimuli enable phagocytosis even in the absence of SIRPα-CD47 blockade. Higher CD11c expression in macrophage-enriched tumors correlates with improved survival in clinical studies. Thus, inflammatory macrophages exploit unconventional pro-phagocytic integrins for improved phagocytosis and anti-tumor immunity.


Subject(s)
CD11a Antigen/metabolism , CD11c Antigen/metabolism , Inflammation/immunology , Macrophages/immunology , Peritoneal Neoplasms/prevention & control , Phagocytosis , Signaling Lymphocytic Activation Molecule Family/physiology , Animals , CD11a Antigen/genetics , CD11c Antigen/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology
15.
J Exp Med ; 218(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33237304

ABSTRACT

Expression of the signaling lymphocytic activation molecule (SLAM)-associated protein (SAP) is critical for the germinal center (GC) reaction and T cell-dependent antibody production. However, when SAP is expressed normally, the role of the associated SLAM family receptors (SFRs) in these processes is nebulous. Herein, we established that in the presence of SAP, SFRs suppressed the expansion of the GC reaction but facilitated the generation of antigen-specific B cells and antibodies. SFRs favored the generation of antigen-reactive B cells and antibodies by boosting expression of pro-survival effectors, such as the B cell antigen receptor (BCR) and Bcl-2, in activated GC B cells. The effects of SFRs on the GC reaction and T cell-dependent antibody production necessitated expression of multiple SFRs, both in T cells and in B cells. Hence, while in the presence of SAP, SFRs inhibit the GC reaction, they are critical for the induction of T cell-mediated humoral immunity by enhancing expression of pro-survival effectors in GC B cells.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Germinal Center/cytology , Immunity, Humoral , Signaling Lymphocytic Activation Molecule Family/metabolism , Animals , Antibodies/metabolism , Antibody Formation/immunology , Antigens, Helminth/metabolism , Apoptosis , Bone Marrow/metabolism , Cell Count , Cell Cycle , Cell Proliferation , Cell Survival , Dose-Response Relationship, Immunologic , Immunization , Immunoglobulin Class Switching , Immunologic Memory , Mice, Knockout , Nematospiroides dubius/physiology , Plasma Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Signaling Lymphocytic Activation Molecule Family/deficiency , Somatic Hypermutation, Immunoglobulin , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Vaccination
16.
Blood Adv ; 4(5): 868-879, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32130407

ABSTRACT

Angioimmunoblastic T-cell lymphoma (AITL) is an aggressive peripheral T-cell lymphoma driven by a pool of neoplastic cells originating from T follicular helper (Tfh) cells and concomitant expansion of B cells. Conventional chemotherapies for AITL have shown limited efficacy, and as such, there is a need for improved therapeutic options. Because AITL originates from Tfh cells, we hypothesized that AITL tumors continue to rely on essential Tfh components and intimate T-cell-B-cell (T-B) interactions. Using a spontaneous AITL mouse model (Roquinsan/+ mice), we found that acute loss of Bcl6 activity in growing tumors drastically reduced tumor size, demonstrating that AITL-like tumors critically depend on the Tfh lineage-defining transcription factor Bcl6. Because Bcl6 can upregulate expression of signaling lymphocytic activation molecule-associated protein (SAP), which is known to promote T-B conjugation, we next targeted the SAP-encoding Sh2d1a gene. We observed that Sh2d1a deletion from CD4+ T cells in fully developed tumors also led to tumor regression. Further, we provide evidence that tumor progression depends on T-B cross talk facilitated by SAP and high-affinity LFA-1. In our study, AITL-like tumors relied heavily on molecular pathways that support Tfh cell identity and T-B collaboration, revealing potential therapeutic targets for AITL.


Subject(s)
Immunoblastic Lymphadenopathy , Lymphoma, T-Cell, Peripheral , Animals , Mice , T-Lymphocytes, Helper-Inducer , Transcription Factors
18.
J Exp Med ; 215(12): 3151-3164, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30498080

ABSTRACT

Primary immunodeficiencies represent naturally occurring experimental models to decipher human immunobiology. We report a patient with combined immunodeficiency, marked by recurrent respiratory tract and DNA-based viral infections, hypogammaglobulinemia, and panlymphopenia. He also developed moderate neutropenia but without prototypical pyogenic infections. Using whole-exome sequencing, we identified a homozygous mutation in the inducible T cell costimulator ligand gene (ICOSLG; c.657C>G; p.N219K). Whereas WT ICOSL is expressed at the cell surface, the ICOSLN219K mutation abrogates surface localization: mutant protein is retained in the endoplasmic reticulum/Golgi apparatus, which is predicted to result from deleterious conformational and biochemical changes. ICOSLN219K diminished B cell costimulation of T cells, providing a compelling basis for the observed defect in antibody and memory B cell generation. Interestingly, ICOSLN219K also impaired migration of lymphocytes and neutrophils across endothelial cells, which normally express ICOSL. These defects likely contributed to the altered adaptive immunity and neutropenia observed in the patient, respectively. Our study identifies human ICOSLG deficiency as a novel cause of a combined immunodeficiency.


Subject(s)
Immunologic Deficiency Syndromes , Inducible T-Cell Co-Stimulator Ligand/deficiency , Mutation, Missense , Amino Acid Substitution , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Line, Transformed , Endothelial Cells/immunology , Endothelial Cells/pathology , Female , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/pathology , Immunologic Memory , Inducible T-Cell Co-Stimulator Ligand/immunology , Male , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Whole Genome Sequencing
19.
J Clin Invest ; 110(1): 81-90, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12093891

ABSTRACT

Systemic administration of antigen/peptide for peripheral T cell tolerance has long been investigated as a potential approach to therapy of autoimmune diseases. The multiple antimyelin T cell reactivities likely to be associated with multiple sclerosis (MS) impose major difficulties in devising such an immune-specific therapeutic approach to the disease, because targeting T cells specific for a single autoantigen/epitope is unlikely to be sufficiently effective. Here, we present a pilot study on the possibility of concomitantly inhibiting multiple potentially pathogenic antimyelin T cell reactivities by tolerogenic administration of an artificial "multiantigen/multiepitope" protein. A synthetic gene was constructed to encode selected disease-relevant epitopes of myelin basic protein (MBP), proteolipid protein (PLP), and myelin oligodendrocyte glycoprotein (MOG). The protein product, hmTAP (synthetic human multitarget autoantigen protein), was adequately processed for antigenic presentation of the relevant integral epitopes, in vitro and in vivo. Systemic administration of hmTAP not only suppressed and treated experimental autoimmune encephalomyelitis (EAE) initiated by autoreactivity to a PLP epitope, but also abrogated complex EAE transferred by multispecific line T cells reactive against encephalitogenic epitopes of MBP, PLP, and MOG. These data indicate that multiantigen/multiepitope-directed therapy of complex autoimmune diseases is effective and can be mediated by the protein product of a specifically designed synthetic gene.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Amino Acid Sequence , Animals , Autoantigens/genetics , Base Sequence , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Epitopes/genetics , Female , Humans , Immunosuppression Therapy , Mice , Mice, Inbred C3H , Molecular Sequence Data , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Myelin Basic Protein/genetics , Myelin Basic Protein/immunology , Myelin Proteins , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/immunology , Myelin-Associated Glycoprotein/genetics , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes/immunology
20.
Cell Rep ; 17(10): 2776-2788, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27926878

ABSTRACT

The transmembrane adaptor PAG (Cbp) has been proposed to mediate membrane recruitment of Csk, a cytoplasmic protein tyrosine kinase playing a critical inhibitory role during T cell activation, by inactivating membrane-associated Src kinases. However, this model has not been validated by genetic evidence. Here, we demonstrate that PAG-deficient mice display enhanced T cell activation responses in effector, but not in naive, T cells. PAG-deficient mice also have augmented T cell-dependent autoimmunity and greater resistance to T cell anergy. Interestingly, in the absence of PAG, Csk becomes more associated with alternative partners; i.e., phosphatase PTPN22 and Dok adaptors. Combining PAG deficiency with PTPN22 or Dok adaptor deficiency further enhances effector T cell responses. Unlike PAG, Cbl ubiquitin ligases inhibit the activation of naive, but not of effector, T cells. Thus, Csk-associating PAG is a critical component of the inhibitory machinery controlling effector T cell activation in cooperation with PTPN22 and Dok adaptors.


Subject(s)
DNA-Binding Proteins/genetics , Membrane Proteins/genetics , Phosphoproteins/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Proto-Oncogene Proteins c-cbl/genetics , RNA-Binding Proteins/genetics , T-Lymphocytes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Intercellular Signaling Peptides and Proteins , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , src-Family Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL