Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Annu Rev Immunol ; 29: 665-705, 2011.
Article in English | MEDLINE | ID: mdl-21219180

ABSTRACT

The signaling lymphocyte activation molecule (SLAM)-associated protein, SAP, was first identified as the protein affected in most cases of X-linked lymphoproliferative (XLP) syndrome, a rare genetic disorder characterized by abnormal responses to Epstein-Barr virus infection, lymphoproliferative syndromes, and dysgammaglobulinemia. SAP consists almost entirely of a single SH2 protein domain that interacts with the cytoplasmic tail of SLAM and related receptors, including 2B4, Ly108, CD84, Ly9, and potentially CRACC. SLAM family members are now recognized as important immunomodulatory receptors with roles in cytotoxicity, humoral immunity, autoimmunity, cell survival, lymphocyte development, and cell adhesion. In this review, we cover recent findings on the roles of SLAM family receptors and the SAP family of adaptors, with a focus on their regulation of the pathways involved in the pathogenesis of XLP and other immune disorders.


Subject(s)
Antigens, CD/immunology , Intracellular Signaling Peptides and Proteins/immunology , Animals , Antigens, CD/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/physiopathology , Mutation
2.
Immunity ; 56(7): 1631-1648.e10, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37392737

ABSTRACT

CD137 (4-1BB)-activating receptor represents a promising cancer immunotherapeutic target. Yet, the cellular program driven by CD137 and its role in cancer immune surveillance remain unresolved. Using T cell-specific deletion and agonist antibodies, we found that CD137 modulates tumor infiltration of CD8+-exhausted T (Tex) cells expressing PD1, Lag-3, and Tim-3 inhibitory receptors. T cell-intrinsic, TCR-independent CD137 signaling stimulated the proliferation and the terminal differentiation of Tex precursor cells through a mechanism involving the RelA and cRel canonical NF-κB subunits and Tox-dependent chromatin remodeling. While Tex cell accumulation induced by prophylactic CD137 agonists favored tumor growth, anti-PD1 efficacy was improved with subsequent CD137 stimulation in pre-clinical mouse models. Better understanding of T cell exhaustion has crucial implications for the treatment of cancer and infectious diseases. Our results identify CD137 as a critical regulator of Tex cell expansion and differentiation that holds potential for broad therapeutic applications.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Mice , Animals , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Cell Differentiation , Cell Proliferation , Receptors, Antigen, T-Cell
3.
Nat Immunol ; 19(9): 986-1000, 2018 09.
Article in English | MEDLINE | ID: mdl-30127432

ABSTRACT

Gain-of-function mutations in the gene encoding the phosphatidylinositol-3-OH kinase catalytic subunit p110δ (PI3Kδ) result in a human primary immunodeficiency characterized by lymphoproliferation, respiratory infections and inefficient responses to vaccines. However, what promotes these immunological disturbances at the cellular and molecular level remains unknown. We generated a mouse model that recapitulated major features of this disease and used this model and patient samples to probe how hyperactive PI3Kδ fosters aberrant humoral immunity. We found that mutant PI3Kδ led to co-stimulatory receptor ICOS-independent increases in the abundance of follicular helper T cells (TFH cells) and germinal-center (GC) B cells, disorganized GCs and poor class-switched antigen-specific responses to immunization, associated with altered regulation of the transcription factor FOXO1 and pro-apoptotic and anti-apoptotic members of the BCL-2 family. Notably, aberrant responses were accompanied by increased reactivity to gut bacteria and a broad increase in autoantibodies that were dependent on stimulation by commensal microbes. Our findings suggest that proper regulation of PI3Kδ is critical for ensuring optimal host-protective humoral immunity despite tonic stimulation from the commensal microbiome.


Subject(s)
B-Lymphocytes/physiology , Gastrointestinal Microbiome/immunology , Germinal Center/physiology , Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , T-Lymphocytes, Helper-Inducer/physiology , Animals , Autoantibodies/blood , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases/genetics , Disease Models, Animal , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Humans , Immunity, Humoral/genetics , Immunoglobulin Class Switching/genetics , Immunologic Deficiency Syndromes/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
4.
Nat Immunol ; 15(1): 88-97, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24165795

ABSTRACT

The p110δ subunit of phosphatidylinositol-3-OH kinase (PI(3)K) is selectively expressed in leukocytes and is critical for lymphocyte biology. Here we report fourteen patients from seven families who were heterozygous for three different germline, gain-of-function mutations in PIK3CD (which encodes p110δ). These patients presented with sinopulmonary infections, lymphadenopathy, nodular lymphoid hyperplasia and viremia due to cytomegalovirus (CMV) and/or Epstein-Barr virus (EBV). Strikingly, they had a substantial deficiency in naive T cells but an over-representation of senescent effector T cells. In vitro, T cells from patients exhibited increased phosphorylation of the kinase Akt and hyperactivation of the metabolic checkpoint kinase mTOR, enhanced glucose uptake and terminal effector differentiation. Notably, treatment with rapamycin to inhibit mTOR activity in vivo partially restored the abundance of naive T cells, largely 'rescued' the in vitro T cell defects and improved the clinical course.


Subject(s)
Cellular Senescence/genetics , Germ-Line Mutation , Immunologic Deficiency Syndromes/genetics , Phosphatidylinositol 3-Kinases/genetics , T-Lymphocytes/metabolism , Antibiotics, Antineoplastic/therapeutic use , Cell Differentiation/genetics , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases , Cytomegalovirus Infections/blood , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Epstein-Barr Virus Infections/blood , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Female , Genes, Dominant , Humans , Immunoblotting , Immunologic Deficiency Syndromes/drug therapy , Male , Pedigree , Phosphatidylinositol 3-Kinases/chemistry , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/metabolism , Viremia/drug therapy , Viremia/genetics , Viremia/virology
5.
Immunol Rev ; 291(1): 154-173, 2019 09.
Article in English | MEDLINE | ID: mdl-31402502

ABSTRACT

Phosphatidylinositol 3 kinases (PI3K) are a family of lipid kinases that are activated by a variety of cell-surface receptors, and regulate a wide range of downstream readouts affecting cellular metabolism, growth, survival, differentiation, adhesion, and migration. The importance of these lipid kinases in lymphocyte signaling has recently been highlighted by genetic analyses, including the recognition that both activating and inactivating mutations of the catalytic subunit of PI3Kδ, p110δ, lead to human primary immunodeficiencies. In this article, we discuss how studies on the human genetic disorder "Activated PI3K-delta syndrome" and mouse models of this disease (Pik3cdE1020K/+ mice) have provided fundamental insight into pathways regulated by PI3Kδ in T and B cells and their contribution to lymphocyte function and disease, including responses to commensal bacteria and the development of autoimmunity and tumors. We highlight critical roles of PI3Kδ in T follicular helper cells and the orchestration of the germinal center reaction, as well as in CD8+ T-cell function. We further  present data demonstrating the ability of the AKT-resistant FOXO1AAA mutant to rescue IgG1 class switching defects in Pik3cdE1020K/+ B cells, as well as data supporting a role for PI3Kδ in promoting multiple T-helper effector cell lineages.


Subject(s)
B-Lymphocytes/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Animals , Autoimmunity , B-Lymphocytes/immunology , Biomarkers , Disease Susceptibility , Energy Metabolism , Humans , Immunotherapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/therapy , Primary Immunodeficiency Diseases/etiology , Primary Immunodeficiency Diseases/metabolism , T-Lymphocytes/immunology
6.
Immunity ; 36(6): 1003-16, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22683123

ABSTRACT

X-linked lymphoproliferative syndrome, characterized by fatal responses to Epstein-Barr virus infection, is caused by mutations affecting the adaptor SAP, which links SLAM family receptors to downstream signaling. Although cytotoxic defects in SAP-deficient T cells are documented, the mechanism remains unclear. We show that SAP-deficient murine CD8(+) T cells exhibited normal cytotoxicity against fibrosarcoma targets, yet had impaired adhesion to and killing of B cell and low-avidity T cell targets. SAP-deficient cytotoxic lymphocytes showed specific defects in immunological synapse organization with these targets, resulting in inefficient actin clearance. In the absence of SAP, signaling through the SLAM family members Ly108 and 2B4 resulted in increased recruitment of the SHP-1 phosphatase, associated with altered SHP-1 localization and decreased activation of Src kinases at the synapse. Hence, SAP and SLAM receptors regulate positive and negative signals required for organizing the T cell:B cell synapse and setting thresholds for cytotoxicity against distinct cellular targets.


Subject(s)
Antigens, CD/immunology , Antigens, Ly/immunology , Immunological Synapses/immunology , Intracellular Signaling Peptides and Proteins/immunology , Receptors, Immunologic/immunology , T-Lymphocytes, Cytotoxic/immunology , Actins/analysis , Animals , B-Lymphocytes , Cell Adhesion , Cell Line, Tumor , Centrosome/ultrastructure , Cytoskeleton/ultrastructure , Cytotoxicity, Immunologic , Fibrosarcoma/pathology , Inositol Polyphosphate 5-Phosphatases , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Lymphoma, T-Cell/pathology , Mice , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/physiology , Phosphorylation , Protein Processing, Post-Translational , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family , T-Lymphocytes , src-Family Kinases/metabolism
7.
Immunity ; 36(6): 986-1002, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22683125

ABSTRACT

Humans and mice deficient in the adaptor protein SAP (Sh2d1a) have a major defect in humoral immunity, resulting from a lack of T cell help for B cells. The role of SAP in this process is incompletely understood. We found that deletion of receptor Ly108 (Slamf6) in CD4(+) T cells reversed the Sh2d1a(-/-) phenotype, eliminating the SAP requirement for germinal centers. This potent negative signaling by Ly108 required immunotyrosine switch motifs (ITSMs) and SHP-1 recruitment, resulting in high amounts of SHP-1 at the T cell:B cell synapse, limiting T cell:B cell adhesion. Ly108-negative signaling was important not only in CD4(+) T cells; we found that NKT cell differentiation was substantially restored in Slamf6(-/-)Sh2d1a(-/-) mice. The ability of SAP to regulate both positive and negative signals in T cells can explain the severity of SAP deficiency and highlights the importance of SAP and SHP-1 competition for Ly108 ITSM binding as a rheostat for the magnitude of T cell help to B cells.


Subject(s)
Antigens, Ly/physiology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Lymphocyte Cooperation/physiology , Lymphopoiesis/physiology , Natural Killer T-Cells/cytology , Amino Acid Motifs , Animals , Antigens, Ly/genetics , Germinal Center/immunology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunological Synapses/immunology , Inositol Polyphosphate 5-Phosphatases , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphoric Monoester Hydrolases/physiology , Phosphorylation , Phosphotyrosine/physiology , Protein Processing, Post-Translational , Signaling Lymphocytic Activation Molecule Associated Protein
8.
Nat Immunol ; 9(11): 1279-87, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18806793

ABSTRACT

Mice deficient in the interferon-gamma (IFN-gamma)-inducible, immunity-related GTPase Irgm1 have defective host resistance to a variety of intracellular pathogens. This greater susceptibility to infection is associated with impaired IFN-gamma-dependent macrophage microbicidal activity in vitro. Here we show that Irgm1 also regulated the survival of mature effector CD4(+) T lymphocytes by protecting them from IFN-gamma-induced autophagic cell death. Mice deficient in both IFN-gamma and Irgm1 were 'rescued' from the lymphocyte depletion and greater mortality that occurs in mice singly deficient in Irgm1 after mycobacterial infection. Our studies identify a feedback mechanism in the T helper type 1 response that limits the detrimental effects of IFN-gamma on effector T lymphocyte survival while promoting the antimicrobial functions of IFN-gamma.


Subject(s)
Autophagy , CD4-Positive T-Lymphocytes/immunology , GTP-Binding Proteins/immunology , Interferon-gamma/immunology , Animals , Autophagy/genetics , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/ultrastructure , Cell Proliferation , Cell Survival , Cells, Cultured , Female , GTP-Binding Proteins/genetics , Interferon-gamma/genetics , Interferon-gamma/pharmacology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium avium/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Tuberculosis/immunology
9.
Immunity ; 35(4): 622-32, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22018472

ABSTRACT

Follicular T helper (Tfh) cells provide critical help to B cells for germinal center (GC) formation. Mutations affecting SLAM-associated protein (SAP) prevent GC formation because of defective T cell-B cell interactions, yet effects on Tfh cell differentiation remain unclear. We describe the in vitro differentiation of functionally competent "Tfh-like" cells that expressed interleukin-21, Tfh cell markers, and Bcl6 and rescued GC formation in SAP-deficient hosts better than other T helper (Th) cells. SAP-deficient Tfh-like cells appeared virtually indistinguishable from wild-type, yet failed to support GCs in vivo. Interestingly, both Tfh-like and in vivo-derived Tfh cells could produce effector cytokines in response to polarizing conditions. Moreover, Th1, Th2, and Th17 cells could be reprogrammed to obtain Tfh cell characteristics. ChIP-Seq analyses revealed positive epigenetic markings on Tbx21, Gata3, and Rorc in Tfh-like and ex vivo Tfh cells and on Bcl6 in non-Tfh cells, supporting the concept of plasticity between Tfh and other Th cell populations.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cells, Cultured , DNA-Binding Proteins/immunology , Germinal Center/cytology , Germinal Center/immunology , Interleukins/biosynthesis , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6 , Signaling Lymphocytic Activation Molecule Associated Protein
10.
Immunity ; 34(3): 435-47, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21419664

ABSTRACT

Vitamin A and its metabolite, retinoic acid (RA) are implicated in the regulation of immune homeostasis via the peripheral induction of regulatory T cells. Here we showed RA was also required to elicit proinflammatory CD4(+) helper T cell responses to infection and mucosal vaccination. Retinoic acid receptor alpha (RARα) was the critical mediator of these effects. Antagonism of RAR signaling and deficiency in RARα (Rara(-/-)) resulted in a cell-autonomous CD4(+) T cell activation defect, which impaired intermediate signaling events, including calcium mobilization. Altogether, these findings reveal a fundamental role for the RA-RARα axis in the development of both regulatory and inflammatory arms of adaptive immunity and establish nutritional status as a broad regulator of adaptive T cell responses.


Subject(s)
Adaptive Immunity/immunology , CD4-Positive T-Lymphocytes/immunology , Receptors, Retinoic Acid/immunology , Tretinoin/immunology , Animals , Female , Homeostasis/immunology , Male , Mice , Mice, Inbred C57BL , Retinoic Acid Receptor alpha , Signal Transduction , Toxoplasmosis/immunology
12.
Immunity ; 32(2): 253-65, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20153220

ABSTRACT

CD4(+) T cells deficient in signaling lymphocyte activation molecule (SLAM)-associated protein (SAP) exhibit a selective impairment in adhesion to antigen-presenting B cells but not dendritic cells (DCs), resulting in defective germinal center formation. However, the nature of this selective adhesion defect remained unclear. We found that whereas T cell:DC interactions were primarily integrin dependent, T cell:B cell interactions had both an early integrin-dependent phase and a sustained phase that also required SAP. We further found that the SLAM family member CD84 was required for prolonged T cell:B cell contact, optimal T follicular helper function, and germinal center formation in vivo. Moreover, both CD84 and another SLAM member, Ly108, mediated T cell adhesion and participated in stable T cell:B cell interactions in vitro. Our results reveal insight into the dynamic regulation of T cell:B cell interactions and identify SLAM family members as critical components of sustained T cell:B cell adhesion required for productive humoral immunity.


Subject(s)
B-Lymphocytes/metabolism , Cell Adhesion/immunology , Germinal Center/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , T-Lymphocytes/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Ly/immunology , Antigens, Ly/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Adhesion/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Germinal Center/pathology , Interferon-gamma/metabolism , Interleukins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family , Signaling Lymphocytic Activation Molecule Family Member 1 , T-Lymphocytes/immunology , T-Lymphocytes/pathology
13.
Trends Immunol ; 34(5): 200-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23395212

ABSTRACT

CD4(+) T helper (Th) cells play an instrumental role in orchestrating adaptive immune responses to invading pathogens through their ability to differentiate into specialized effector subsets. Part of this customized response requires the development of T follicular helper (Tfh) cells, which provide help to B cells for the generation of germinal centers (GCs) and long-term protective humoral responses. Although initially viewed as terminally differentiated, we now recognize that Th cell subsets, including Tfh cells, display substantial flexibility and overlap in their characteristics. In this review, we highlight advances in our understanding of Tfh cell development, cytokine production, and the potential plasticity that allows Tfh cells to possess characteristics of other effector Th cell populations.


Subject(s)
B-Lymphocytes/immunology , Cytokines/immunology , Germinal Center/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Differentiation , Cell Lineage , Epigenesis, Genetic/immunology , Humans , Immunologic Memory , Immunomodulation
14.
J Immunol ; 192(5): 2156-66, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24489092

ABSTRACT

CD4(+) T follicular helper cells (TFH) are critical for the formation and function of B cell responses to infection or immunization, but also play an important role in autoimmunity. The factors that contribute to the differentiation of this helper cell subset are incompletely understood, although several cytokines including IL-6, IL-21, and IL-12 can promote TFH cell formation. Yet, none of these factors, nor their downstream cognate STATs, have emerged as nonredundant, essential drivers of TFH cells. This suggests a model in which multiple factors can contribute to the phenotypic characteristics of TFH cells. Because type I IFNs are often generated in immune responses, we set out to investigate whether these factors are relevant to TFH cell differentiation. Type I IFNs promote Th1 responses, thus one possibility was these factors antagonized TFH-expressed genes. However, we show that type I IFNs (IFN-α/ß) induced B cell lymphoma 6 (Bcl6) expression, the master regulator transcription factor for TFH cells, and CXCR5 and programmed cell death-1 (encoded by Pdcd1), key surface molecules expressed by TFH cells. In contrast, type I IFNs failed to induce IL-21, the signature cytokine for TFH cells. The induction of Bcl6 was regulated directly by STAT1, which bound to the Bcl6, Cxcr5, and Pdcd1 loci. These data suggest that type I IFNs (IFN-α/ß) and STAT1 can contribute to some features of TFH cells but are inadequate in inducing complete programming of this subset.


Subject(s)
DNA-Binding Proteins/immunology , Interferon Type I/immunology , STAT1 Transcription Factor/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/immunology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Interferon Type I/biosynthesis , Interferon Type I/genetics , Mice , Mice, Knockout , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Proto-Oncogene Proteins c-bcl-6 , Quantitative Trait Loci/physiology , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism
15.
J Immunol ; 191(4): 1704-15, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23851691

ABSTRACT

Differentiation of CD8 single-positive (SP) T cells is predicated by the ability of lymphocyte progenitors to integrate multiple signaling cues provided by the thymic microenvironment. In the thymus and the OP9-DL1 system for T cell development, Notch signals are required for progenitors to commit to the T cell lineage and necessary for their progression to the CD4(+)CD8(+) double-positive (DP) stage of T cell development. However, it remains unclear whether Notch is a prerequisite for the differentiation of DP cells to the CD8 SP stage of development. In this study, we demonstrate that Notch receptor-ligand interactions allow for efficient differentiation and selection of conventional CD8 T cells from bone marrow-derived hematopoietic stem cells. However, bone marrow-derived hematopoietic stem cells isolated from Itk(-/-)Rlk(-/-) mice gave rise to T cells with decreased IFN-γ production, but gained the ability to produce IL-17. We further reveal that positive and negative selection in vitro are constrained by peptide-MHC class I expressed on OP9 cells. Finally, using an MHC class I-restricted TCR-transgenic model, we show that the commitment of DP precursors to the CD8 T cell lineage is dependent on Notch signaling. Our findings further establish the requirement for Notch receptor-ligand interactions throughout T cell differentiation, including the final step of CD8 SP selection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Clonal Selection, Antigen-Mediated , Intercellular Signaling Peptides and Proteins/immunology , Lymphopoiesis/immunology , Receptors, Notch/physiology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Actins/immunology , Animals , Antigens, Viral/immunology , CD4 Antigens/analysis , CD8 Antigens/analysis , Calcium-Binding Proteins , Cell Lineage , Cells, Cultured , Cellular Microenvironment , Coculture Techniques , Crosses, Genetic , H-2 Antigens/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Histocompatibility Antigen H-2D/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/immunology , Specific Pathogen-Free Organisms , Stromal Cells/cytology , Stromal Cells/immunology
16.
J Immunol ; 190(5): 2121-8, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23355739

ABSTRACT

The promyelocytic zinc finger transcription factor (PLZF) is required for the development of activated phenotypes in NKT and other innate T lymphocytes. Although strong TCR stimulation has been implicated in the induction of PLZF, the factors regulating PLZF expression are incompletely understood. We show in this study that costimulation of preselection double-positive thymocytes through the signaling lymphocyte activation molecule family receptor Ly108 markedly enhanced PLZF expression compared with that induced by TCR stimulation alone. Costimulation with Ly108 increased expression of early growth response protein (Egr)-2 and binding of Egr-2 to the promoter of Zbtb16, which encodes PLZF, and resulted in PLZF levels similar to those seen in NKT cells. In contrast, costimulation with anti-CD28 failed to enhance Egr-2 binding and Zbtb16 expression. Moreover, mice lacking Ly108 showed decreased numbers of PLZF-expressing CD4(+) T cells. Together, these results support a potential role for Ly108 in the induction of PLZF.


Subject(s)
Antigens, Ly/genetics , Cell Differentiation/immunology , Kruppel-Like Transcription Factors/genetics , Thymocytes/cytology , Animals , Antibodies/pharmacology , Antigens, Ly/immunology , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/immunology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Calcium/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/immunology , Gene Expression Regulation/drug effects , Kruppel-Like Transcription Factors/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Transgenic , Promoter Regions, Genetic , Promyelocytic Leukemia Zinc Finger Protein , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction/drug effects , Thymocytes/drug effects , Thymocytes/immunology
17.
N Engl J Med ; 365(7): 611-9, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21793738

ABSTRACT

BACKGROUND: The Proteus syndrome is characterized by the overgrowth of skin, connective tissue, brain, and other tissues. It has been hypothesized that the syndrome is caused by somatic mosaicism for a mutation that is lethal in the nonmosaic state. METHODS: We performed exome sequencing of DNA from biopsy samples obtained from patients with the Proteus syndrome and compared the resultant DNA sequences with those of unaffected tissues obtained from the same patients. We confirmed and extended an observed association, using a custom restriction-enzyme assay to analyze the DNA in 158 samples from 29 patients with the Proteus syndrome. We then assayed activation of the AKT protein in affected tissues, using phosphorylation-specific antibodies on Western blots. RESULTS: Of 29 patients with the Proteus syndrome, 26 had a somatic activating mutation (c.49G→A, p.Glu17Lys) in the oncogene AKT1, encoding the AKT1 kinase, an enzyme known to mediate processes such as cell proliferation and apoptosis. Tissues and cell lines from patients with the Proteus syndrome harbored admixtures of mutant alleles that ranged from 1% to approximately 50%. Mutant cell lines showed greater AKT phosphorylation than did control cell lines. A pair of single-cell clones that were established from the same starting culture and differed with respect to their mutation status had different levels of AKT phosphorylation. CONCLUSIONS: The Proteus syndrome is caused by a somatic activating mutation in AKT1, proving the hypothesis of somatic mosaicism and implicating activation of the PI3K-AKT pathway in the characteristic clinical findings of overgrowth and tumor susceptibility in this disorder. (Funded by the Intramural Research Program of the National Human Genome Research Institute.).


Subject(s)
Mosaicism , Mutation , Proteus Syndrome/genetics , Proto-Oncogene Proteins c-akt/genetics , Child , DNA Mutational Analysis , Exons/genetics , Genotype , Humans , Male , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
18.
J Immunol ; 199(5): 1531-1533, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28827384
19.
Nature ; 455(7214): 764-9, 2008 Oct 09.
Article in English | MEDLINE | ID: mdl-18843362

ABSTRACT

Generation of long-term antibody-mediated immunity depends on the germinal centre reaction, which requires cooperation between antigen-specific T and B lymphocytes. In human X-linked lymphoproliferative disease and its gene-targeted mouse model, loss-of-function mutations in signalling lymphocyte activation molecule-associated protein (SAP, encoded by SH2D1a) cause a profound defect in germinal centre formation by an as yet unknown mechanism. Here, using two-photon intravital imaging, we show that SAP deficiency selectively impairs the ability of CD4(+) T cells to stably interact with cognate B cells but not antigen-presenting dendritic cells. This selective defect results in a failure of antigen-specific B cells to receive adequate levels of contact-dependent T-cell help to expand normally, despite Sap(-/-) T cells exhibiting the known characteristics of otherwise competent helper T cells. Furthermore, the lack of stable interactions with B cells renders Sap(-/-) T cells unable to be efficiently recruited to and retained in a nascent germinal centre to sustain the germinal centre reaction. These results offer an explanation for the germinal centre defect due to SAP deficiency and provide new insights into the bi-directional communication between cognate T and B cells in vivo.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Germinal Center/cytology , Germinal Center/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Cell Adhesion , Cell Communication , Cells, Cultured , Chimera/immunology , Dendritic Cells/immunology , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Lymphocyte Activation , Mice , Signaling Lymphocytic Activation Molecule Associated Protein
20.
PLoS Genet ; 6(10): e1001157, 2010 Oct 14.
Article in English | MEDLINE | ID: mdl-20976248

ABSTRACT

A remarkable characteristic of the human major histocompatibility complex (MHC) is its extreme genetic diversity, which is maintained by balancing selection. In fact, the MHC complex remains one of the best-known examples of natural selection in humans, with well-established genetic signatures and biological mechanisms for the action of selection. Here, we present genetic and functional evidence that another gene with a fundamental role in MHC class I presentation, endoplasmic reticulum aminopeptidase 2 (ERAP2), has also evolved under balancing selection and contains a variant that affects antigen presentation. Specifically, genetic analyses of six human populations revealed strong and consistent signatures of balancing selection affecting ERAP2. This selection maintains two highly differentiated haplotypes (Haplotype A and Haplotype B), with frequencies 0.44 and 0.56, respectively. We found that ERAP2 expressed from Haplotype B undergoes differential splicing and encodes a truncated protein, leading to nonsense-mediated decay of the mRNA. To investigate the consequences of ERAP2 deficiency on MHC presentation, we correlated surface MHC class I expression with ERAP2 genotypes in primary lymphocytes. Haplotype B homozygotes had lower levels of MHC class I expressed on the surface of B cells, suggesting that naturally occurring ERAP2 deficiency affects MHC presentation and immune response. Interestingly, an ERAP2 paralog, endoplasmic reticulum aminopeptidase 1 (ERAP1), also shows genetic signatures of balancing selection. Together, our findings link the genetic signatures of selection with an effect on splicing and a cellular phenotype. Although the precise selective pressure that maintains polymorphism is unknown, the demonstrated differences between the ERAP2 splice forms provide important insights into the potential mechanism for the action of selection.


Subject(s)
Aminopeptidases/genetics , Antigen Presentation , Haplotypes/genetics , Selection, Genetic , Aminopeptidases/immunology , Asian People/genetics , Black People/genetics , Gene Frequency , Genetics, Population , Genotype , Histocompatibility Antigens Class I/immunology , Humans , Indians, North American/genetics , Minor Histocompatibility Antigens , Phylogeny , Polymorphism, Single Nucleotide , Protein Biosynthesis , RNA Processing, Post-Transcriptional , RNA Splicing , White People/genetics
SELECTION OF CITATIONS
SEARCH DETAIL