Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Circ Res ; 126(10): e61-e79, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32151196

ABSTRACT

RATIONALE: Monocytes are key effectors of the mononuclear phagocyte system, playing critical roles in regulating tissue homeostasis and coordinating inflammatory reactions, including those involved in chronic inflammatory diseases such as atherosclerosis. Monocytes have traditionally been divided into 2 major subsets termed conventional monocytes and patrolling monocytes (pMo) but recent systems immunology approaches have identified marked heterogeneity within these cells, and much of what regulates monocyte population homeostasis remains unknown. We and others have previously identified LYN tyrosine kinase as a key negative regulator of myeloid cell biology; however, LYN's role in regulating specific monocyte subset homeostasis has not been investigated. OBJECTIVE: We sought to comprehensively profile monocytes to elucidate the underlying heterogeneity within monocytes and dissect how Lyn deficiency affects monocyte subset composition, signaling, and gene expression. We further tested the biological significance of these findings in a model of atherosclerosis. METHODS AND RESULTS: Mass cytometric analysis of monocyte subsets and signaling pathway activation patterns in conventional monocytes and pMos revealed distinct baseline signaling profiles and far greater heterogeneity than previously described. Lyn deficiency led to a selective expansion of pMos and alterations in specific signaling pathways within these cells, revealing a critical role for LYN in pMo physiology. LYN's role in regulating pMos was cell-intrinsic and correlated with an increased circulating half-life of Lyn-deficient pMos. Furthermore, single-cell RNA sequencing revealed marked perturbations in the gene expression profiles of Lyn-/- monocytes with upregulation of genes involved in pMo development, survival, and function. Lyn deficiency also led to a significant increase in aorta-associated pMos and protected Ldlr-/- mice from high-fat diet-induced atherosclerosis. CONCLUSIONS: Together our data identify LYN as a key regulator of pMo development and a potential therapeutic target in inflammatory diseases regulated by pMos.


Subject(s)
Atherosclerosis/genetics , Flow Cytometry , Genetic Heterogeneity , Monocytes/enzymology , RNA-Seq , Signal Transduction/genetics , Single-Cell Analysis , Transcriptome , src-Family Kinases/genetics , Animals , Atherosclerosis/enzymology , Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Survival , Cells, Cultured , Cellular Senescence , Disease Models, Animal , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Phenotype , Receptors, LDL/deficiency , Receptors, LDL/genetics , src-Family Kinases/deficiency
2.
J Immunol ; 205(2): 329-334, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32540993

ABSTRACT

A layer of mucus functions to segregate contents of the intestinal lumen from the intestinal epithelium. The MUC2 mucin is the primary constituent of intestinal mucus and plays critical protective roles against luminal microbes and other noxious agents. In this study, we investigated whether MUC2 helps maintain CD8 T cell tolerance toward intestinal luminal Ags by gavaging wild-type and Muc2-/- mice with a model Ag and monitoring immune responses posttreatment. We report that orally delivered OVA rapidly disseminates through the blood of Muc2-/- (but not control) mice and causes immune activation of Ag-specific CD8 T cells at both local and distal sites. Further, the administration of oral OVA to Muc2-/- mice led to its presentation by thymic dendritic cells and the deletion of Ag-specific thymocytes. Collectively, our findings suggest that intestinal mucus helps limit the shaping of the TCR repertoire of developing thymocytes by intestinal luminal Ags.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Intestines/physiology , Mucin-2/metabolism , Mucus/metabolism , Administration, Oral , Animals , Antigens/immunology , Cell Differentiation , Cell Proliferation , Clonal Deletion , Immune Tolerance , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-2/genetics
3.
J Immunol ; 205(4): 945-956, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32641384

ABSTRACT

C-type lectin CLEC16A is located next to CIITA, the master transcription factor of HLA class II (HLA-II), at a susceptibility locus for several autoimmune diseases, including multiple sclerosis (MS). We previously found that CLEC16A promotes the biogenesis of HLA-II peptide-loading compartments (MIICs) in myeloid cells. Given the emerging role of B cells as APCs in these diseases, in this study, we addressed whether and how CLEC16A is involved in the BCR-dependent HLA-II pathway. CLEC16A was coexpressed with surface class II-associated invariant chain peptides (CLIP) in human EBV-positive and not EBV-negative B cell lines. Stable knockdown of CLEC16A in EBV-positive Raji B cells resulted in an upregulation of surface HLA-DR and CD74 (invariant chain), whereas CLIP was slightly but significantly reduced. In addition, IgM-mediated Salmonella uptake was decreased, and MIICs were less clustered in CLEC16A-silenced Raji cells, implying that CLEC16A controls both HLA-DR/CD74 and BCR/Ag processing in MIICs. In primary B cells, CLEC16A was only induced under CLIP-stimulating conditions in vitro and was predominantly expressed in CLIPhigh naive populations. Finally, CLIP-loaded HLA-DR molecules were abnormally enriched, and coregulation with CLEC16A was abolished in blood B cells of patients who rapidly develop MS. These findings demonstrate that CLEC16A participates in the BCR-dependent HLA-II pathway in human B cells and that this regulation is impaired during MS disease onset. The abundance of CLIP already on naive B cells of MS patients may point to a chronically induced stage and a new mechanism underlying B cell-mediated autoimmune diseases such as MS.


Subject(s)
Autoimmunity/immunology , B-Lymphocytes/immunology , Genes, MHC Class II/immunology , Lectins, C-Type/immunology , Monosaccharide Transport Proteins/immunology , Receptors, Antigen, B-Cell/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Autoimmune Diseases/immunology , Cell Line , Cell Line, Tumor , HLA-DR Antigens/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immunoglobulin M/immunology , Multiple Sclerosis/immunology , Signal Transduction/immunology
4.
Nat Chem Biol ; 15(3): 304-313, 2019 03.
Article in English | MEDLINE | ID: mdl-30692685

ABSTRACT

MALT1 paracaspase is central for lymphocyte antigen-dependent responses including NF-κB activation. We discovered nanomolar, selective allosteric inhibitors of MALT1 that bind by displacing the side chain of Trp580, locking the protease in an inactive conformation. Interestingly, we had previously identified a patient homozygous for a MALT1 Trp580-to-serine mutation who suffered from combined immunodeficiency. We show that the loss of tryptophan weakened interactions between the paracaspase and C-terminal immunoglobulin MALT1 domains resulting in protein instability, reduced protein levels and functions. Upon binding of allosteric inhibitors of increasing potency, we found proportionate increased stabilization of MALT1-W580S to reach that of wild-type MALT1. With restored levels of stable MALT1 protein, the most potent of the allosteric inhibitors rescued NF-κB and JNK signaling in patient lymphocytes. Following compound washout, MALT1 substrate cleavage was partly recovered. Thus, a molecular corrector rescues an enzyme deficiency by substituting for the mutated residue, inspiring new potential precision therapies to increase mutant enzyme activity in other deficiencies.


Subject(s)
Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Gene Expression Regulation , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/therapy , Lymphocytes/metabolism , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Male , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/ultrastructure , NF-kappa B/metabolism , Neoplasm Proteins , Signal Transduction
5.
Immunity ; 35(3): 349-60, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21835646

ABSTRACT

The transport of calcium ions (Ca(2+)) to the cytosol is essential for immunoreceptor signaling, regulating lymphocyte differentiation, activation, and effector function. Increases in cytosolic-free Ca(2+) concentrations are thought to be mediated through two interconnected and complementary mechanisms: the release of endoplasmic reticulum Ca(2+) "stores" and "store-operated" Ca(2+) entry via plasma membrane channels. However, the identity of molecular components conducting Ca(2+) currents within developing and mature T cells is unclear. Here, we have demonstrated that the L-type "voltage-dependent" Ca(2+) channel Ca(V)1.4 plays a cell-intrinsic role in the function, development, and survival of naive T cells. Plasma membrane Ca(V)1.4 was found to be essential for modulation of intracellular Ca(2+) stores and T cell receptor (TCR)-induced rises in cytosolic-free Ca(2+), impacting activation of Ras-extracellular signal-regulated kinase (ERK) and nuclear factor of activated T cells (NFAT) pathways. Collectively, these studies revealed that Ca(V)1.4 functions in controlling naive T cell homeostasis and antigen-driven T cell immune responses.


Subject(s)
Calcium Channels/genetics , Calcium Channels/metabolism , Homeostasis , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/immunology , Animals , Calcium/metabolism , Calcium Channels/deficiency , Calcium Channels, L-Type , Cell Proliferation , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout , NFATC Transcription Factors/metabolism , T-Lymphocytes/cytology , ras Proteins/metabolism
6.
J Immunol ; 193(12): 5841-53, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25362182

ABSTRACT

IL-17 plays critical roles in host defenses, combating bacterial and fungal infections, as well as the pathogenesis of autoimmune diseases such as experimental autoimmune encephalomyelitis (EAE). The signaling adaptor SAP is essential for normal immune homeostasis and mutations within SH2D1A, the locus encoding this protein, result in serious and sometimes fatal syndromes, including X-linked lymphoproliferative disease and severe cases of common variable immunodeficiency. However, the precise cellular basis of how SAP deficiency contributes to immune dysfunction remains incompletely understood. In this study, we found that CD4 and CD8 T cells lacking SAP had a diminished capacity to differentiate into IL-17-producing Th17 and T cytotoxic (Tc17) cells relative to wild-type lymphocytes. The use of costimulating SLAM Abs was found to augment the differentiation of IL-17-secreting effectors in wild-type but not Sh2d1a(-/-) splenic T cells under IL-17-polarizing conditions. In addition, SAP's regulation of IL-17-secreting T cells was shown to be a T cell-intrinsic role, as purified naive Sh2d1a(-/-) CD4 and CD8 T cells were inherently defective at converting into Th17 and Tc17 cells in vitro and in vivo. Furthermore, Sh2d1a(-/-) mice were protected from EAE and exhibited greatly decreased numbers of CNS-infiltrating Th17 and Tc17 effector T cells and reduced disease severity. Collectively, these results suggest that SLAM-SAP signaling drives the differentiation and function of Th17 and Tc17 cells in vitro and in vivo and contributes to the pathogenesis of autoimmunity in EAE.


Subject(s)
Antigens, CD/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Antigens, CD/genetics , Cell Differentiation , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/genetics , Gene Expression , Immunization , Immunophenotyping , Interferon-gamma/metabolism , Interleukin-17/biosynthesis , Interleukin-4/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/immunology , Phenotype , Receptors, Cell Surface/genetics , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family Member 1 , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/cytology
7.
J Immunol ; 193(10): 5249-63, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25339668

ABSTRACT

The Lyn tyrosine kinase governs the development and function of various immune cells, and its dysregulation has been linked to malignancy and autoimmunity. Using models of chemically induced colitis and enteric infection, we show that Lyn plays a critical role in regulating the intestinal microbiota and inflammatory responses as well as protection from enteric pathogens. Lyn(-/-) mice were highly susceptible to dextran sulfate sodium (DSS) colitis, characterized by significant wasting, rectal bleeding, colonic pathology, and enhanced barrier permeability. Increased DSS susceptibility in Lyn(-/-) mice required the presence of T but not B cells and correlated with dysbiosis and increased IFN-γ(+) and/or IL-17(+) colonic T cells. This dysbiosis was characterized by an expansion of segmented filamentous bacteria, associated with altered intestinal production of IL-22 and IgA, and was transmissible to wild-type mice, resulting in increased susceptibility to DSS. Lyn deficiency also resulted in an inability to control infection by the enteric pathogens Salmonella enterica serovar Typhimurium and Citrobacter rodentium. Lyn(-/-) mice exhibited profound cecal inflammation, bacterial dissemination, and morbidity following S. Typhimurium challenge and greater colonic inflammation throughout the course of C. rodentium infection. These results identify Lyn as a key regulator of the mucosal immune system, governing pathophysiology in multiple models of intestinal disease.


Subject(s)
Colitis/immunology , Dysbiosis/immunology , Enterobacteriaceae Infections/immunology , Salmonella Infections/immunology , src-Family Kinases/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/microbiology , Citrobacter rodentium/immunology , Citrobacter rodentium/pathogenicity , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Dextran Sulfate , Disease Susceptibility , Dysbiosis/genetics , Dysbiosis/microbiology , Dysbiosis/pathology , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Female , Gene Expression , Immunoglobulin A/genetics , Immunoglobulin A/metabolism , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukins/genetics , Interleukins/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Knockout , Microbiota/immunology , Salmonella Infections/genetics , Salmonella Infections/microbiology , Salmonella Infections/pathology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Severity of Illness Index , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , src-Family Kinases/deficiency , src-Family Kinases/genetics , Interleukin-22
8.
Brain ; 138(Pt 6): 1531-47, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25823473

ABSTRACT

C-type lectins are key players in immune regulation by driving distinct functions of antigen-presenting cells. The C-type lectin CLEC16A gene is located at 16p13, a susceptibility locus for several autoimmune diseases, including multiple sclerosis. However, the function of this gene and its potential contribution to these diseases in humans are poorly understood. In this study, we found a strong upregulation of CLEC16A expression in the white matter of multiple sclerosis patients (n = 14) compared to non-demented controls (n = 11), mainly in perivascular leukocyte infiltrates. Moreover, CLEC16A levels were significantly enhanced in peripheral blood mononuclear cells of multiple sclerosis patients (n = 69) versus healthy controls (n = 46). In peripheral blood mononuclear cells, CLEC16A was most abundant in monocyte-derived dendritic cells, in which it strongly co-localized with human leukocyte antigen class II. Treatment of these professional antigen-presenting cells with vitamin D, a key protective environmental factor in multiple sclerosis, downmodulated CLEC16A in parallel with human leukocyte antigen class II. Knockdown of CLEC16A in distinct types of model and primary antigen-presenting cells resulted in severely impaired cytoplasmic distribution and formation of human leucocyte antigen class II-positive late endosomes, as determined by immunofluorescence and electron microscopy. Mechanistically, CLEC16A participated in the molecular machinery of human leukocyte antigen class II-positive late endosome formation and trafficking to perinuclear regions, involving the dynein motor complex. By performing co-immunoprecipitations, we found that CLEC16A directly binds to two critical members of this complex, RILP and the HOPS complex. CLEC16A silencing in antigen-presenting cells disturbed RILP-mediated recruitment of human leukocyte antigen class II-positive late endosomes to perinuclear regions. Together, we identify CLEC16A as a pivotal gene in multiple sclerosis that serves as a direct regulator of the human leukocyte antigen class II pathway in antigen-presenting cells. These findings are a first step in coupling multiple sclerosis-associated genes to the regulation of the strongest genetic factor in multiple sclerosis, human leukocyte antigen class II.


Subject(s)
Endosomes/metabolism , Genetic Predisposition to Disease/genetics , Histocompatibility Antigens Class II/biosynthesis , Lectins, C-Type/physiology , Monosaccharide Transport Proteins/physiology , Multiple Sclerosis/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adolescent , Adult , Aged , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , Case-Control Studies , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Gene Knockdown Techniques , Humans , Lectins, C-Type/genetics , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/ultrastructure , Male , Middle Aged , Monosaccharide Transport Proteins/genetics , Protein Transport/genetics , RNA, Small Interfering/pharmacology , Up-Regulation/drug effects , Vitamin D/pharmacology , White Matter/metabolism , Young Adult
9.
Blood ; 122(15): 2600-8, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-23974196

ABSTRACT

Individuals with X-linked lymphoproliferative disease lack invariant natural killer T (iNKT) cells and are exquisitely susceptible to Epstein-Barr virus (EBV) infection. To determine whether iNKT cells recognize or regulate EBV, resting B cells were infected with EBV in the presence or absence of iNKT cells. The depletion of iNKT cells increased both viral titers and the frequency of EBV-infected B cells. However, EBV-infected B cells rapidly lost expression of the iNKT cell receptor ligand CD1d, abrogating iNKT cell recognition. To determine whether induced CD1d expression could restore iNKT recognition in EBV-infected cells, lymphoblastoid cell lines (LCL) were treated with AM580, a synthetic retinoic acid receptor-α agonist that upregulates CD1d expression via the nuclear protein, lymphoid enhancer-binding factor 1 (LEF-1). AM580 significantly reduced LEF-1 association at the CD1d promoter region, induced CD1d expression on LCL, and restored iNKT recognition of LCL. CD1d-expressing LCL elicited interferon γ secretion and cytotoxicity by iNKT cells even in the absence of exogenous antigen, suggesting an endogenous iNKT antigen is expressed during EBV infection. These data indicate that iNKT cells may be important for early, innate control of B cell infection by EBV and that downregulation of CD1d may allow EBV to circumvent iNKT cell-mediated immune recognition.


Subject(s)
B-Lymphocytes/immunology , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Lymphoproliferative Disorders/immunology , Natural Killer T-Cells/immunology , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , B-Lymphocytes/virology , Cell Communication/immunology , Cell Transformation, Neoplastic/immunology , Epstein-Barr Virus Infections/complications , Humans , Immunity, Innate/immunology , Lymphoid Enhancer-Binding Factor 1/immunology , Lymphoid Enhancer-Binding Factor 1/metabolism , Lymphoproliferative Disorders/virology , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/virology , Palatine Tonsil/immunology , Palatine Tonsil/virology
10.
J Immunol ; 188(4): 1638-46, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22250093

ABSTRACT

P-selectin glycoprotein ligand-1 (PSGL-1), a heavily glycosylated sialomucin expressed on most leukocytes, has dual function as a selectin ligand for leukocyte rolling on vascular selectins expressed in inflammation and as a facilitator of resting T cell homing into lymphoid organs. In this article, we document disturbances in T cell homeostasis present in PSGL-1(null) mice. Naive CD4(+) and CD8(+) T cell frequencies were profoundly reduced in blood, whereas T cell numbers in lymph nodes and spleen were at or near normal levels. Although PSGL-1(null) T cells were less efficient at entering lymph nodes, they also remained in lymph nodes longer than PSGL-1(+/+) T cells, suggesting that PSGL-1 supports T cell egress. In addition, PSGL-1(null) CD8(+) T cell proliferation was observed under steady-state conditions and PSGL-1(null) CD8(+) T cells were found to be hyperresponsive to homeostatic cytokines IL-2, IL-4, and IL-15. Despite these disturbances in T cell homeostasis, PSGL-1(null) mice exhibited a normal acute response (day 8) to lymphocytic choriomeningitis virus infection but generated an increased frequency of memory T cells (day 40). Our observations demonstrate a novel pleiotropic influence of PSGL-1 deficiency on several aspects of T cell homeostasis that would not have been anticipated based on the mild phenotype of PSGL-1(null) mice. These potentially offsetting effects presumably account for the near-normal cellularity seen in lymph nodes of PSGL-1(null) mice.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/physiology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , Cell Movement , Cells, Cultured , Homeostasis , Interleukin-1/immunology , Interleukin-2/immunology , Interleukin-4/immunology , Lymph Nodes/immunology , Lymphocyte Activation , Lymphocytic choriomeningitis virus/immunology , Membrane Glycoproteins/blood , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL
11.
J Immunol ; 188(10): 5094-105, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22491248

ABSTRACT

The innate immune response is a first line of defense against invading pathogens; however, the magnitude of this response must be tightly regulated, as hyper- or suboptimal responses can be detrimental to the host. Systemic inflammation resulting from bacterial infection can lead to sepsis, which remains a serious problem with high mortality rates. Lyn tyrosine kinase plays a key role in adaptive immunity, although its role in innate immunity remains unclear. In this study, we show that Lyn gain-of-function (Lyn(up/up)) mice display enhanced sensitivity to endotoxin and succumb to upregulated proinflammatory cytokine production at a dose well tolerated by control animals. Endotoxin sensitivity in Lyn(up/up) mice depends on dendritic cells (DCs) and NK cells and occurs though a mechanism involving increased maturation and activation of the DC compartment, leading to elevated production of IFN-γ by NK cells. We further show that modulation of endotoxin-induced signal transduction in DCs by Lyn involves the phosphatases Src homology 2 domain-containing phosphatase-1 and SHIP-1. Collectively, we demonstrate that Lyn regulates DC physiology such that alterations in Lyn-dependent signaling have profound effects on the nature and magnitude of inflammatory responses. Our studies highlight how perturbations in signaling pathways controlling DC/NK cell-regulated responses to microbial products can profoundly affect the magnitude of innate immune responses.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Signal Transduction/immunology , src-Family Kinases/physiology , Animals , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Lymphocyte Activation/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Signal Transduction/genetics , src-Family Kinases/deficiency
12.
J Exp Med ; 204(3): 619-31, 2007 Mar 19.
Article in English | MEDLINE | ID: mdl-17325202

ABSTRACT

Loss of interleukin (IL)-7 or the IL-7 receptor alpha (IL-7Ralpha, CD127) results in severe immunodeficiencies in mice and humans. To more precisely identify signals governing IL-7 function in vivo, we have disrupted the IL-7Ralpha Y449XXM motif in mice by knock-in mutagenesis (IL-7Ralpha(449F)). Thymic precursors were reduced in number in IL-7Ralpha(449F) mice, but in marked contrast to IL-7Ralpha(-/-) knockout mice, thymocytes and peripheral T cells developed normally. Strikingly, Listeria infection revealed that CD4 and CD8 T cells had different requirements for IL-7Ralpha signals. CD4 T cells failed to mount a primary response, but despite normal CD8 primary responses, maintenance of CD8 memory was impaired in IL-7Ralpha(449F) mice. Furthermore, we show that Bcl-2 is IL-7Ralpha Y449 independent and insufficient for IL-7-mediated maintenance of CD8 memory.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/genetics , Receptors, Interleukin-7/genetics , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mutagenesis, Site-Directed , Signal Transduction/genetics
14.
J Immunol ; 187(6): 2898-904, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21844383

ABSTRACT

Autoimmune diabetes in NOD mice can be prevented by application of Ags derived from Mycobacterium tuberculosis in the form of bacillus Calmette-Guérin or CFA. Disease protection by CFA is associated with a reduction in the numbers of pathogenic ß-cell specific, self-reactive CTLs, a phenomenon dependent on the presence and function of NK cells. However, the mechanisms by which NK cells are activated and recruited by heat-killed M. tuberculosis within CFA are unclear. In this study, we report that CFA-mediated NK cell activation and mobilization is dependent on CD1d expression. The administration of M. tuberculosis from CFA results in rapid NKT cell activation and IFN-γ secretion both in vitro and in vivo. CFA-induced NKT cell activation is intact in MyD88(-/-) mice suggesting that the mechanism is independent of TLR signaling. Furthermore, CD1d expression was found to be essential for both M. tuberculosis-triggered NKT cell activation and CFA-mediated protection of NOD mice from diabetes. Collectively, these findings reveal hitherto previously unidentified roles for NKT cells in the adjuvant-promoting effects of CFA on innate and adaptive immunity.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Freund's Adjuvant/immunology , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Animals , Cell Separation , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout
16.
Cell Mol Gastroenterol Hepatol ; 15(2): 425-438, 2023.
Article in English | MEDLINE | ID: mdl-36244647

ABSTRACT

BACKGROUND & AIMS: Increased intestinal permeability is seen in a variety of inflammatory conditions such as enteric infections and inflammatory bowel disease. Because barrier function can provide a key biomarker of disease severity, it often is assayed in animal models. A common methodology involves gavaging mice with fluorescein isothiocyanate-conjugated dextran (FITC-D), followed by cardiac puncture to assay plasma fluorescence on a spectrophotometer. Although the FITC-D method is relatively simple, its sensitivity is limited and enables only a single measurement because the test requires killing the subject. Herein, we describe a novel flow cytometry-based method of intestinal permeability measurement based on detection of orally gavaged ovalbumin (OVA) that leaks out of the gut. Our approach uses minute blood volumes collected from the tail vein, permitting repeated testing of the same subject at multiple time points. By comparing this assay against the gold standard FITC-D method, we show the expanded utility of our OVA assay in measuring intestinal permeability. METHODS: We directly compared our OVA assay against the FITC-D assay by co-administering both probes orally to the same animals and subsequently using their respective methodologies to measure intestinal permeability by detecting probe levels in the plasma. Permeability was assessed in mice genetically deficient in intestinal mucus production or glycosylation. In addition, wild-type mice undergoing dextran sodium sulfate-induced colitis or infected by the enteric bacterial pathogen Citrobacter rodentium also were tested. RESULTS: The OVA assay showed very high efficacy in all animal models of intestinal barrier dysfunction tested. Besides identifying intestinal barrier dysfunction in mice with impaired mucin glycosylation, the assay also allowed for repeated tracking of intestinal permeability within the same animal over time, providing data that cannot be easily acquired with other currently applied methods. CONCLUSIONS: The OVA assay is a highly sensitive and effective method of measuring intestinal permeability in mouse models of barrier dysfunction and experimental colitis.


Subject(s)
Colitis , Dextrans , Mice , Animals , Dextrans/adverse effects , Intestinal Mucosa , Flow Cytometry , Fluorescein-5-isothiocyanate/adverse effects , Colitis/chemically induced , Disease Models, Animal , Permeability
17.
Cancer Res Commun ; 3(3): 404-419, 2023 03.
Article in English | MEDLINE | ID: mdl-36911097

ABSTRACT

While immunotherapies such as immune checkpoint blockade and adoptive T-cell therapy improve survival for a subset of human malignancies, many patients fail to respond. Phagocytes including dendritic cells (DC), monocytes, and macrophages (MF) orchestrate innate and adaptive immune responses against tumors. However, tumor-derived factors may limit immunotherapy effectiveness by altering phagocyte signal transduction, development, and activity. Using Cytometry by Time-of-Flight, we found that tumor-derived GCSF altered myeloid cell distribution both locally and systemically. We distinguished a large number of GCSF-induced immune cell subset and signal transduction pathway perturbations in tumor-bearing mice, including a prominent increase in immature neutrophil/myeloid-derived suppressor cell (Neut/MDSC) subsets and tumor-resident PD-L1+ Neut/MDSCs. GCSF expression was also linked to distinct tumor-associated MF populations, decreased conventional DCs, and splenomegaly characterized by increased splenic progenitors with diminished DC differentiation potential. GCSF-dependent dysregulation of DC development was recapitulated in bone marrow cultures in vitro, using medium derived from GCSF-expressing tumor cell cultures. Importantly, tumor-derived GCSF impaired T-cell adoptive cell therapy effectiveness and was associated with increased tumor volume and diminished survival of mice with mammary cancer. Treatment with neutralizing anti-GCSF antibodies reduced colonic and circulatory Neut/MDSCs, normalized colonic immune cell composition and diminished tumor burden in a spontaneous model of mouse colon cancer. Analysis of human colorectal cancer patient gene expression data revealed a significant correlation between survival and low GCSF and Neut/MDSC gene expression. Our data suggest that normalizing GCSF bioactivity may improve immunotherapy in cancers associated with GCSF overexpression. Significance: Tumor-derived GCSF leads to systemic immune population changes. GCSF blockade restores immune populations, improves immunotherapy, and reduces tumor size, paralleling human colorectal cancer data. GCSF inhibition may synergize with current immunotherapies to treat GCSF-secreting tumors.


Subject(s)
Colonic Neoplasms , Myeloid-Derived Suppressor Cells , Humans , Mice , Animals , Myeloid Cells , Signal Transduction , Lymphocytes, Tumor-Infiltrating , Colonic Neoplasms/metabolism
18.
J Immunol ; 185(7): 3814-8, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20810982

ABSTRACT

CD4(+)FOXP3(+) regulatory T cells are essential for immune tolerance, and murine studies suggest that their dysfunction can lead to type 1 diabetes (T1D). Human studies assessing regulatory T cell dysfunction in T1D have relied on analysis of FOXP3-expressing cells. Recently, distinct subsets of CD4(+)FOXP3(+) T cells with differing function were identified. Notably, CD45RA(-)CD25(int)FOXP3(low) T cells lack suppressive function and secrete the proinflammatory cytokine IL-17. Therefore, we evaluated whether the relative fractions of CD4(+)FOXP3(+) subsets are altered in new-onset T1D subjects. We report that children with new-onset T1D have an increased proportion of CD45RA(-)CD25(int)FOXP3(low) cells that are not suppressive and secrete significantly more IL-17 than other FOXP3(+) subsets. Moreover, these T1D subjects had a higher proportion of both CD4(+) and CD8(+) T cells that secrete IL-17. The bias toward IL-17-secreting T cells in T1D suggests a role for this proinflammatory cytokine in the pathogenesis of disease.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Interleukin-17/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Separation , Child , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/immunology , Humans , Interleukin-17/metabolism , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/immunology , Leukocyte Common Antigens/biosynthesis , Leukocyte Common Antigens/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism
19.
J Immunol ; 184(2): 666-76, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20007535

ABSTRACT

Ag encounter by naive CD8 T cells initiates a developmental program consisting of cellular proliferation, changes in gene expression, and the formation of effector and memory T cells. The strength and duration of TCR signaling are known to be important parameters regulating the differentiation of naive CD8 T cells, although the molecular signals arbitrating these processes remain poorly defined. The Ras-guanyl nucleotide exchange factor RasGRP1 has been shown to transduce TCR-mediated signals critically required for the maturation of developing thymocytes. To elucidate the role of RasGRP1 in CD8 T cell differentiation, in vitro and in vivo experiments were performed with 2C TCR transgenic CD8 T cells lacking RasGRP1. In this study, we report that RasGRP1 regulates the threshold of T cell activation and Ag-induced expansion, at least in part, through the regulation of IL-2 production. Moreover, RasGRP1(-/-) 2C CD8 T cells exhibit an anergic phenotype in response to cognate Ag stimulation that is partially reversible upon the addition of exogenous IL-2. By contrast, the capacity of IL-2/IL-2R interactions to mediate Ras activation and CD8 T cell expansion and differentiation appears to be largely RasGRP1-independent. Collectively, our results demonstrate that RasGRP1 plays a selective role in T cell signaling, controlling the initiation and duration of CD8 T cell immune responses.


Subject(s)
Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Guanine Nucleotide Exchange Factors/physiology , Animals , Cell Differentiation/immunology , Cell Proliferation , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/immunology , Interleukin-2/pharmacology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Signal Transduction/immunology , Transgenes
20.
JCI Insight ; 7(24)2022 12 22.
Article in English | MEDLINE | ID: mdl-36546480

ABSTRACT

Primary atopic disorders are a group of inborn errors of immunity that skew the immune system toward severe allergic disease. Defining the biology underlying these extreme monogenic phenotypes reveals shared mechanisms underlying common polygenic allergic disease and identifies potential drug targets. Germline gain-of-function (GOF) variants in JAK1 are a cause of severe atopy and eosinophilia. Modeling the JAK1GOF (p.A634D) variant in both zebrafish and human induced pluripotent stem cells (iPSCs) revealed enhanced myelopoiesis. RNA-Seq of JAK1GOF human whole blood, iPSCs, and transgenic zebrafish revealed a shared core set of dysregulated genes involved in IL-4, IL-13, and IFN signaling. Immunophenotypic and transcriptomic analysis of patients carrying a JAK1GOF variant revealed marked Th cell skewing. Moreover, long-term ruxolitinib treatment of 2 children carrying the JAK1GOF (p.A634D) variant remarkably improved their growth, eosinophilia, and clinical features of allergic inflammation. This work highlights the role of JAK1 signaling in atopic immune dysregulation and the clinical impact of JAK1/2 inhibition in treating eosinophilic and allergic disease.


Subject(s)
Eosinophilia , Hypersensitivity, Immediate , Hypersensitivity , Induced Pluripotent Stem Cells , Child , Animals , Humans , Gain of Function Mutation , Zebrafish , Hypersensitivity/genetics , Inflammation/genetics , Eosinophilia/genetics , Janus Kinase 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL