Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 20(9)2019 May 10.
Article in English | MEDLINE | ID: mdl-31083300

ABSTRACT

TCDD-inducible poly-ADP-ribose polymerase (TIPARP) is an aryl hydrocarbon receptor (AHR) target gene that functions as part of a negative feedback loop to repress AHR activity. Tiparp-/- mice exhibit increased sensitivity to the toxicological effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), including lethal wasting syndrome. However, it is not known whether Tiparp-/- mice also exhibit increased sensitivity to other AHR ligands. In this study, we treated male Tiparp-/- or wild type (WT) mice with a single injection of 100 mg/kg 3-methylcholanthrene (3MC). Consistent with TIPARP's role as a repressor of AHR signaling, 3MC-treated Tiparp-/- mice exhibited increased hepatic Cyp1a1 and Cyp1b1 levels compared with WT mice. No 3MC-treated Tiparp-/- mice survived beyond day 16 and the mice exhibited chylous ascites characterized by an accumulation of fluid in the peritoneal cavity. All WT mice survived the 30-day treatment and showed no signs of fluid accumulation. Treated Tiparp-/- mice also exhibited a transient and mild hepatotoxicity with inflammation. 3MC-treated WT, but not Tiparp-/- mice, developed mild hepatic steatosis. Lipid deposits accumulated on the surface of the liver and other abdominal organs in the 3MC-Tiparp-/- mice. Our study reveals that Tiparp-/- mice have increased sensitivity to 3MC-induced liver toxicity, but unlike with TCDD, lethality is due to chylous ascites rather than wasting syndrome.


Subject(s)
Chylous Ascites/chemically induced , Chylous Ascites/enzymology , Methylcholanthrene/toxicity , Poly(ADP-ribose) Polymerases/metabolism , Polychlorinated Dibenzodioxins/toxicity , Adipose Tissue/drug effects , Adipose Tissue/pathology , Animals , Azo Compounds/pharmacology , Chylous Ascites/pathology , Cytokines/metabolism , Fatty Liver/enzymology , Fatty Liver/pathology , Gene Expression Regulation, Enzymologic/drug effects , Inflammation/pathology , Inflammation Mediators/metabolism , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Mice, Knockout , Poly(ADP-ribose) Polymerases/genetics , Pyrazoles/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Survival Analysis
2.
J Immunol ; 202(8): 2482-2492, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30867239

ABSTRACT

The TNFR superfamily member 4-1BB is important in the establishment of tissue-resident memory T cells (Trm) in the lung tissue following influenza infection. Moreover, supraphysiological boosting of 4-1BB in the airways during the boost phase of a prime-boost immunization regimen increases the long-lived Trm population, correlating with increased protection against heterotypic challenge. However, little is known about how 4-1BB contributes to the establishment of the lung Trm population. In this study, we show that effects of 4-1BB on lung Trm accumulation are already apparent at the effector stage, suggesting that the major role of 4-1BB in Trm formation is to allow persistence of CD8 T effector cells in the lung as they transition to Trm. Using supraphysiological stimulation of 4-1BB in the boost phase of a prime-boost immunization, we show that the effect of 4-1BB on Trm generation requires local delivery of both Ag and costimulation, is inhibited by rapamycin treatment during secondary CD8 effector T cell expansion, and is dependent on the signaling adaptor TRAF1. The decrease in lung Trm following early rapamycin treatment is accompanied by increased circulating memory T cells, as well as fewer effectors, suggesting a role for mammalian target of rapamycin (mTOR) in the formation of Trm through effects on the accumulation of effector precursors. Taken together, these data point to an important role for 4-1BB, TRAF1, and mTOR in the persistence of CD8 effector T cells in the lung parenchyma, thereby allowing the transition to Trm.


Subject(s)
4-1BB Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Influenza A virus/immunology , Lung Diseases/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , TNF Receptor-Associated Factor 1/immunology , TOR Serine-Threonine Kinases/immunology , 4-1BB Ligand/genetics , Animals , CD8-Positive T-Lymphocytes/pathology , Lung/pathology , Lung/virology , Lung Diseases/genetics , Lung Diseases/pathology , Lung Diseases/virology , Mice , Mice, Knockout , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , TNF Receptor-Associated Factor 1/genetics , TOR Serine-Threonine Kinases/genetics
3.
Mucosal Immunol ; 12(2): 363-377, 2019 03.
Article in English | MEDLINE | ID: mdl-30487647

ABSTRACT

T-cell responses in the lung are critical for protection against respiratory pathogens. TNFR superfamily members play important roles in providing survival signals to T cells during respiratory infections. However, whether these signals take place mainly during priming in the secondary lymphoid organs and/or in the peripheral tissues remains unknown. Here we show that under conditions of competition, GITR provides a T-cell intrinsic advantage to both CD4 and CD8 effector T cells in the lung tissue, as well as for the formation of CD4 and CD8 tissue-resident memory T cells during respiratory influenza infection in mice. In contrast, under non-competitive conditions, GITR has a preferential effect on CD8 over CD4 T cells. The nucleoprotein-specific CD8 T-cell response partially compensated for GITR deficiency by expansion of higher affinity T cells; whereas, the polymerase-specific response was less flexible and more GITR dependent. Following influenza infection, GITR is expressed on lung T cells and GITRL is preferentially expressed on lung monocyte-derived inflammatory antigen presenting cells. Accordingly, we show that GITR+/+ T cells in the lung parenchyma express more phosphorylated-ribosomal protein S6 than their GITR-/- counterparts. Thus, GITR signaling within the lung tissue critically regulates effector and tissue-resident memory T-cell accumulation.


Subject(s)
Antigen-Presenting Cells/immunology , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Influenza A virus/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factors/metabolism , Animals , Cell Differentiation , Cell Movement , Cells, Cultured , Glucocorticoid-Induced TNFR-Related Protein/genetics , Immunologic Memory , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Ribosomal Protein S6 Kinases/metabolism , Tumor Necrosis Factors/genetics
4.
Cell Metab ; 28(6): 922-934.e4, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30174303

ABSTRACT

T cells represent a critical effector of cell-mediated immunity. Activated T cells engage in metabolic reprogramming during effector differentiation to accommodate dynamic changes in energy demands. Here, we show that the hormone, insulin, and downstream signaling through its insulin receptor shape adaptive immune function through modulating T cell metabolism. T cells lacking insulin receptor expression (LckCre+ Insrfl/fl) show reduced antigen-specific proliferation and compromised production of pro-inflammatory cytokines. In vivo, T cell-specific insulin receptor deficiency reduces T cell-driven colonic inflammation. In a model of severe influenza infection with A/PR8 (H1N1), lack of insulin receptor on T cells curtails antigen-specific immunity to influenza viral antigens. Mechanistically, insulin receptor signaling reinforces a metabolic program that supports T cell nutrient uptake and associated glycolytic and respiratory capacities. These data highlight insulin receptor signaling as an important node integrating immunometabolic pathways to drive optimal T cell effector function in health and disease.


Subject(s)
Antigens, CD/immunology , Immunity, Cellular/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Lymphocyte Activation/immunology , Receptor, Insulin/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/genetics , Cytokines/immunology , Cytokines/metabolism , Glycolysis/immunology , Humans , Inflammation/immunology , Inflammation/virology , Insulin/metabolism , Lymph Nodes , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections , Receptor, Insulin/genetics , Signal Transduction , Spleen , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
5.
Immunity ; 47(5): 943-958.e9, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29150240

ABSTRACT

T cell antigen-presenting cell (APC) interactions early during chronic viral infection are crucial for determining viral set point and disease outcome, but how and when different APC subtypes contribute to these outcomes is unclear. The TNF receptor superfamily (TNFRSF) member GITR is important for CD4+ T cell accumulation and control of chronic lymphocytic choriomeningitis virus (LCMV). We found that type I interferon (IFN-I) induced TNFSF ligands GITRL, 4-1BBL, OX40L, and CD70 predominantly on monocyte-derived APCs and CD80 and CD86 predominantly on classical dendritic cells (cDCs). Mice with hypofunctional GITRL in Lyz2+ cells had decreased LCMV-specific CD4+ T cell accumulation and increased viral load. GITR signals in CD4+ T cells occurred after priming to upregulate OX40, CD25, and chemokine receptor CX3CR1. Thus IFN-I (signal 3) induced a post-priming checkpoint (signal 4) for CD4+ T cell accumulation, revealing a division of labor between cDCs and monocyte-derived APCs in regulating T cell expansion.


Subject(s)
Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Tumor Necrosis Factors/analysis , Animals , CD27 Ligand/analysis , CX3C Chemokine Receptor 1/analysis , Dendritic Cells/immunology , Female , Glucocorticoid-Induced TNFR-Related Protein/analysis , Glucocorticoid-Induced TNFR-Related Protein/physiology , Membrane Glycoproteins/analysis , Mice , Mice, Inbred C57BL , Monocytes/cytology , OX40 Ligand
6.
Front Immunol ; 8: 1156, 2017.
Article in English | MEDLINE | ID: mdl-28993768

ABSTRACT

CD30 is a tumor necrosis factor receptor (TNFR) family member whose expression is associated with Hodgkin's disease, anaplastic large cell lymphomas, and other T and B lymphoproliferative disorders in humans. A limited number of studies have assessed the physiological role of CD30/CD30 ligand interactions in control of infection in mice. Here, we assess the role of CD30 in T-cell immunity to acute influenza and chronic lymphocytic choriomeningitis virus (LCMV) clone 13 infection, two viral infections in which other members of the TNFR superfamily are important for T-cell responses. We show that CD30 is expressed on activated but not resting CD4 and CD8 T cells in vitro, as well as on regulatory T cells and marginally on T helper 1 cells in vivo during influenza infection. Despite this, CD4 and CD8 T-cell expansion in response to influenza virus was comparable in CD30+/+ and CD30-/- littermates, with no discernable role for the pathway in the outcome of influenza infection. Similarly, during persistent infection with LCMV clone 13, CD30 plays no obvious role in CD4 or CD8 T-cell responses, the level of T-cell exhaustion or viral control. In contrast, in the steady state, we observed increased numbers of total CD4 and CD8 T cells as well as increased numbers of regulatory T cells in unimmunized older (~8 months) CD30+/+ but not in CD30-/- age-matched littermates. Naive T-cell numbers were unchanged in the aged CD30+/+ mice compared to their CD30-/- littermate controls, rather the T-cell expansions were explained by an increase in CD4+ and CD8+ CD44mid-hiCD62L- effector memory cells, with a similar trend in the central memory T-cell compartment. In contrast, CD30 did not impact the numbers of T cells in young mice. These data suggest a role for CD30 in the homeostatic regulation of T cells during aging, contributing to memory T-cell expansions, which may have relevance for CD30 expression in human T-cell lymphoproliferative diseases.

7.
Nat Immunol ; 17(6): 687-94, 2016 06.
Article in English | MEDLINE | ID: mdl-27089381

ABSTRACT

Aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that mediates the toxic activity of many environmental xenobiotics. However, its role in innate immune responses during viral infection is not fully understood. Here we demonstrate that constitutive AHR signaling negatively regulates the type I interferon (IFN-I) response during infection with various types of virus. Virus-induced IFN-ß production was enhanced in AHR-deficient cells and mice and resulted in restricted viral replication. We found that AHR upregulates expression of the ADP-ribosylase TIPARP, which in turn causes downregulation of the IFN-I response. Mechanistically, TIPARP interacted with the kinase TBK1 and suppressed its activity by ADP-ribosylation. Thus, this study reveals the physiological importance of endogenous activation of AHR signaling in shaping the IFN-I-mediated innate response and, further, suggests that the AHR-TIPARP axis is a potential therapeutic target for enhancing antiviral responses.


Subject(s)
Poly(ADP-ribose) Polymerases/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Virus Diseases/immunology , Animals , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly(ADP-ribose) Polymerases/genetics , RNA, Small Interfering/genetics , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Transcriptional Activation , Virus Replication
8.
PLoS Pathog ; 11(1): e1004517, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25590581

ABSTRACT

CD4 T cells are critical for control of persistent infections; however, the key signals that regulate CD4 T help during chronic infection remain incompletely defined. While several studies have addressed the role of inhibitory receptors and soluble factors such as PD-1 and IL-10, significantly less work has addressed the role of T cell co-stimulatory molecules during chronic viral infection. Here we show that during a persistent infection with lymphocytic choriomeningitis virus (LCMV) clone 13, mice lacking the glucocorticoid-induced tumor necrosis factor receptor related protein (GITR) exhibit defective CD8 T cell accumulation, increased T cell exhaustion and impaired viral control. Differences in CD8 T cells and viral control between GITR+/+ and GITR-/- mice were lost when CD4 T cells were depleted. Moreover, mixed bone marrow chimeric mice, as well as transfer of LCMV epitope-specific CD4 or CD8 T cells, demonstrated that these effects of GITR are largely CD4 T cell-intrinsic. GITR is dispensable for initial CD4 T cell proliferation and differentiation, but supports the post-priming accumulation of IFNγ+IL-2+ Th1 cells, facilitating CD8 T cell expansion and early viral control. GITR-dependent phosphorylation of the p65 subunit of NF-κB as well as phosphorylation of the downstream mTORC1 target, S6 ribosomal protein, were detected at day three post-infection (p.i.), and defects in CD4 T cell accumulation in GITR-deficient T cells were apparent starting at day five p.i. Consistently, we pinpoint IL-2-dependent CD4 T cell help for CD8 T cells to between days four and eight p.i. GITR also increases the ratio of T follicular helper to T follicular regulatory cells and thereby enhances LCMV-specific IgG production. Together, these findings identify a CD4 T cell-intrinsic role for GITR in sustaining early CD8 and late humoral responses to collectively promote control of chronic LCMV clone 13 infection.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/physiology , Glucocorticoid-Induced TNFR-Related Protein/physiology , Lymphopoiesis/genetics , Virus Diseases/immunology , Animals , CD4 Lymphocyte Count , Cell Differentiation/genetics , Cells, Cultured , Chronic Disease , Cricetinae , Female , Immunity, Humoral/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Helper-Inducer/physiology , Virus Diseases/genetics
9.
J Immunol ; 193(10): 5033-43, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25281716

ABSTRACT

The costimulatory TNFR family member GITR can provide important survival signals for CD8 T cells. However, little is known about the regulation of this pathway during a chronic infection. In this study, we show that GITR ligand (GITRL) is maximally induced on APCs at day 2 post-lymphocytic choriomeningitis virus (LCMV) clone 13 infection, but is downregulated to below baseline levels by day 8 postinfection (p.i.), and remains so at the chronic stage of infection. At its peak, GITRL expression is highest on macrophages, with lower expression on conventional and plasmacytoid dendritic cells. GITR expression was highest on T regulatory cells but was also detected on Th1 and LCMV-specific CD8 T cells at day 8 p.i. and was maintained at low, but above baseline levels at the chronic stage of LCMV infection. As GITRL was limiting at the chronic stage of infection, we investigated the potential of therapeutic stimulation of GITR at this stage using agonistic anti-GITR Ab. Anti-GITR treatment at day 21 p.i. increased the frequency and number of LCMV-specific CD8 T cells, resulting in increased in vivo CTL activity and a concomitant decrease in viral load, despite the persistence of PD-1 expression. These effects of anti-GITR were CD8 T cell intrinsic, with no detectable effects on Th1 or T regulatory cells. In contrast to other TNFR agonists, such as anti-4-1BB, which can cause immune pathology, a single therapeutic dose of anti-GITR did not induce splenomegaly or increase serum alanine transaminase. These studies identify GITR as a promising therapeutic target for chronic infection.


Subject(s)
Antibodies/pharmacology , CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/therapy , Lymphocytic choriomeningitis virus/immunology , Tumor Necrosis Factors/agonists , Animals , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Female , Gene Expression Regulation , Host-Pathogen Interactions , Immunotherapy , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/virology , Macrophages/immunology , Macrophages/pathology , Macrophages/virology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/virology , Th1 Cells/immunology , Th1 Cells/pathology , Th1 Cells/virology , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...