Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Nat Immunol ; 24(5): 841-854, 2023 05.
Article in English | MEDLINE | ID: mdl-36928412

ABSTRACT

Regulatory T (Treg) cells are an immunosuppressive population that are required to maintain peripheral tolerance and prevent tissue damage from immunopathology, via anti-inflammatory cytokines, inhibitor receptors and metabolic disruption. Here we show that Treg cells acquire an effector-like state, yet remain stable and functional, when exposed to interferon gamma (IFNγ) during infection with lymphocytic choriomeningitis and influenza A virus. Treg cell-restricted deletion of the IFNγ receptor (encoded by Ifngr1), but not the interleukin 12 (IL12) receptor (encoded by Il12rb2), prevented TH1-like polarization (decreased expression of T-bet, CXC motif chemokine receptor 3 and IFNγ) and promoted TH2-like polarization (increased expression of GATA-3, CCR4 and IL4). TH1-like Treg cells limited CD8+ T cell effector function, proliferation and memory formation during acute and chronic infection. These findings provide fundamental insights into how Treg cells sense inflammatory cues from the environment (such as IFNγ) during viral infection to provide guidance to the effector immune response. This regulatory circuit prevents prolonged immunoinflammatory responses and shapes the quality and quantity of the memory T cell response.


Subject(s)
Interferon-gamma , T-Lymphocytes, Regulatory , Interferon-gamma/metabolism , Cytokines/metabolism , CD8-Positive T-Lymphocytes , Antiviral Agents/metabolism , Th1 Cells
2.
J Immunol ; 212(3): 466-474, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38108417

ABSTRACT

Tim-3 is a transmembrane protein that is best known for being highly expressed on terminally exhausted CD8+ T cells associated with chronic infection and tumors, although its expression is not limited to those settings. Tim-3 is also expressed by CD8+ T cells during acute infection and by multiple other immune cell types, including CD4+ Th1 and regulatory T cells, dendritic cells, and mast cells. In this study, we investigated the role of Tim-3 signaling on CD8+ T cell memory using a Tim-3 conditional knockout mouse model and mice lacking the signaling portion of the Tim-3 cytoplasmic domain. Together, our results indicate that Tim-3 has at most a modest effect on the formation and function of CD8+ memory T cells.


Subject(s)
Lymphocytic Choriomeningitis , Lymphocytic choriomeningitis virus , Animals , Mice , CD8-Positive T-Lymphocytes , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Memory T Cells , Signal Transduction
3.
J Immunol ; 205(8): 2100-2108, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32887751

ABSTRACT

PI3K plays multiple roles throughout the life of a B cell. As such, its signaling is tightly regulated. The importance of this is illustrated by the fact that both loss- and gain-of-function mutations in PI3K can cause immunodeficiency in humans. PIK3IP1, also known as TrIP, is a transmembrane protein that has been shown to inhibit PI3K in T cells. Results from the ImmGen Consortium indicate that PIK3IP1 expression fluctuates throughout B cell development in a manner inversely correlated with PI3K activity; however, its role in B cells is poorly understood. In this study, we define the consequences of B cell-specific deletion of PIK3IP1. B cell development, basal Ig levels, and T-independent responses were unaffected by loss of PIK3IP1. However, there was a significant delay in the production of IgG during T-dependent responses, and secondary responses were impaired. This is likely due to a role for PIK3IP1 in the extrafollicular response because germinal center formation and affinity maturation were normal, and PIK3IP1 is not appreciably expressed in germinal center B cells. Consistent with a role early in the response, PIK3IP1 was downregulated at late time points after B cell activation, in a manner dependent on PI3K. Increased activation of the PI3K pathway was observed in PIK3IP1-deficient B cells in response to engagement of both the BCR and CD40 or strong cross-linking of CD40 alone. Taken together, these observations suggest that PIK3IP1 promotes extrafollicular responses by limiting PI3K signaling during initial interactions between B and T cells.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Immunity, Cellular , Immunoglobulin Class Switching , Intracellular Signaling Peptides and Proteins/immunology , Membrane Proteins/immunology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/cytology , CD40 Antigens/genetics , CD40 Antigens/immunology , Germinal Center/cytology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , T-Lymphocytes/cytology
4.
Biochem J ; 478(22): 3999-4004, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34813649

ABSTRACT

Tim-3 is a transmembrane protein that is highly expressed on subsets of chronically stimulated CD4+ helper and CD8+ cytotoxic T cells, with more transient expression during acute activation and infection. Tim-3 is also constitutively expressed by multiple types of myeloid cells. Like other TIM family members, Tim-3 can bind to phosphatidylserine displayed by apoptotic cells, and this interaction has been shown to mediate uptake of such cells by dendritic cells and cross-presentation of antigens to CD8+ T cells. In contrast, how the recognition of PS by Tim-3 might regulate the function of Tim-3+ T cells is not known. In their recent paper, Lemmon and colleagues demonstrate for the first time that recognition of PS by Tim-3 leads to enhanced T cell activation.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Phosphatidylserines , Hepatitis A Virus Cellular Receptor 2/genetics , Lymphocyte Activation , Membrane Proteins/genetics , Phagocytosis
5.
J Virol ; 94(2)2020 01 06.
Article in English | MEDLINE | ID: mdl-31645447

ABSTRACT

Herpes simplex virus 1 (HSV-1) causes a lifelong infection of neurons that innervate barrier sites like the skin and mucosal surfaces like the eye. After primary infection of the cornea, the virus enters latency within the trigeminal ganglion (TG), from which it can reactivate throughout the life of the host. Viral latency is maintained, in part, by virus-specific CD8+ T cells that nonlethally interact with infected neurons. When CD8+ T cell responses are inhibited, HSV-1 can reactivate, and these recurrent reactivation events can lead to blinding scarring of the cornea. In the C57BL/6 mouse, CD8+ T cells specific for the immunodominant epitope from glycoprotein B maintain functionality throughout latency, while CD8+ T cells specific for subdominant epitopes undergo functional impairment that is associated with the expression of the inhibitory checkpoint molecule programmed death 1 (PD-1). Here, we investigate the checkpoint molecule T cell immunoglobulin and mucin domain-containing 3 (Tim-3), which has traditionally been associated with CD8+ T cell exhaustion. Unexpectedly, we found that Tim-3 was preferentially expressed on highly functional ganglionic CD8+ T cells during acute and latent HSV-1 infection. This, paired with data that show that Tim-3 expression on CD8+ T cells in the latently infected TG is influenced by viral gene expression, suggests that Tim-3 is an indicator of recent T cell stimulation, rather than functional compromise, in this model. We conclude that Tim-3 expression is not sufficient to define functional compromise during latency; however, it may be useful in identifying activated cells within the TG during HSV-1 infection.IMPORTANCE Without an effective means of eliminating HSV-1 from latently infected neurons, efforts to control the virus have centered on preventing viral reactivation from latency. Virus-specific CD8+ T cells within the infected TG have been shown to play a crucial role in inhibiting viral reactivation, and with a portion of these cells exhibiting functional impairment, checkpoint molecule immunotherapies have presented a potential solution to enhancing the antiviral response of these cells. In pursuing this potential treatment strategy, we found that Tim-3 (often associated with CD8+ T cell functional exhaustion) is not upregulated on impaired cells but instead is upregulated on highly functional cells that have recently received antigenic stimulation. These findings support a role for Tim-3 as a marker of activation rather than exhaustion in this model, and we provide additional evidence for the hypothesis that there is persistent viral gene expression in the HSV-1 latently infected TG.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Gene Expression Regulation/immunology , Hepatitis A Virus Cellular Receptor 2/immunology , Herpesvirus 1, Human/physiology , Lymphocyte Activation , Programmed Cell Death 1 Receptor/immunology , Trigeminal Ganglion , Virus Latency/immunology , Animals , Biomarkers , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Female , Mice , Trigeminal Ganglion/immunology , Trigeminal Ganglion/pathology , Trigeminal Ganglion/virology
6.
J Immunol ; 203(3): 665-675, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31209103

ABSTRACT

ß-site amyloid precursor protein-cleaving enzyme 1 (BACE1) is best known for its role in Alzheimer's disease amyloid plaque formation but also contributes to neurodegenerative processes triggered by CNS injury. In this article, we report that BACE1 is expressed in murine CD4+ T cells and regulates signaling through the TCR. BACE1-deficient T cells have reduced IL-17A expression under Th17 conditions and reduced CD73 expression in Th17 and inducible T regulatory cells. However, induction of the Th17 and T regulatory transcription factors RORγt and Foxp3 was unaffected. BACE1-deficient T cells showed impaired pathogenic function in experimental autoimmune encephalomyelitis. These data identify BACE1 as a novel regulator of T cell signaling pathways that impact autoimmune inflammatory T cell function.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Autoimmunity/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , 5'-Nucleotidase/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Forkhead Transcription Factors/biosynthesis , Interleukin-17/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis
7.
Proc Natl Acad Sci U S A ; 115(10): 2455-2460, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29463725

ABSTRACT

Tim-3 is highly expressed on a subset of T cells during T cell exhaustion in settings of chronic viral infection and tumors. Using lymphocytic choriomeningitis virus (LCMV) Clone 13, a model for chronic infection, we found that Tim-3 was neither necessary nor sufficient for the development of T cell exhaustion. Nonetheless, expression of Tim-3 was sufficient to drive resistance to PD-L1 blockade therapy during chronic infection. Strikingly, expression of Tim-3 promoted the development of short-lived effector T cells, at the expense of memory precursor development, after acute LCMV infection. These effects were accompanied by increased Akt/mTOR signaling in T cells expressing endogenous or ectopic Tim-3. Conversely, Akt/mTOR signaling was reduced in effector T cells from Tim-3-deficient mice. Thus, Tim-3 is essential for optimal effector T cell responses, and may also contribute to exhaustion by restricting the development of long-lived memory T cells. Taken together, our results suggest that Tim-3 is actually more similar to costimulatory receptors that are up-regulated after T cell activation than to a dominant inhibitory protein like PD-1. These findings have significant implications for the development of anti-Tim-3 antibodies as therapeutic agents.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/immunology , Lymphocyte Activation/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Animals , Chronic Disease , Hepatitis A Virus Cellular Receptor 2/genetics , Lymphocytic Choriomeningitis , Lymphocytic choriomeningitis virus/immunology , Mice , Phenotype , Receptors, Antigen, T-Cell/immunology , TOR Serine-Threonine Kinases/metabolism
8.
J Immunol ; 198(2): 640-644, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27974455

ABSTRACT

There is growing appreciation that cellular metabolic and bioenergetic pathways do not play merely passive roles in activated leukocytes. Rather, metabolism has important roles in controlling cellular activation, differentiation, survival, and effector function. Much of this work has been performed in T cells; however, there is still very little information regarding mast cell metabolic reprogramming and its effect on cellular function. Mast cells perform important barrier functions and help control type 2 immune responses. In this study we show that murine bone marrow-derived mast cells rapidly alter their metabolism in response to stimulation through the FcεRI. We also demonstrate that specific metabolic pathways appear to be differentially required for the control of mast cell function. Manipulation of metabolic pathways may represent a novel point for the manipulation of mast cell activation.


Subject(s)
Mast Cells/immunology , Mast Cells/metabolism , Animals , Cell Differentiation/immunology , Cells, Cultured , Mice , Mice, Inbred C57BL
9.
J Immunol ; 194(10): 4615-9, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25855357

ABSTRACT

Signaling via the Akt/mammalian target of rapamycin pathway influences CD4(+) T cell differentiation; low levels favor regulatory T cell induction and high levels favor Th induction. Although the lipid phosphatase phosphatase and tensin homolog (PTEN) suppresses Akt activity, the control of PTEN activity is poorly studied in T cells. In this study, we identify multiple mechanisms that regulate PTEN expression. During Th induction, PTEN function is suppressed via lower mRNA levels, lower protein levels, and an increase in C-terminal phosphorylation. Conversely, during regulatory T cell induction, PTEN function is maintained through the stabilization of PTEN mRNA transcription and sustained protein levels. We demonstrate that differential Akt/mammalian target of rapamycin signaling regulates PTEN transcription via the FoxO1 transcription factor. A mathematical model that includes multiple modes of PTEN regulation recapitulates our experimental findings and demonstrates how several feedback loops determine differentiation outcomes. Collectively, this work provides novel mechanistic insights into how differential regulation of PTEN controls alternate CD4(+) T cell fate outcomes.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/immunology , Lymphocyte Activation/immunology , Oncogene Protein v-akt/immunology , PTEN Phosphohydrolase/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Lineage , Chromatin Immunoprecipitation , Flow Cytometry , Forkhead Box Protein O1 , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Models, Theoretical , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology
10.
Int J Mol Sci ; 18(3)2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28300768

ABSTRACT

Cancer immunotherapy has produced impressive clinical results in recent years. Despite the success of the checkpoint blockade strategies targeting cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed death receptor 1 (PD-1), a large portion of cancer patients have not yet benefited from this novel therapy. T cell immunoglobulin and mucin domain 3 (TIM-3) has been shown to mediate immune tolerance in mouse models of infectious diseases, alloimmunity, autoimmunity, and tumor Immunity. Thus, targeting TIM-3 emerges as a promising approach for further improvement of current immunotherapy. Despite a large amount of experimental data showing an immune suppressive function of TIM-3 in vivo, the exact mechanisms are not well understood. To enable effective targeting of TIM-3 for tumor immunotherapy, further in-depth mechanistic studies are warranted. These studies will also provide much-needed insight for the rational design of novel combination therapy with other checkpoint blockers. In this review, we summarize key evidence supporting an immune regulatory role of TIM-3 and discuss possible mechanisms of action.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/immunology , Immunotherapy/methods , Neoplasms/therapy , Animals , Humans , Immune Tolerance , Ligands , Neoplasms/immunology , T-Lymphocyte Subsets/immunology
11.
J Immunol ; 193(4): 1525-30, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25086175

ABSTRACT

T cell exhaustion is thought to be a natural mechanism for limiting immune pathology, although it may be desirable to circumvent this mechanism to help eliminate viral reservoirs or tumors. Although there are no definitive markers, a fingerprint for exhausted T cells has been described that includes the transmembrane proteins PD-1, LAG3, and Tim-3. However, apart from the recruitment of tyrosine phosphatases to PD-1, little is known about the biochemical mechanisms by which these proteins contribute to the development or maintenance of exhaustion. Tim-3 contains no known motifs for the recruitment of inhibitory phosphatases, but it may actually increase signaling downstream of TCR/CD3, at least under acute conditions. Other studies showed that T cell exhaustion results from chronic stimulation that extends the effector phase of T cell activation, at the expense of T cell memory. We suggest that Tim-3 may contribute to T cell exhaustion by enhancing TCR-signaling pathways.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Membrane Proteins/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Animals , Antigens, CD/biosynthesis , Antigens, CD/immunology , Cell Proliferation , Cytokines/biosynthesis , HIV-1/immunology , Hepacivirus/immunology , Hepatitis A Virus Cellular Receptor 2 , Humans , Lymphocyte Activation/immunology , Membrane Proteins/biosynthesis , Mice , Positive Regulatory Domain I-Binding Factor 1 , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/genetics , Repressor Proteins/biosynthesis , Tumor Microenvironment/immunology , Lymphocyte Activation Gene 3 Protein
12.
J Immunol ; 193(7): 3288-95, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25149466

ABSTRACT

Systemic lupus erythematosus (SLE) is a type I IFN (IFN-I)-driven autoimmune disorder with exaggerated B and Th cell responses. Th17 cells, a recently identified Th cell subset, have been strongly implicated in the pathogenesis of SLE. Because IFN-I suppresses the generation and expansion of Th17 cells in an IL-27-dependent manner, it is unclear how pathogenic Th17 cells are generated in SLE in the presence of an environment characterized by high IFN-I levels. In this study, we showed that activation of c5aR on murine macrophages blocked IFN-I-mediated IL-27 production, thus permitting the development of Th17 cells. C5aR activation on IFN-I-responsive macrophages inhibits IRF-1-mediated transactivation of IL-27 gene expression via the PI3K/Akt pathway. Consistently, C5aR-deficient mice exhibited increased IL-27 expression and fewer Th17 cells and consequently developed reduced lupus nephritis in comparison with wild-type mice. In support of these findings in mice, we found that C5a inhibited IFN-I-induced IL-27 production from macrophages of lupus subjects. Moreover, the level of serum C5a correlated with Th17 frequency in peripheral blood. Collectively, these data indicate an essential role for C5a in the generation of pathogenic Th17 responses in SLE. Thus, therapeutic strategies to block C5aR activation may be beneficial for controlling pathogenic Th17-mediated inflammation in SLE.


Subject(s)
Complement C5a/immunology , Interferon Type I/immunology , Lupus Nephritis/immunology , Macrophages/immunology , Th17 Cells/immunology , Transcriptional Activation/immunology , Animals , Complement C5a/genetics , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/immunology , Interferon Type I/genetics , Interleukins/genetics , Interleukins/immunology , Lupus Nephritis/genetics , Lupus Nephritis/pathology , Lupus Nephritis/therapy , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/immunology , Th17 Cells/pathology , Transcriptional Activation/genetics
13.
bioRxiv ; 2024 May 02.
Article in English | MEDLINE | ID: mdl-38746242

ABSTRACT

The protein known as PI3K-interacting protein (PIK3IP1), or transmembrane inhibitor of PI3K (TrIP), is highly expressed by T cells and can modulate PI3K activity in these cells. Several studies have also revealed that TrIP is rapidly downregulated following T cell activation. However, it is unclear as to how this downregulation is controlled. Using a novel monoclonal antibody that robustly stains cell-surface TrIP, we demonstrate that TrIP is lost from the surface of activated T cells in a manner dependent on the strength of signaling through the T cell receptor (TCR) and specific downstream signaling pathways. In addition, TrIP expression returns after 24 hours, suggesting that it may play a role in resetting TCR signaling at later time points. Finally, by expressing truncated forms of TrIP in cells, we identify the region in the extracellular stalk domain of TrIP that is targeted for proteolytic cleavage by metalloprotease ADAM17.

14.
Eur J Immunol ; 42(10): 2754-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22706993

ABSTRACT

The PI-3 kinase (PI3K) pathway is critical for T-cell development and activation. Several negative regulators of this pathway have already been described and characterized: the lipid phosphatases SHIP, inositol polyphosphate-4-phosphatase, type II (INPP4B), and phosphatase and tensin homolog (PTEN), the latter of which are tumor suppressors. PIK3IP1 (PI3K interacting protein 1) is a recently described transmembrane protein that has the ability to bind the catalytic protein p110 and prevent its activation by the p85 family adaptor proteins. Thus far, nothing is known about the possible role of PIK3IP1 in the regulation of lymphocyte development or activation. Here, we show for the first time that PIK3IP1 is expressed in T cells. Ectopic expression of PIK3IP1 in Jurkat or D10 T-cell lines inhibited activation of an NFAT/AP-1 transcriptional reporter. Conversely, siRNA-mediated silencing of PIK3IP1 in the same cell lines modestly augmented Akt phosphorylation, T-cell activation, and production of IL-2. These results suggest that the novel PI3K regulator PIK3IP1 plays an inhibitory role in T-cell activation.


Subject(s)
Membrane Proteins/metabolism , T-Lymphocytes/immunology , Humans , Immunosuppression Therapy , Interleukin-2/metabolism , Intracellular Signaling Peptides and Proteins , Jurkat Cells , Lymphocyte Activation/genetics , Membrane Proteins/genetics , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Oncogene Protein v-akt/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcriptional Activation/genetics , Transgenes/genetics
15.
Blood Adv ; 7(22): 6949-6963, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37738167

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) and macrophage activation syndrome (MAS) are life-threatening hyperinflammatory syndromes. Familial HLH is caused by genetic impairment of granule-mediated cytotoxicity (eg, perforin deficiency). MAS is linked to excess activity of the inflammasome-activated cytokine interleukin-18 (IL-18). Though individually tolerated, mice with dual susceptibility (Prf1⁻/⁻Il18tg; DS) succumb to spontaneous, lethal hyperinflammation. We hypothesized that understanding how these susceptibility factors synergize would uncover key pathomechanisms in the activation, function, and persistence of hyperactivated CD8 T cells. In IL-18 transgenic (Il18tg) mice, IL-18 effects on CD8 T cells drove MAS after a viral (lymphocytic choriomeningitis virus), but not innate (toll like receptor 9), trigger. In vitro, CD8 T cells also required T-cell receptor (TCR) stimulation to fully respond to IL-18. IL-18 induced but perforin deficiency impaired immunoregulatory restimulation-induced cell death (RICD). Paralleling hyperinflammation, DS mice displayed massive postthymic oligoclonal CD8 T-cell hyperactivation in their spleens, livers, and bone marrow as early as 3 weeks. These cells increased proliferation and interferon gamma production, which contrasted with increased expression of receptors and transcription factors associated with exhaustion. Broad-spectrum antibiotics and antiretrovirals failed to ameliorate the disease. Attempting to genetically "fix" TCR antigen-specificity instead demonstrated the persistence of spontaneous HLH and hyperactivation, chiefly on T cells that had evaded TCR fixation. Thus, drivers of HLH may preferentially act on CD8 T cells: IL-18 amplifies activation and demand for RICD, whereas perforin supplies critical immunoregulation. Together, these factors promote a terminal CD8 T-cell activation state, combining features of exhaustion and effector function. Therefore, susceptibility to hyperinflammation may converge on a unique, unrelenting, and antigen-dependent state of CD8 T-cell hyperactivation.


Subject(s)
Lymphohistiocytosis, Hemophagocytic , Mice , Animals , Lymphohistiocytosis, Hemophagocytic/etiology , Perforin/genetics , Perforin/metabolism , Interleukin-18/metabolism , CD8-Positive T-Lymphocytes/metabolism , Receptors, Antigen, T-Cell/metabolism
16.
J Leukoc Biol ; 114(4): 368-372, 2023 09 27.
Article in English | MEDLINE | ID: mdl-37350502

ABSTRACT

TIM-3 expression is increased on peripheral regulatory T cells (Tregs) of virally suppressed persons with HIV-1 on antiretroviral therapy (PWH-ART). However, the relevance of TIM-3 expression in this setting is unclear. We used flow cytometry to evaluate the suppressive phenotype and signaling pathways in peripheral TIM-3- vs TIM-3+ Tregs in PWH-ART. TIM-3+ Tregs showed increased expression of IL-10 compared with persons without HIV-1. In addition, TIM-3+ Tregs displayed elevated signaling and activation, relative to TIM-3- Tregs from the same PWH-ART. Dramatically, TIM-3 blockade restrained the in vitro suppressive capacity of peripheral Tregs. Therefore, our data demonstrate not only that TIM-3 expression by Tregs is associated with an immunosuppressive response among PWH-ART, but also that TIM-3 contributes directly to the enhanced suppressive activity of Tregs in this setting.


Subject(s)
HIV Infections , Hepatitis A Virus Cellular Receptor 2 , Humans , Hepatitis A Virus Cellular Receptor 2/metabolism , T-Lymphocytes, Regulatory/metabolism , HIV Infections/drug therapy , HIV Infections/metabolism
17.
Sci Immunol ; 8(82): eadd8454, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37083450

ABSTRACT

Our understanding of tissue-resident memory T (TRM) cell biology has been largely developed from acute infection models in which antigen is cleared and sterilizing immunity is achieved. Less is known about TRM cells in the context of chronic antigen persistence and inflammation. We investigated factors that underlie TRM maintenance in a kidney transplantation model in which TRM cells drive rejection. In contrast to acute infection, we found that TRM cells declined markedly in the absence of cognate antigen, antigen presentation, or antigen sensing by the T cells. Depletion of graft-infiltrating dendritic cells or interruption of antigen presentation after TRM cells were established was sufficient to disrupt TRM maintenance and reduce allograft pathology. Likewise, removal of IL-15 transpresentation or of the IL-15 receptor on T cells during TRM maintenance led to a decline in TRM cells, and IL-15 receptor blockade prevented chronic rejection. Therefore, antigen and IL-15 presented by dendritic cells play nonredundant key roles in CD8 TRM cell maintenance in settings of antigen persistence and inflammation. These findings provide insights that could lead to improved treatment of chronic transplant rejection and autoimmunity.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-15 , Humans , Antigens , Inflammation , Memory T Cells
18.
J Biol Chem ; 286(41): 36076-36085, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21862580

ABSTRACT

Activation of the NF-κB signaling pathway is critical for leukocyte activation and development. Although previous studies suggested a role for the Akt kinase in coupling the T cell antigen receptor and CD28 to NF-κB activation in T cells, the nature of the role of Akt in this pathway is still unclear. Using a targeted gene profiling approach, we found that a subset of NF-κB-dependent genes required Akt for optimal up-regulation during T cell activation. The selective effects of Akt were manifest at the level of mRNA transcription and p65/RelA binding to upstream promoters and appear to be due to altered formation of the Carma1-Bcl10 complex. The proinflammatory cytokine TNF-α was found to be particularly sensitive to Akt inhibition or knockdown, including in primary human blood T cells and a murine model of rheumatoid arthritis. Our findings are consistent with a hierarchy in the expression of NF-κB-dependent genes, controlled by the strength and/or duration of NF-κB signaling. More broadly, our results suggest that defining the more graded effects of signaling, such as those demonstrated here for Akt and the NF-κB pathway, is important to understanding how cells can fine-tune signaling responses for optimal sensitivity and specificity.


Subject(s)
Gene Expression Regulation/physiology , Lymphocyte Activation/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , T-Lymphocytes/metabolism , Transcription Factor RelA/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , B-Cell CLL-Lymphoma 10 Protein , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Humans , Jurkat Cells , Promoter Regions, Genetic/physiology , Proto-Oncogene Proteins c-akt/genetics , T-Lymphocytes/cytology , Transcription Factor RelA/genetics
19.
J Immunol ; 184(6): 2743-9, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20200285

ABSTRACT

Proteins of the transmembrane (or T cell) Ig and mucin domain (TIM) family are expressed by multiple cell types within the immune systems of rodents and humans. Studies over the last several years have suggested that these proteins may be promising targets for therapeutic manipulation of immune responses. This review discusses the progress that has been made in understanding TIM protein function in the immune system, as well as some of the unresolved issues that remain on the road to eventually targeting TIM proteins for enhancing or inhibiting immunity.


Subject(s)
Immunity, Cellular , Immunoglobulins/physiology , Mucins/physiology , T-Lymphocytes/immunology , Animals , Cell Cycle Proteins/physiology , Humans , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/classification , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Protein Structure, Tertiary , T-Lymphocytes/metabolism
20.
J Immunol ; 185(2): 1063-70, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20554964

ABSTRACT

IL-17 mediates essential inflammatory responses in host defense and autoimmunity. The IL-17A-IL-17F signaling complex is composed of IL-17RA and IL-17RC, both of which are necessary for signal transduction. To date, the specific contribution of IL-17RC to downstream signaling remains poorly understood. To define the regions within the IL-17RC cytoplasmic tail required for signal transduction, we assayed signaling by a panel of IL-17RC deletion mutants. These findings reveal that IL-17RC inducibly associates with a specific glycosylated IL-17RA isoform, in a manner independent of the IL-17RC cytoplasmic tail. Using expression of the IL-17 target genes IL-6 and 24p3/lipocalin-2 as a readout, functional reconstitution of signaling in IL-17RC(-/-) fibroblasts required the SEF/IL-17R signaling domain (SEFIR), a conserved motif common to IL-17R family members. Unexpectedly, the IL-17RC SEFIR alone was not sufficient to reconstitute IL-17-dependent signaling. Rather, an additional sequence downstream of the SEFIR was also necessary. We further found that IL-17RC interacts directly with the adaptor/E3 ubiquitin ligase Act1, and that the functional IL-17RC isoforms containing the extended SEFIR region interact specifically with a phosphorylated isoform of Act1. Finally, we show that IL-17RC is required for in vivo IL-17-dependent responses during oral mucosal infections caused by the human commensal fungus Candida albicans. These results indicate that IL-17RC is vital for IL-17-dependent signaling both in vitro and in vivo. Insight into the mechanisms by which IL-17RC signals helps shed light on IL-17-dependent inflammatory responses and may ultimately provide an avenue for therapeutic intervention in IL-17-mediated diseases.


Subject(s)
Amino Acid Motifs , Fibroblasts/immunology , Receptors, Interleukin-17/immunology , Signal Transduction/immunology , Amino Acid Sequence , Animals , Binding Sites , Blotting, Western , Candidiasis, Oral/genetics , Candidiasis, Oral/immunology , Cell Line , Cells, Cultured , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Genetic Predisposition to Disease , Humans , Interleukin-17/pharmacology , Mice , Mice, Knockout , Mutation , Oropharynx/immunology , Oropharynx/microbiology , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/metabolism , Signal Transduction/drug effects , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL