Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
J Leukoc Biol ; 116(2): 210-223, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38489541

ABSTRACT

Toll-like receptors are pattern recognition receptors that bridge the innate and adaptive immune responses and are critical for host defense. Most studies of Toll-like receptors have focused upon their roles in myeloid cells. B lymphocytes express most Toll-like receptors and are responsive to Toll-like receptor ligands, yet Toll-like receptor-mediated signaling in B cells is relatively understudied. This is an important knowledge gap, as Toll-like receptor functions can be cell type specific. In striking contrast to myeloid cells, TRAF3 inhibits TLR-mediated functions in B cells. TRAF3-deficient B cells display enhanced IRF3 and NFκB activation, cytokine production, immunoglobulin isotype switching, and antibody production in response to Toll-like receptors 3, 4, 7, and 9. Here, we address the question of how TRAF3 impacts initial B-cell Toll-like receptor signals to regulate downstream activation. We found that TRAF3 in B cells associated with proximal Toll-like receptor 4 and 7 signaling proteins, including MyD88, TRAF6, and the tyrosine kinase Syk. In the absence of TRAF3, TRAF6 showed a greater association with several Toll-like receptor signaling proteins, suggesting that TRAF3 may inhibit TRAF6 access to Toll-like receptor signaling complexes and thus early Toll-like receptor signaling. In addition, our results highlight a key role for Syk in Toll-like receptor signaling in B cells. In the absence of TRAF3, Syk activation was enhanced in response to ligands for Toll-like receptors 4 and 7, and Syk inhibition reduced downstream Toll-like receptor-mediated NFκB activation and proinflammatory cytokine production. This study reveals multiple mechanisms by which TRAF3 serves as a key negative regulator of early Toll-like receptor signaling events in B cells.


Subject(s)
B-Lymphocytes , Signal Transduction , TNF Receptor-Associated Factor 3 , TNF Receptor-Associated Factor 6 , Toll-Like Receptors , TNF Receptor-Associated Factor 3/metabolism , TNF Receptor-Associated Factor 3/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Animals , Toll-Like Receptors/metabolism , Mice , TNF Receptor-Associated Factor 6/metabolism , Syk Kinase/metabolism , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Mice, Knockout , Mice, Inbred C57BL , NF-kappa B/metabolism
2.
J Immunol ; 212(1): 1-2, 2024 01 01.
Article in English | MEDLINE | ID: mdl-38118105
3.
PLoS Pathog ; 19(10): e1011720, 2023 10.
Article in English | MEDLINE | ID: mdl-37824591

ABSTRACT

Sepsis, an amplified immune response to systemic infection, is characterized by a transient cytokine storm followed by chronic immune dysfunction. Consequently, sepsis survivors are highly susceptible to newly introduced infections, suggesting sepsis can influence the function and composition of the naïve CD8 T cell pool and resulting pathogen-induced primary CD8 T cell responses. Here, we explored the extent to which sepsis induces phenotypic and functional changes within the naïve CD8 T cell pool. To interrogate this, the cecal ligation and puncture (CLP) mouse model of polymicrobial sepsis was used. In normal, non-septic mice, we show type-I interferon (IFN I)-mediated signaling plays an important role in driving the phenotypic and functional heterogeneity in the naïve CD8 T cell compartment leading to increased representation of Ly6C+ naïve CD8 T cells. In response to viral infection after sepsis resolution, naïve Ly6C+ CD8 T cells generated more primary effector and memory CD8 T cells with slower conversion to a central memory CD8 T cell phenotype (Tcm) than Ly6C- naïve CD8 T cells. Importantly, as a potent inducer of cytokine storm and IFN I production, sepsis leads to increased representation of Ly6C+ naïve CD8 T cells that maintained their heightened ability to respond (i.e., effector and memory CD8 T cell accumulation and cytokine production) to primary LCMV infection. Lastly, longitudinal analyses of peripheral blood samples obtained from septic patients revealed profound changes in CD8 T cell subset composition and frequency compared to healthy controls. Thus, sepsis has the capacity to alter the composition of naïve CD8 T cells, directly influencing primary CD8 T cell responses to newly introduced infections.


Subject(s)
Cytokine Release Syndrome , Sepsis , Humans , Mice , Animals , CD8-Positive T-Lymphocytes , Immunity, Innate , Phenotype , Mice, Inbred C57BL , Immunologic Memory
4.
Viruses ; 15(6)2023 06 12.
Article in English | MEDLINE | ID: mdl-37376652

ABSTRACT

Macrophages are critical in the pathogenesis of a diverse group of viral pathogens, both as targets of infection and for eliciting primary defense mechanisms. Our prior in vitro work identified that CD40 signaling in murine peritoneal macrophages protects against several RNA viruses by eliciting IL-12, which stimulates the production of interferon gamma (IFN-γ). Here, we examine the role of CD40 signaling in vivo. We show that CD40 signaling is a critical, but currently poorly appreciated, component of the innate immune response using two distinct infectious agents: mouse-adapted influenza A virus (IAV, PR8) and recombinant VSV encoding the Ebola virus glycoprotein (rVSV-EBOV GP). We find that stimulation of CD40 signaling decreases early IAV titers, whereas loss of CD40 elevated early titers and compromised lung function by day 3 of infection. Protection conferred by CD40 signaling against IAV is dependent on IFN-γ production, consistent with our in vitro studies. Using rVSV-EBOV GP that serves as a low-biocontainment model of filovirus infection, we demonstrate that macrophages are a CD40-expressing population critical for protection within the peritoneum and T-cells are the key source of CD40L (CD154). These experiments reveal the in vivo mechanisms by which CD40 signaling in macrophages regulates the early host responses to RNA virus infection and highlight how CD40 agonists currently under investigation for clinical use may function as a novel class of broad antiviral treatments.


Subject(s)
CD40 Antigens , RNA Virus Infections , RNA Viruses , Animals , Mice , CD40 Antigens/metabolism , Interferon-gamma , Macrophages , RNA Virus Infections/immunology
5.
Front Immunol ; 14: 1129251, 2023.
Article in English | MEDLINE | ID: mdl-36814922

ABSTRACT

Tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) is an adapter protein with many context-specific functions. Early studies of lymphocyte TRAF3 hinted at TRAF3's importance for T cell function, but elucidation of specific mechanisms was delayed by early lethality of globally TRAF3-/- mice. Development of a conditional TRAF3-deficient mouse enabled important descriptive and mechanistic insights into how TRAF3 promotes optimal T cell function. Signaling through the T cell antigen receptor (TCR) fails to induce normal proliferation and survival in TRAF3 -/- T cells, and TCR-activated cells in vitro and in vivo have deficient cytokine production. These defects can be traced to incorrect localization and function of negative regulatory phosphatases acting at different parts of the signaling cascade, as can dysregulated effector responses and memory T cell homeostasis in vivo and an enlarged regulatory T cell (Treg) compartment. The important regulatory activity of TRAF3 is also evident at members of the TNFR superfamily and cytokine receptors. Here, we review significant advances in mechanistic understanding of how TRAF3 regulates T cell differentiation and function, through modulation of signaling through the TCR, costimulatory receptors, and cytokine receptors. Finally, we briefly discuss the recent identification of families carrying single allele loss-of-function mutations in TRAF3, and compare the findings in their T cells with the T cell defects identified in mice whose T cells completely lack T cell TRAF3. Together, the body of work describing TRAF3-mediated regulation of T cell effector function and differentiation frame TRAF3 as an important modulator of T cell signal integration.


Subject(s)
Signal Transduction , T-Lymphocytes, Regulatory , TNF Receptor-Associated Factor 3 , Animals , Mice , Receptors, Antigen, T-Cell/metabolism , Receptors, Cytokine/metabolism , TNF Receptor-Associated Factor 3/metabolism
6.
Cancers (Basel) ; 14(20)2022 Oct 14.
Article in English | MEDLINE | ID: mdl-36291813

ABSTRACT

TNF receptor-associated factor 3 (TRAF3) is an adapter protein that inhibits many signals that promote B cell survival and activation. Mice with a B cell-specific TRAF3 deficiency and humans with a rare haploinsufficiency in TRAF3 have enhanced development of BCLs as they age. Loss-of-function mutations in TRAF3 are common in B cell malignancies. Recent studies show that pharmacological inhibition of the enzyme glycogen synthase kinase 3 (GSK3), which regulates cellular growth, survival, and metabolism, inhibits growth and survival of BCL-derived B cells. In this study, we found that TRAF3 and GSK3 associated in B cells. The relative levels of TRAF3 in BCL cell lines correlated positively with the ratio of inactive to total GSK3ß, and negatively correlated with susceptibility to GSK3 inhibition by the GSK3 inhibitory drug 9-ING-41, currently in clinical trials. Uniquely in BCLs with low TRAF3, GSK3 inhibition caused increased loss of the TRAF3-regulated, anti-apoptotic protein Mcl-1. GSK3 inhibition also blocked hyperresponsiveness to IL-6 receptor signaling in TRAF3-deficient BCL cells. Together, these results support the utility of 9-ING-41 as a treatment for BCL, and suggest that a decrease or loss of TRAF3 in BCLs could act as a biomarker for increased susceptibility to GSK3 inhibitor treatment.

7.
Sci Signal ; 15(753): eabn5507, 2022 Sep 27.
Article in English | MEDLINE | ID: mdl-36166512

ABSTRACT

Type I interferons (IFNs) are among the most powerful tools that host cells deploy against intracellular pathogens. Their effectiveness is due both to the rapid, directly antiviral effects of IFN-stimulated gene products and to the effects of type I IFN on responding immune cells. Type I IFN signaling through its receptor, IFNAR, is tightly regulated at multiple steps in the signaling cascade, including at the level of IFNAR downstream effectors, which include the kinase JAK1 and the transcriptional regulator STAT1. Here, we found that tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) enhanced the activation of JAK1 and STAT1 specifically in CD4+ T cells by preventing recruitment of the negative regulatory phosphatase PTPN22 to the IFNAR complex. The balance between signals through IFNAR and other cytokine receptors influences CD4+ T cell differentiation and function during infections. Our work reveals TRAF3 and PTPN22 as key regulators of CD4+ T cell activation by type I IFNs.


Subject(s)
Interferon Type I , TNF Receptor-Associated Factor 3 , Antiviral Agents/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Phosphoric Monoester Hydrolases/metabolism , Receptor, Interferon alpha-beta/genetics , Receptors, Tumor Necrosis Factor/metabolism , T-Lymphocytes/metabolism , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism
8.
Sci Immunol ; 7(74): eabn3800, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35960817

ABSTRACT

Tumor necrosis factor receptor-associated factor 3 (TRAF3) is a central regulator of immunity. TRAF3 is often somatically mutated in B cell malignancies, but its role in human immunity is not defined. Here, in five unrelated families, we describe an immune dysregulation syndrome of recurrent bacterial infections, autoimmunity, systemic inflammation, B cell lymphoproliferation, and hypergammaglobulinemia. Affected individuals each had monoallelic mutations in TRAF3 that reduced TRAF3 expression. Immunophenotyping showed that patients' B cells were dysregulated, exhibiting increased nuclear factor-κB 2 activation, elevated mitochondrial respiration, and heightened inflammatory responses. Patients had mild CD4+ T cell lymphopenia, with a reduced proportion of naïve T cells but increased regulatory T cells and circulating T follicular helper cells. Guided by this clinical phenotype, targeted analyses demonstrated that common genetic variants, which also reduce TRAF3 expression, are associated with an increased risk of B cell malignancies, systemic lupus erythematosus, higher immunoglobulin levels, and bacterial infections in the wider population. Reduced TRAF3 conveys disease risks by driving B cell hyperactivity via intrinsic activation of multiple intracellular proinflammatory pathways and increased mitochondrial respiration, with a likely contribution from dysregulated T cell help. Thus, we define monogenic TRAF3 haploinsufficiency syndrome and demonstrate how common TRAF3 variants affect a range of human diseases.


Subject(s)
Neoplasms , TNF Receptor-Associated Factor 3 , Autoimmunity/genetics , B-Lymphocytes , Humans , Mutation , Neoplasms/pathology , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism
9.
J Biol Chem ; 297(3): 101097, 2021 09.
Article in English | MEDLINE | ID: mdl-34418432

ABSTRACT

Tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) plays context-specific roles in multiple receptor-mediated signaling pathways in different cell types. Mice lacking TRAF3 in T cells display defective T-cell-mediated immune responses to immunization and infection and demonstrate defective early signaling via the TCR complex. However, the role of TRAF3 in the function of GITR/TNFRSF18, an important costimulatory member of the TNFR superfamily, is unclear. Here we investigated the impact of T cell TRAF3 status on both GITR expression and activation of specific kinases in the GITR signaling pathway in T cells. Our results indicate that TRAF3 negatively regulates GITR functions in several ways. First, expression of GITR protein was elevated in TRAF3-deficient T cells, resulting from both transcriptional and posttranslational regulation that led to greater GITR transcript levels, as well as enhanced GITR protein stability. TRAF3 associated with T cell GITR in a manner dependent upon GITR ligation. TRAF3 also inhibited several events of the GITR mediated early signaling cascade, in a manner independent of recruitment of phosphatases, a mechanism by which TRAF3 inhibits signaling through several other cytokine receptors. These results add new information to our understanding of GITR signaling and function in T cells, which is relevant to the potential use of GITR to enhance immune therapies.


Subject(s)
Costimulatory and Inhibitory T-Cell Receptors/metabolism , Glucocorticoid-Induced TNFR-Related Protein/metabolism , TNF Receptor-Associated Factor 3/metabolism , Animals , Female , Glucocorticoid-Induced TNFR-Related Protein/physiology , Interleukin-2/metabolism , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/immunology , T-Lymphocytes/metabolism , TNF Receptor-Associated Factor 3/physiology
10.
Cells ; 10(8)2021 08 12.
Article in English | MEDLINE | ID: mdl-34440839

ABSTRACT

Uncontrolled inflammation is associated with neurodegenerative conditions in central nervous system tissues, including the retina and brain. We previously found that the neural retina (NR) plays an important role in retinal immunity. Tumor necrosis factor Receptor-Associated Factor 3 (TRAF3) is a known immune regulator expressed in the retina; however, whether TRAF3 regulates retinal immunity is unknown. We have generated the first conditional NR-Traf3 knockout mouse model (Chx10-Cre/Traf3f/f) to enable studies of neuronal TRAF3 function. Here, we evaluated NR-Traf3 depletion effects on whole retinal TRAF3 protein expression, visual acuity, and retinal structure and function. Additionally, to determine if NR-Traf3 plays a role in retinal immune regulation, we used flow cytometry to assess immune cell infiltration following acute local lipopolysaccharide (LPS) administration. Our results show that TRAF3 protein is highly expressed in the NR and establish that NR-Traf3 depletion does not affect basal retinal structure or function. Importantly, NR-Traf3 promoted LPS-stimulated retinal immune infiltration. Thus, our findings propose NR-Traf3 as a positive regulator of retinal immunity. Further, the NR-Traf3 mouse provides a tool for investigations of neuronal TRAF3 as a novel potential target for therapeutic interventions aimed at suppressing retinal inflammatory disease and may also inform treatment approaches for inflammatory neurodegenerative brain conditions.


Subject(s)
Homeodomain Proteins/genetics , Neurons/metabolism , Retina/metabolism , TNF Receptor-Associated Factor 3/genetics , Transcription Factors/genetics , Animals , Disease Models, Animal , Electroretinography , Immunity/drug effects , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Neurons/immunology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Retina/physiology , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/metabolism , Transcription Factors/deficiency , Uveitis/etiology , Uveitis/immunology , Uveitis/metabolism , Visual Acuity
11.
J Immunol ; 207(1): 322-332, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34145060

ABSTRACT

The adaptor protein TNFR-associated factor 3 (TRAF3) is required for in vivo T cell effector functions and for normal TCR/CD28 signaling. TRAF3-mediated enhancement of TCR function requires engagement of both CD3 and CD28, but the molecular mechanisms underlying how TRAF3 interacts with and impacts TCR/CD28-mediated complexes to enhance their signaling remains an important knowledge gap. We investigated how TRAF3 is recruited to, and regulates, CD28 as a TCR costimulator. Direct association with known signaling motifs in CD28 was dispensable for TRAF3 recruitment; rather, TRAF3 associated with the CD28-interacting protein linker of activated T cells (LAT) in human and mouse T cells. TRAF3-LAT association required the TRAF3 TRAF-C domain and a newly identified TRAF2/3 binding motif in LAT. TRAF3 inhibited function of the LAT-associated negative regulatory protein Dok1, which is phosphorylated at an inhibitory tyrosine residue by the tyrosine kinase breast tumor kinase (Brk/PTK6). TRAF3 regulated Brk activation in T cells, limiting the association of protein tyrosine phosphatase 1B (PTP1B) with the LAT complex. In TRAF3-deficient cells, LAT complex-associated PTP1B was associated with dephosphorylation of Brk at an activating tyrosine residue, potentially reducing its ability to inhibit Dok1. Consistent with these findings, inhibiting PTP1B activity in TRAF3-deficient T cells rescued basal and TCR/CD28-mediated activation of Src family kinases. These results reveal a new mechanism for promotion of TCR/CD28-mediated signaling through restraint of negative regulation of LAT by TRAF3, enhancing the understanding of regulation of the TCR complex.


Subject(s)
CD28 Antigens/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , TNF Receptor-Associated Factor 3/immunology , Animals , Cells, Cultured , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/immunology , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/genetics
12.
Cancer Immunol Immunother ; 70(11): 3093-3103, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33765210

ABSTRACT

Cancer vaccines that utilize patient antigen-presenting cells to fight their own tumors have shown exciting promise in many preclinical studies, but have proven quite challenging to translate to clinical feasibility. Dendritic cells have typically been the cell of choice for such vaccine platforms, due to their ability to endocytose antigens nonspecifically, and their expression of multiple surface molecules that enhance antigen presentation. However, dendritic cells are present in low numbers in human peripheral blood and must be matured in culture before use in vaccines. Mature B lymphocytes, in contrast, are relatively abundant in peripheral blood, and can be quickly activated and expanded in overnight cultures. We devised an optimal stimulation cocktail that engages the B cell antigen receptor, CD40, TLR4 and TLR7, to activate B cells to present antigens from lysates of the recipient's tumor cells, precluding the need for known tumor antigens. This B cell vaccine (Bvac) improved overall survival from B16F1 melanoma challenge, as well as reduced tumor size and increased time to tumor appearance. Bvac upregulated B cell antigen presentation molecules, stimulated activation of both CD4+ and CD8+ T cells, and induced T cell migration. Bvac provides an alternative cellular vaccine strategy that has considerable practical advantages for translation to clinical settings.


Subject(s)
Antigens, Neoplasm/immunology , B-Lymphocytes/immunology , Cancer Vaccines/immunology , Melanoma, Experimental/immunology , Skin Neoplasms/immunology , Animals , Antigen Presentation/immunology , Cancer Vaccines/pharmacology , Chemotaxis, Leukocyte/immunology , Female , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology
13.
J Biol Chem ; 296: 100465, 2021.
Article in English | MEDLINE | ID: mdl-33639170

ABSTRACT

TRAF3 has diverse signaling functions, which vary by cell type. Uniquely in B lymphocytes, TRAF3 inhibits homeostatic survival. Highlighting the role of TRAF3 as a tumor suppressor, loss-of-function TRAF3 mutations are associated with human B-cell malignancies, while B-cell-specific deletion of TRAF3 in mice leads to autoimmunity and lymphoma development. The role of TRAF3 in inhibiting noncanonical NF-κB activation, CD40 and BAFF-R signaling to B cells is well documented. In contrast, TRAF3 enhances many T-cell effector functions, through associating with and enhancing signaling by the T-cell receptor (TCR)-CD28 complex. The present study was designed to determine the role of TRAF3 in signaling via the B-cell antigen receptor (BCR). The BCR is crucial for antigen recognition, survival, proliferation, and antibody production, and defects in BCR signaling can promote abnormal survival of malignant B cells. Here, we show that TRAF3 is associated with both CD79B and the BCR-activated kinases Syk and Btk following BCR stimulation. BCR-induced phosphorylation of Syk and additional downstream kinases was increased in TRAF3-/- B cells, with regulation observed in both follicular and marginal zone B-cell subsets. BCR stimulation of TRAF3-/- B cells resulted in increased surface expression of MHC-II, CD80, and CD86 molecules. Interestingly, increased survival of TRAF3-/- primary B cells was resistant to inhibition of Btk, while TRAF3-deficient malignant B-cell lines showed enhanced sensitivity. TRAF3 serves to restrain normal and malignant BCR signaling, with important implications for its role in normal B-cell biology and abnormal survival of malignant B cells.


Subject(s)
Receptors, Antigen, B-Cell/metabolism , TNF Receptor-Associated Factor 3/metabolism , Agammaglobulinaemia Tyrosine Kinase/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD79 Antigens/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Receptors, Antigen, B-Cell/genetics , Signal Transduction/genetics , Syk Kinase/metabolism , T-Lymphocytes/metabolism , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 3/genetics
14.
J Leukoc Biol ; 109(2): 309-325, 2021 02.
Article in English | MEDLINE | ID: mdl-32441445

ABSTRACT

Many acute viral infections target tissue Mϕs, yet the mechanisms of Mϕ-mediated control of viruses are poorly understood. Here, we report that CD40 expressed by peritoneal Mϕs restricts early infection of a broad range of RNA viruses. Loss of CD40 expression enhanced virus replication as early as 12-24 h of infection and, conversely, stimulation of CD40 signaling with an agonistic Ab blocked infection. With peritoneal cell populations infected with the filovirus, wild-type (WT) Ebola virus (EBOV), or a BSL2 model virus, recombinant vesicular stomatitis virus encoding Ebola virus glycoprotein (rVSV/EBOV GP), we examined the mechanism conferring protection. Here, we demonstrate that restricted virus replication in Mϕs required CD154/CD40 interactions that stimulated IL-12 production through TRAF6-dependent signaling. In turn, IL-12 production resulted in IFN-γ production, which induced proinflammatory polarization of Mϕs, protecting the cells from infection. These CD40-dependent events protected mice against virus challenge. CD40-/- mice were exquisitely sensitive to intraperitoneal challenge with a dose of rVSV/EBOV GP that was sublethal to CD40+/+ mice, exhibiting viremia within 12 h of infection and rapidly succumbing to infection. This study identifies a previously unappreciated role for Mϕ-intrinsic CD40 signaling in controlling acute virus infection.


Subject(s)
CD40 Antigens/metabolism , Immunity, Innate , Macrophages/immunology , Macrophages/virology , RNA Viruses/physiology , Signal Transduction , Virus Diseases/immunology , Virus Replication/physiology , Acute Disease , Animals , CD40 Ligand/metabolism , Ebolavirus/physiology , Glycoproteins/immunology , Humans , Interferon-gamma/metabolism , Interleukin-12/biosynthesis , Mice, Inbred C57BL , Models, Biological , Peritoneum/pathology , Peritoneum/virology , TNF Receptor-Associated Factor 6/metabolism , Virus Diseases/virology
15.
Front Immunol ; 11: 606936, 2020.
Article in English | MEDLINE | ID: mdl-33613527

ABSTRACT

Systemic lupus erythematosus (SLE) and other autoimmune diseases are propelled by immune dysregulation and pathogenic, disease-specific autoantibodies. Autoimmunity against the lupus autoantigen Sm is associated with cross-reactivity to Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA-1). Additionally, EBV latent membrane protein-1 (LMP1), initially noted for its oncogenic activity, is an aberrantly active functional mimic of the B cell co-stimulatory molecule CD40. Mice expressing a transgene (Tg) for the mCD40-LMP1 hybrid molecule (containing the cytoplasmic tail of LMP1) have mild autoantibody production and other features of immune dysregulation by 2-3 months of age, but no overt autoimmune disease. This study evaluates whether exposure to the EBV molecular mimic, EBNA-1, stimulates antigen-specific and concurrently-reactive humoral and cellular immunity, as well as lupus-like features. After immunization with EBNA-1, mCD40-LMP1 Tg mice exhibited enhanced, antigen-specific, cellular and humoral responses compared to immunized WT congenic mice. EBNA-1 specific proliferative and inflammatory cytokine responses, including IL-17 and IFN-γ, were significantly increased (p<0.0001) in mCD40-LMP1 Tg mice, as well as antibody responses to amino- and carboxy-domains of EBNA-1. Of particular interest was the ability of mCD40-LMP1 to drive EBNA-1 associated molecular mimicry with the lupus-associated autoantigen, Sm. EBNA-1 immunized mCD40-LMP1 Tg mice exhibited enhanced proliferative and cytokine cellular responses (p<0.0001) to the EBNA-1 homologous epitope PPPGRRP and the Sm B/B' cross-reactive sequence PPPGMRPP. When immunized with the SLE autoantigen Sm, mCD40-LMP1 Tg mice again exhibited enhanced cellular and humoral immune responses to both Sm and EBNA-1. Cellular immune dysregulation with EBNA-1 immunization in mCD40-LMP1 Tg mice was accompanied by enhanced splenomegaly, increased serum blood urea nitrogen (BUN) and creatinine levels, and elevated anti-dsDNA and antinuclear antibody (ANA) levels (p<0.0001 compared to mCD40 WT mice). However, no evidence of immune-complex glomerulonephritis pathology was noted, suggesting that a combination of EBV and genetic factors may be required to drive lupus-associated renal disease. These data support that the expression of LMP1 in the context of EBNA-1 may interact to increase immune dysregulation that leads to pathogenic, autoantigen-specific lupus inflammation.


Subject(s)
Autoantigens/immunology , Autoimmunity , Epstein-Barr Virus Nuclear Antigens/immunology , Immunity, Cellular , Immunity, Humoral , Lupus Erythematosus, Systemic/immunology , Molecular Mimicry , Viral Matrix Proteins/immunology , snRNP Core Proteins/immunology , Animals , Antibodies, Antinuclear/blood , Autoantigens/administration & dosage , CD40 Antigens/genetics , CD40 Antigens/immunology , CD40 Antigens/metabolism , Cross Reactions , Epitopes , Epstein-Barr Virus Nuclear Antigens/administration & dosage , Immunization , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/genetics , Mice, Inbred C57BL , Mice, Transgenic , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , snRNP Core Proteins/administration & dosage
16.
J Leukoc Biol ; 107(6): 907-915, 2020 06.
Article in English | MEDLINE | ID: mdl-31749173

ABSTRACT

The TNFR superfamily of receptors, the major focus of the recent TNFR Superfamily Conference held in June 2019, employ the TNFR-associated factor (TRAF) family of adaptor proteins in key aspects of their signaling pathways. Although many early studies investigated TRAF functions via exogenous overexpression in nonhematopoietic cell lines, it has subsequently become clear that whereas TRAFs share some overlap in function, each also plays unique biologic roles, that can be highly context dependent. This brief review summarizes the current state of knowledge of functions of each of the TRAF molecules that mediate important functions in T lymphocytes: TRAFs 1, 2, 3, 5, and 6. Due to our current appreciation of the contextual nature of TRAF function, our focus is upon findings made specifically in T lymphocytes. Key T cell functions for each TRAF are detailed, as well as future knowledge gaps of interest and importance.


Subject(s)
Immunologic Deficiency Syndromes/genetics , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Gene Expression Regulation , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/pathology , Immunologic Memory , Mice , Mice, Knockout , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction/immunology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/deficiency , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/immunology , Tumor Necrosis Factor-alpha/immunology
17.
Sci Rep ; 9(1): 17502, 2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31745166

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

18.
Sci Rep ; 9(1): 12884, 2019 09 09.
Article in English | MEDLINE | ID: mdl-31501481

ABSTRACT

TRAF3 is a versatile intracellular adapter protein with multiple context-specific roles. Uniquely in B cells, TRAF3 deficiency enhances survival and increases the risk of transformation, as loss of TRAF3 is observed in several types of B cell cancers. Here, we report a new mechanism for TRAF3 in the restraint of B cell survival. We found that TRAF3 deficiency was associated with induction of the pro-survival kinase Pim2 in mouse primary B cells and human malignant B cell lines. The increase in Pim2 was independent of NF-κB2 activation but was ameliorated with inhibition of STAT3 expression or function. TRAF3 deficiency also led to a Pim2-dependent increase in c-Myc protein levels and was associated with reduced c-Myc ubiquitination. TRAF3-deficient primary B cells were less sensitive to cell death induced by the Pim inhibitors SGI-1776 and TP-3654. Interestingly, human malignant B cell lines with low expression of TRAF3 were more sensitive to Pim inhibition-induced cell death. Combination treatment of TRAF3-deficient B cells and B cell tumor lines with c-Myc inhibitors enhanced their sensitivity to Pim inhibition, suggesting a possible therapeutic strategy. TRAF3 thus suppresses a Pim2-mediated B cell survival axis, which can be a potential target for treatment of B cell malignancies.


Subject(s)
Gene Expression Regulation, Neoplastic , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins/metabolism , TNF Receptor-Associated Factor 3/metabolism , Animals , B-Lymphocytes/metabolism , Cell Line, Tumor , Cell Survival , Humans , Mice , Phosphorylation , STAT3 Transcription Factor/metabolism , TNF Receptor-Associated Factor 3/deficiency
19.
Blood ; 134(4): 363-373, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31101621

ABSTRACT

Targeting the B-cell receptor and phosphatidylinositol 3-kinase/mTOR signaling pathways has shown meaningful, but incomplete, antitumor activity in lymphoma. Glycogen synthase kinase 3 (GSK3) α and ß are 2 homologous and functionally overlapping serine/threonine kinases that phosphorylate multiple protein substrates in several key signaling pathways. To date, no agent targeting GSK3 has been approved for lymphoma therapy. We show that lymphoma cells abundantly express GSK3α and GSK3ß compared with normal B and T lymphocytes at the messenger RNA and protein levels. Utilizing a new GSK3 inhibitor 9-ING-41 and by genetic deletion of GSK3α and GSK3ß genes using CRISPR/CAS9 knockout, GSK3 was demonstrated to be functionally important to lymphoma cell growth and proliferation. GSK3ß binds to centrosomes and microtubules, and lymphoma cells treated with 9-ING-41 become arrested in mitotic prophase, supporting the notion that GSK3ß is necessary for the progression of mitosis. By analyzing recently published RNA sequencing data on 234 diffuse large B-cell lymphoma patients, we found that higher expression of GSK3α or GSK3ß correlates well with shorter overall survival. These data provide rationale for testing GSK3 inhibitors in lymphoma patient trials.


Subject(s)
Glycogen Synthase Kinase 3/genetics , Lymphoma/etiology , Molecular Targeted Therapy , Animals , Biomarkers, Tumor , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Disease Models, Animal , Gene Expression , Gene Targeting/methods , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Indoles/pharmacology , Lymphoma/diagnosis , Lymphoma/mortality , Lymphoma/therapy , Maleimides/pharmacology , Mice , Mice, Transgenic , Mitosis/drug effects , Mitosis/genetics , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/methods , Spindle Apparatus/drug effects , Treatment Outcome , Xenograft Model Antitumor Assays
20.
J Clin Invest ; 129(7): 2888-2897, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31038471

ABSTRACT

Influenza A virus (IAV)-specific T cell responses are important correlates of protection during primary and subsequent infections. Generation and maintenance of robust IAV-specific T cell responses relies on T cell interactions with dendritic cells (DCs). In this study, we explore the role of nucleotide-binding domain leucine-rich repeat containing receptor family member NLRC4 in modulating the DC phenotype during IAV infection. Nlrc4-/- mice had worsened survival and increased viral titers during infection, normal innate immune cell recruitment and IAV-specific CD8 T cell responses, but severely blunted IAV-specific CD4 T cell responses compared to wild-type mice. The defect in the pulmonary IAV-specific CD4 T cell response was not a result of defective priming or migration of these cells in Nlrc4-/- mice but was instead due to an increase in FasL+ DCs, resulting in IAV-specific CD4 T cell death. Together, our data support a novel role for NLRC4 in regulating the phenotype of lung DCs during a respiratory viral infection, and thereby influencing the magnitude of protective T cell responses.


Subject(s)
Apoptosis Regulatory Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , Calcium-Binding Proteins/immunology , Dendritic Cells/immunology , Fas Ligand Protein/immunology , Gene Expression Regulation/immunology , Influenza A virus/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Animals , Apoptosis Regulatory Proteins/genetics , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , Calcium-Binding Proteins/genetics , Dendritic Cells/pathology , Fas Ligand Protein/genetics , Lung/pathology , Mice , Mice, Knockout , Orthomyxoviridae Infections/pathology
SELECTION OF CITATIONS
SEARCH DETAIL