Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
J Exp Med ; 218(5)2021 05 03.
Article in English | MEDLINE | ID: mdl-33724364

ABSTRACT

The spleen contains a myriad of conventional dendritic cell (cDC) subsets that protect against systemic pathogen dissemination by bridging antigen detection to the induction of adaptive immunity. How cDC subsets differentiate in the splenic environment is poorly understood. Here, we report that LTα1ß2-expressing Rorgt+ ILC3s, together with B cells, control the splenic cDC niche size and the terminal differentiation of Sirpα+CD4+Esam+ cDC2s, independently of the microbiota and of bone marrow pre-cDC output. Whereas the size of the splenic cDC niche depended on lymphotoxin signaling only during a restricted time frame, the homeostasis of Sirpα+CD4+Esam+ cDC2s required continuous lymphotoxin input. This latter property made Sirpα+CD4+Esam+ cDC2s uniquely susceptible to pharmacological interventions with LTßR agonists and antagonists and to ILC reconstitution strategies. Together, our findings demonstrate that LTα1ß2-expressing Rorgt+ ILC3s drive splenic cDC differentiation and highlight the critical role of ILC3s as perpetual regulators of lymphoid tissue homeostasis.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate , Lymphoid Tissue/immunology , Lymphotoxin-alpha/immunology , Signal Transduction/immunology , Spleen/immunology , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Dendritic Cells/metabolism , Female , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/immunology , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signal Transduction/genetics , Spleen/cytology , Spleen/metabolism
3.
Immunity ; 53(5): 1015-1032.e8, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33207209

ABSTRACT

Solitary intestinal lymphoid tissues such as cryptopatches (CPs) and isolated lymphoid follicles (ILFs) constitute steady-state activation hubs containing group 3 innate lymphoid cells (ILC3) that continuously produce interleukin (IL)-22. The outer surface of CPs and ILFs is demarcated by a poorly characterized population of CD11c+ cells. Using genome-wide single-cell transcriptional profiling of intestinal mononuclear phagocytes and multidimensional flow cytometry, we found that CP- and ILF-associated CD11c+ cells were a transcriptionally distinct subset of intestinal cDCs, which we term CIA-DCs. CIA-DCs required programming by CP- and ILF-resident CCR6+ ILC3 via lymphotoxin-ß receptor signaling in cDCs. CIA-DCs differentially expressed genes associated with immunoregulation and were the major cellular source of IL-22 binding protein (IL-22BP) at steady state. Mice lacking CIA-DC-derived IL-22BP exhibited diminished expression of epithelial lipid transporters, reduced lipid resorption, and changes in body fat homeostasis. Our findings provide insight into the design principles of an immunoregulatory checkpoint controlling nutrient absorption.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunity, Innate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Peyer's Patches/cytology , Peyer's Patches/immunology , Receptors, Interleukin/biosynthesis , Animals , Biomarkers , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Immunophenotyping , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Lipid Metabolism , Mice , Mice, Transgenic , RNA, Small Cytoplasmic/genetics , Receptors, Interleukin/genetics , Signal Transduction
4.
Nat Neurosci ; 23(7): 842-853, 2020 07.
Article in English | MEDLINE | ID: mdl-32424282

ABSTRACT

Many immunotherapies act by enhancing the ability of cytotoxic T cells to kill tumor cells. Killing depends on T cell recognition of antigens presented by class I major histocompatibility complex (MHC-I) proteins on tumor cells. In this study, we showed that medulloblastomas lacking the p53 tumor suppressor do not express surface MHC-I and are therefore resistant to immune rejection. Mechanistically, this is because p53 regulates expression of the peptide transporter Tap1 and the aminopeptidase Erap1, which are required for MHC-I trafficking to the cell surface. In vitro, tumor necrosis factor (TNF) or lymphotoxin-ß receptor agonist can rescue expression of Erap1, Tap1 and MHC-I on p53-mutant tumor cells. In vivo, low doses of TNF prolong survival and synergize with immune checkpoint inhibitors to promote tumor rejection. These studies identified p53 as a key regulator of immune evasion and suggest that TNF could be used to enhance sensitivity of tumors to immunotherapy.


Subject(s)
Cerebellar Neoplasms/immunology , Medulloblastoma/immunology , Tumor Escape/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Suppressor Protein p53/immunology , Animals , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Medulloblastoma/genetics , Medulloblastoma/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
J Biol Chem ; 292(51): 21060-21070, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29061848

ABSTRACT

The human cytomegalovirus opening reading frame UL144 is an ortholog of the TNF receptor superfamily member, herpesvirus entry mediator (HVEM; TNFRSF14). HVEM binds the TNF ligands, LIGHT and LTa; the immunoglobulin inhibitory receptor, B and T lymphocyte attenuator (BTLA); and the natural killer cell-activating receptor CD160. However, UL144 selectively binds BTLA, avoiding activation of inflammatory signaling initiated by CD160 in natural killer cells. BTLA and CD160 cross-compete for binding HVEM, but the structural basis for the ligand selectivity by UL144 and how it acts as an anti-inflammatory agonist remains unclear. Here, we modeled the UL144 structure and characterized its binding with BTLA. The UL144 structure was predicted to closely mimic the surface of HVEM, and we also found that both HVEM and UL144 bind a common epitope of BTLA, whether engaged in trans or in cis, that is shared with a BTLA antibody agonist. On the basis of the UL144 selectivity, we engineered a BTLA-selective HVEM protein to understand the basis for ligand selectivity and BTLA agonism to develop novel anti-inflammatory agonists. This HVEM mutein did not bind CD160 or TNF ligands but did bind BTLA with 10-fold stronger affinity than wild-type HVEM and retained potent inhibitory activity that reduced T-cell receptor, B-cell receptor, and interferon signaling in B cells. In conclusion, using a viral immune evasion strategy that shows broad immune-ablating activity, we have identified a novel anti-inflammatory BTLA-selective agonist.


Subject(s)
B-Lymphocytes/metabolism , Killer Cells, Natural/metabolism , Membrane Glycoproteins/metabolism , Models, Molecular , Receptors, Immunologic/agonists , Receptors, Tumor Necrosis Factor, Member 14/metabolism , T-Lymphocytes/metabolism , Viral Proteins/metabolism , Amino Acid Substitution , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antigens, CD/chemistry , Antigens, CD/genetics , Antigens, CD/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Binding Sites , Cell Line, Tumor , Drug Design , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HEK293 Cells , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Kinetics , Ligands , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mutation , Protein Conformation , Protein Engineering , Protein Interaction Domains and Motifs , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/chemistry , Receptors, Tumor Necrosis Factor, Member 14/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Viral Proteins/chemistry , Viral Proteins/genetics
6.
Gut ; 65(10): 1765-75, 2016 10.
Article in English | MEDLINE | ID: mdl-26206664

ABSTRACT

OBJECTIVES: The relative contributions of inflammatory signalling and sequential oncogenic dysregulation driving liver cancer pathogenesis remain incompletely understood. Lymphotoxin-ß receptor (LTßR) signalling is critically involved in hepatitis and liver tumorigenesis. Therefore, we explored the interdependence of inflammatory lymphotoxin signalling and specific oncogenic pathways in the progression of hepatic cancer. DESIGN: Pathologically distinct liver tumours were initiated by hydrodynamic transfection of oncogenic V-Akt Murine Thymoma Viral Oncogene Homolog 1 (AKT)/ß-catenin or AKT/Notch expressing plasmids. To investigate the relationship of LTßR signalling and specific oncogenic pathways, LTßR antagonist (LTßR-Fc) or agonist (anti-LTßR) were administered post oncogene transfection. Initiated livers/tumours were investigated for changes in oncogene expression, tumour proliferation, progression, latency and pathology. Moreover, specific LTßR-mediated molecular events were investigated in human liver cancer cell lines and through transcriptional analyses of samples from patients with intrahepatic cholangiocarcinoma (ICC). RESULTS: AKT/ß-catenin-transfected livers displayed increased expression of LTß and LTßR, with antagonism of LTßR signalling reducing tumour progression and enhancing survival. Conversely, enforced LTßR-activation of AKT/ß-catenin-initiated tumours induced robust increases in proliferation and progression of hepatic tumour phenotypes in an AKT-dependent manner. LTßR-activation also rapidly accelerated ICC progression initiated by AKT/Notch, but not Notch alone. Moreover, LTßR-accelerated development coincides with increases of Notch, Hes1, c-MYC, pAKT and ß-catenin. We further demonstrate LTßR signalling in human liver cancer cell lines to be a regulator of Notch, pAKTser473 and ß-catenin. Transcriptome analysis of samples from patients with ICC links increased LTßR network expression with poor patient survival, increased Notch1 expression and Notch and AKT/PI3K signalling. CONCLUSIONS: Our findings link LTßR and oncogenic AKT signalling in the development of ICC.


Subject(s)
Carcinogenesis/metabolism , Cholangiocarcinoma , Liver Neoplasms , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-beta/metabolism , Signal Transduction/physiology , Animals , Cell Proliferation/physiology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Disease Progression , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Statistics as Topic
7.
EMBO J ; 34(4): 466-74, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25599993

ABSTRACT

Antinuclear antibodies are a hallmark feature of generalized autoimmune diseases, including systemic lupus erythematosus and systemic sclerosis. However, the processes underlying the loss of tolerance against nuclear self-constituents remain largely unresolved. Using mice deficient in lymphotoxin and Hox11, we report that approximately 25% of mice lacking secondary lymphoid organs spontaneously develop specific antinuclear antibodies. Interestingly, we find this phenotype is not caused by a defect in central tolerance. Rather, cell-specific deletion and in vivo lymphotoxin blockade link these systemic autoimmune responses to the formation of gut-associated lymphoid tissue in the neonatal period of life. We further demonstrate antinuclear antibody production is influenced by the presence of commensal gut flora, in particular increased colonization with segmented filamentous bacteria, and IL-17 receptor signaling. Together, these data indicate that neonatal colonization of gut microbiota influences generalized autoimmunity in adult life.


Subject(s)
Autoimmunity/immunology , Microbiota/immunology , Animals , Antibodies, Antinuclear/genetics , Antibodies, Antinuclear/immunology , Autoimmunity/genetics , Female , Flow Cytometry , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
8.
J Immunol ; 191(9): 4611-8, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24078690

ABSTRACT

Innate lymphoid cells encompass a diverse array of lymphocyte subsets with unique phenotype that initiate inflammation and provide host defenses in specific microenvironments. In this study, we identify a rare human CD4(+)CD3(-) innate-like lymphoid population with high TNF expression that is enriched in blood from patients with rheumatoid arthritis. These CD4(+)CD3(-) cells belong to the T cell lineage, but the lack of AgR at the cell surface renders them nonresponsive to TCR-directed stimuli. By developing a culture system that sustains survival, we show that CD4(+)CD3(-) innate-like T cells display IL-7-dependent induction of surface lymphotoxin-αß, demonstrating their potential to modify tissue microenvironments. Furthermore, expression of CCR6 on the CD4(+)CD3(-) population defines a CD127(high) subset that is highly responsive to IL-7. This CD4(+)CD3(-) population is enriched in the peripheral blood from rheumatoid arthritis patients, suggesting a link to their involvement in chronic inflammatory disease.


Subject(s)
Arthritis, Rheumatoid/metabolism , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/metabolism , Lymphotoxin alpha1, beta2 Heterotrimer/metabolism , Tumor Necrosis Factors/metabolism , Adult , Aged , Animals , Arthritis, Rheumatoid/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Female , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Inflammation , Interleukin-7/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphotoxin alpha1, beta2 Heterotrimer/immunology , Male , Mice , Mice, Knockout , Middle Aged , Receptors, CCR6/metabolism
9.
J Immunol ; 191(2): 828-36, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23761635

ABSTRACT

Lymphocyte activation is regulated by costimulatory and inhibitory receptors, of which both B and T lymphocyte attenuator (BTLA) and CD160 engage herpesvirus entry mediator (HVEM). Notably, it remains unclear how HVEM functions with each of its ligands during immune responses. In this study, we show that HVEM specifically activates CD160 on effector NK cells challenged with virus-infected cells. Human CD56(dim) NK cells were costimulated specifically by HVEM but not by other receptors that share the HVEM ligands LIGHT, Lymphotoxin-α, or BTLA. HVEM enhanced human NK cell activation by type I IFN and IL-2, resulting in increased IFN-γ and TNF-α secretion, and tumor cell-expressed HVEM activated CD160 in a human NK cell line, causing rapid hyperphosphorylation of serine kinases ERK1/2 and AKT and enhanced cytolysis of target cells. In contrast, HVEM activation of BTLA reduced cytolysis of target cells. Together, our results demonstrate that HVEM functions as a regulator of immune function that activates NK cells via CD160 and limits lymphocyte-induced inflammation via association with BTLA.


Subject(s)
Antigens, CD/metabolism , Killer Cells, Natural/immunology , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/metabolism , CD56 Antigen/metabolism , Cell Line , Enzyme Activation , GPI-Linked Proteins/metabolism , HEK293 Cells , Humans , Inflammation , Interferon Type I/metabolism , Interferon-gamma/metabolism , Interleukin-2/metabolism , Killer Cells, Natural/metabolism , Lymphocyte Activation , Lymphotoxin-alpha/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Tumor Necrosis Factor, Member 14/immunology , Signal Transduction , T-Lymphocytes/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Cell Host Microbe ; 13(3): 324-35, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23498957

ABSTRACT

Death receptors (DRs) of the TNFR superfamily contribute to antiviral immunity by promoting apoptosis and regulating immune homeostasis during infection, and viral inhibition of DR signaling can alter immune defenses. Here we identify the human cytomegalovirus (HCMV) UL141 glycoprotein as necessary and sufficient to restrict TRAIL DR function. Despite showing no primary sequence homology to TNF family cytokines, UL141 binds the ectodomains of both human TRAIL DRs with affinities comparable to the natural ligand TRAIL. UL141 binding promotes intracellular retention of the DRs, thus protecting virus infected cells from TRAIL and TRAIL-dependent NK cell-mediated killing. The identification of UL141 as a herpesvirus modulator of the TRAIL DRs strongly implicates this pathway as a regulator of host defense to HCMV and highlights UL141 as a pleiotropic inhibitor of NK cell effector function.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Immunity, Innate , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Viral Proteins/immunology , Viral Proteins/metabolism , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Host-Pathogen Interactions , Humans , Killer Cells, Natural/immunology , Membrane Glycoproteins/genetics , Protein Binding , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Viral Proteins/genetics
11.
J Immunol ; 188(10): 4885-96, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22490863

ABSTRACT

The cosignaling network mediated by the herpesvirus entry mediator (HVEM; TNFRSF14) functions as a dual directional system that involves proinflammatory ligand, lymphotoxin that exhibits inducible expression and competes with HSV glycoprotein D for HVEM, a receptor expressed by T lymphocytes (LIGHT; TNFSF14), and the inhibitory Ig family member B and T lymphocyte attenuator (BTLA). To dissect the differential contributions of HVEM/BTLA and HVEM/LIGHT interactions, topographically-specific, competitive, and nonblocking anti-HVEM Abs that inhibit BTLA binding, but not LIGHT, were developed. We demonstrate that a BTLA-specific competitor attenuated the course of acute graft-versus-host reaction in a murine F(1) transfer semiallogeneic model. Selective HVEM/BTLA blockade did not inhibit donor T cell infiltration into graft-versus-host reaction target organs, but decreased the functional activity of the alloreactive T cells. These results highlight the critical role of HVEM/BTLA pathway in the control of the allogeneic immune response and identify a new therapeutic target for transplantation and autoimmune diseases.


Subject(s)
Graft vs Host Reaction/immunology , Receptors, Immunologic/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Member 14/antagonists & inhibitors , Signal Transduction/immunology , Adoptive Transfer , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/transplantation , CHO Cells , Cell Movement/genetics , Cell Movement/immunology , Cricetinae , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rats , Rats, Inbred Lew , Receptors, Immunologic/physiology , Receptors, Tumor Necrosis Factor, Member 14/administration & dosage , Receptors, Tumor Necrosis Factor, Member 14/genetics , Recombinant Fusion Proteins/administration & dosage , Spleen/cytology , Spleen/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation
12.
Nat Med ; 17(5): 596-603, 2011 May.
Article in English | MEDLINE | ID: mdl-21499267

ABSTRACT

Individuals with chronic asthma show a progressive decline in lung function that is thought to be due to structural remodeling of the airways characterized by subepithelial fibrosis and smooth muscle hyperplasia. Here we show that the tumor necrosis factor (TNF) family member LIGHT is expressed on lung inflammatory cells after allergen exposure. Pharmacological inhibition of LIGHT using a fusion protein between the IgG Fc domain and lymphotoxin ß receptor (LTßR) reduces lung fibrosis, smooth muscle hyperplasia and airway hyperresponsiveness in mouse models of chronic asthma, despite having little effect on airway eosinophilia. LIGHT-deficient mice also show a similar impairment in fibrosis and smooth muscle accumulation. Blockade of LIGHT suppresses expression of lung transforming growth factor-ß (TGF-ß) and interleukin-13 (IL-13), cytokines implicated in remodeling in humans, whereas exogenous administration of LIGHT to the airways induces fibrosis and smooth muscle hyperplasia, Thus, LIGHT may be targeted to prevent asthma-related airway remodeling.


Subject(s)
Airway Remodeling/physiology , Asthma/physiopathology , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology , Animals , Asthma/etiology , Asthma/pathology , Disease Models, Animal , Humans , Inflammation Mediators/physiology , Interleukin-13/physiology , Lung/pathology , Lung/physiopathology , Lymphotoxin alpha1, beta2 Heterotrimer/antagonists & inhibitors , Lymphotoxin alpha1, beta2 Heterotrimer/physiology , Mice , Mice, Knockout , Signal Transduction , Transforming Growth Factor beta/physiology , Tumor Necrosis Factor Ligand Superfamily Member 14/antagonists & inhibitors , Tumor Necrosis Factor Ligand Superfamily Member 14/deficiency , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
13.
J Exp Med ; 208(4): 797-809, 2011 Apr 11.
Article in English | MEDLINE | ID: mdl-21402741

ABSTRACT

Memory T helper cells (Th cells) play an important role in host defense against pathogens but also contribute to the pathogenesis of inflammatory disorders. We found that a soluble decoy lymphotoxin ß receptor (LT-ßR)-Fc, which can block tumor necrosis factor (TNF)-related ligands LIGHT (TNFSF14) and LT-αß binding to the herpesvirus entry mediator (HVEM) and the LT-ßR, inhibited the accumulation of memory Th2 cells after antigen encounter and correspondingly reduced inflammatory responses in vivo. Showing that this was a function of the receptor for LIGHT, antigen-specific memory CD4 T cells deficient in HVEM were also unable to persist, despite having a normal immediate response to recall antigen. HVEM(-/-) memory Th2 cells displayed reduced activity of PKB (protein kinase B; Akt), and constitutively active Akt rescued their survival and restored strong inflammation after antigen rechallenge. This was not restricted to Th2 memory cells as HVEM-deficient Th1 memory cells were also impaired in surviving after encounter with recall antigen. Furthermore, the absence of LIGHT on T cells recapitulated the defect seen with the absence of HVEM, suggesting that activated T cells communicate through LIGHT-HVEM interactions. Collectively, our results demonstrate a critical role of HVEM signals in the persistence of large pools of memory CD4 T cells.


Subject(s)
Immunologic Memory , Receptors, Tumor Necrosis Factor, Member 14/physiology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Survival , Female , Inflammation/etiology , Lymphotoxin beta Receptor/pharmacology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes, Helper-Inducer/cytology , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology
14.
J Immunol ; 185(3): 1949-58, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20592286

ABSTRACT

The TNF superfamily member homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for herpesvirus entry mediator (HVEM), a receptor expressed by T lymphocytes (LIGHT) [TNF superfamily (SF)-14], is a key cytokine that activates T cells and dendritic cells and is implicated as a mediator of inflammatory, metabolic, and malignant diseases. LIGHT engages the lymphotoxin-beta receptor (LTbetaR) and HVEM (TNFRSF14), but is competitively limited in activating these receptors by soluble decoy receptor-3 (DcR3; TNFRSF6B). Two variants in the human LIGHT alter the protein at E214K (rs344560) in the receptor-binding domain and S32L (rs2291667) in the cytosolic domain; however, the functional impact of these polymorphisms is unknown. A neutralizing Ab failed to bind the LIGHT-214K variant, indicating this position as a part of the receptor-binding region. Relative to the predominant reference variant S32/E214, the other variants showed altered avidity with LTbetaR and less with HVEM. Heterotrimers of the LIGHT variants decreased binding avidity to DcR3 and minimized the inhibitory effect of DcR3 toward LTbetaR-induced activation of NF-kappaB. In patients with immune-mediated inflammatory diseases, such as rheumatoid arthritis, DcR3 protein levels were significantly elevated. Immunohistochemistry revealed synoviocytes as a significant source of DcR3 production, and DcR3 hyperexpression is controlled by posttranscriptional mechanisms. The increased potential for LTbetaR signaling, coupled with increased bioavailability due to lower DcR3 avidity, provides a mechanism of how polymorphic variants in LIGHT could contribute to the pathogenesis of inflammatory diseases.


Subject(s)
Genetic Variation/immunology , Polymorphism, Single Nucleotide/immunology , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Amino Acid Sequence , Biological Availability , Coculture Techniques , HeLa Cells , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Models, Immunological , Molecular Sequence Data , Mutagenesis, Site-Directed , NF-kappa B/antagonists & inhibitors , Protein Binding/genetics , Protein Binding/immunology , Receptors, Tumor Necrosis Factor, Member 14/physiology , Receptors, Tumor Necrosis Factor, Member 6b/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology
15.
J Biol Chem ; 285(22): 17148-55, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20348096

ABSTRACT

The lymphotoxin-beta receptor (LTbetaR) activates the NF-kappaB2 transcription factors, p100 and RelB, by regulating the NF-kappaB-inducing kinase (NIK). Constitutive proteosomal degradation of NIK limits NF-kappaB activation in unstimulated cells by the ubiquitin:NIK E3 ligase comprised of subunits TNFR-associated factors (TRAF)3, TRAF2, and cellular inhibitor of apoptosis (cIAP). However, the mechanism releasing NIK from constitutive degradation remains unclear. We found that insertion of a charge-repulsion mutation in the receptor-binding crevice of TRAF3 ablated binding of both LTbetaR and NIK suggesting a common recognition site. A homologous mutation in TRAF2 inhibited cIAP interaction and blocked NIK degradation. Furthermore, the recruitment of TRAF3 and TRAF2 to the ligated LTbetaR competitively displaced NIK from TRAF3. Ligated LTbetaR complexed with TRAF3 and TRAF2 redirected the specificity of the ubiquitin ligase reaction to polyubiquitinate TRAF3 and TRAF2, leading to their proteosomal degradation. Stimulus-dependent degradation of TRAF3 required the RING domain of TRAF2, but not of TRAF3, implicating TRAF2 as a key E3 ligase in TRAF turnover. The combined action of competitive displacement of NIK and TRAF degradation halted NIK turnover, and promoted its association with IKKalpha and signal transmission. These results indicate the LTbetaR modifies the ubiquitin:NIK E3 ligase, and also acts as an allosteric regulator of the ubiquitin:TRAF E3 ligase.


Subject(s)
Gene Expression Regulation, Enzymologic , Lymphotoxin beta Receptor/chemistry , Protein Serine-Threonine Kinases/chemistry , TNF Receptor-Associated Factor 3/chemistry , Ubiquitin-Protein Ligases/chemistry , Ubiquitin/chemistry , Allosteric Site , Animals , Binding Sites , Humans , I-kappa B Kinase/metabolism , Mice , Mice, Transgenic , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases/metabolism , NF-kappaB-Inducing Kinase
16.
J Immunol ; 183(11): 7286-96, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19915044

ABSTRACT

The inhibitory cosignaling pathway formed between the TNF receptor herpesvirus entry mediator (HVEM, TNFRSF14) and the Ig superfamily members, B and T lymphocyte attenuator (BTLA) and CD160, limits the activation of T cells. However, BTLA and CD160 can also serve as activating ligands for HVEM when presented in trans by adjacent cells, thus forming a bidirectional signaling pathway. BTLA and CD160 can directly activate the HVEM-dependent NF-kappaB RelA transcriptional complex raising the question of how NF-kappaB activation is repressed in naive T cells. In this study, we show BTLA interacts with HVEM in cis, forming a heterodimeric complex in naive T cells that inhibits HVEM-dependent NF-kappaB activation. The cis-interaction between HVEM and BTLA is the predominant form expressed on the surface of naive human and mouse T cells. The BTLA ectodomain acts as a competitive inhibitor blocking BTLA and CD160 from binding in trans to HVEM and initiating NF-kappaB activation. The TNF-related ligand, LIGHT (homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for HVEM, a receptor expressed by T lymphocytes, or TNFSF14) binds HVEM in the cis-complex, but NF-kappaB activation was attenuated, suggesting BTLA prevents oligomerization of HVEM in the cis-complex. Genetic deletion of BTLA or pharmacologic disruption of the HVEM-BTLA cis-complex in T cells promoted HVEM activation in trans. Interestingly, herpes simplex virus envelope glycoprotein D formed a cis-complex with HVEM, yet surprisingly, promoted the activation NF-kappaB RelA. We suggest that the HVEM-BTLA cis-complex competitively inhibits HVEM activation by ligands expressed in the surrounding microenvironment, thus helping maintain T cells in the naive state.


Subject(s)
Lymphocyte Activation/immunology , Receptors, Immunologic/immunology , Receptors, Tumor Necrosis Factor, Member 14/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Flow Cytometry , Humans , Immunoprecipitation , Mice , Mice, Knockout , Receptors, Immunologic/chemistry , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/chemistry , Receptors, Tumor Necrosis Factor, Member 14/metabolism , T-Lymphocytes/chemistry , T-Lymphocytes/metabolism
17.
Proc Natl Acad Sci U S A ; 106(15): 6244-9, 2009 Apr 14.
Article in English | MEDLINE | ID: mdl-19332782

ABSTRACT

The herpesvirus entry mediator (HVEM; TNFRSF14) activates NF-kappaB through the canonical TNF-related cytokine LIGHT, serving as a costimulatory pathway during activation of T cells. HVEM also functions as a ligand for the Ig superfamily members B and T lymphocyte attenuator (BTLA) and CD160, both of which limit inflammatory responses initiated by T cells. Emerging evidence indicates BTLA also promotes T cell survival, but its structural differences from LIGHT intimate BTLA is unlikely to function as an activator of HVEM. We demonstrate here that BTLA, CD160, and herpes simplex virus envelope glycoprotein D (gD) function as activating ligands for HVEM, promoting NF-kappaB activation and cell survival. Membrane-expressed BTLA and CD160, as well as soluble dimeric receptor surrogates BTLA-Fc and gD-Fc specifically activated HVEM-dependent NF-kappaB. BTLA and CD160 engagement induced recruitment of TNF receptor-associated factor 2 (TRAF2), but not TRAF3, to HVEM that specifically activated the RelA but not the RelB form of NF-kappaB in a mucosal epithelial tumor cell line. Moreover, Btla(-/-) T cells survived poorly following activation but were rescued with BTLA-Fc, indicating HVEM-BTLA bidirectional signaling may serve as a critical cell-survival system for lymphoid and epithelial cells.


Subject(s)
Receptors, Tumor Necrosis Factor, Member 14/immunology , Signal Transduction/immunology , Animals , Antigens, CD/immunology , Cell Line , Cell Survival/immunology , GPI-Linked Proteins , Humans , Immunoglobulins/immunology , Ligands , Lymphocyte Activation/immunology , Mice , Receptors, Immunologic/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , TNF Receptor-Associated Factor 2/metabolism , Transcription Factor RelA/metabolism , Viral Envelope Proteins/immunology
18.
J Immunol ; 180(2): 940-7, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18178834

ABSTRACT

The B and T lymphocyte attenuator (BTLA) appears to act as a negative regulator of T cell activation and growth. BTLA specifically interacts with herpesvirus entry mediator (HVEM), a member of the TNFR family. Herein, we have undertaken surface plasmon resonance studies to quantitatively assess BTLA and HVEM ectodomain interactions. We find that soluble BALB/cJ BTLA engages HVEM with an equilibrium affinity of 0.97+/-0.19 microM while the C57BL/6 BTLA binds slightly better with an equilibrium affinity of 0.42+/-0.06 microM. Despite its lower affinity for HVEM, the kinetic half-life of BALB/cJ BTLA complexes are twice as long as observed for C57BL/6 BTLA (4 vs 2 s). To further explore these interactions, we solved the crystal structure of a murine BTLA (BALB/cJ) ectodomain at 1.8-A resolution, revealing a beta sandwich fold with strong similarity to I-set members of the Ig superfamily. Using a structure-based mutagenesis strategy, we then examined the individual contributions of 26 BTLA surface-exposed residues toward HVEM binding. Four single-site substitutions were identified that decrease HVEM binding below detectable levels and two that decrease binding by more than half. All six of these cluster at the edge of the beta sandwich in a membrane distal patch formed primarily from the A and G strands. This patch falls within the contacting surface recently revealed in the crystal structure of the human BTLA-HVEM cocomplex. The critical binding residues identified here are highly conserved across species, suggesting that BTLA employs a conserved binding mode for HVEM recognition.


Subject(s)
Receptors, Immunologic/chemistry , Receptors, Tumor Necrosis Factor, Member 14/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Conserved Sequence , Crystallography, X-Ray , DNA Mutational Analysis , Dogs , Humans , Mice , Molecular Sequence Data , Protein Conformation , Protein Interaction Mapping , Rats , Receptors, Immunologic/genetics , Surface Plasmon Resonance
19.
J Immunol ; 180(1): 238-48, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18097025

ABSTRACT

Proliferation of dendritic cells (DC) in the spleen is regulated by positive growth signals through the lymphotoxin (LT)-beta receptor; however, the countering inhibitory signals that achieve homeostatic control are unresolved. Mice deficient in LTalpha, LTbeta, LTbetaR, and the NFkappaB inducing kinase show a specific loss of CD8- DC subsets. In contrast, the CD8alpha- DC subsets were overpopulated in mice deficient in the herpesvirus entry mediator (HVEM) or B and T lymphocyte attenuator (BTLA). HVEM- and BTLA-deficient DC subsets displayed a specific growth advantage in repopulating the spleen in competitive replacement bone marrow chimeric mice. Expression of HVEM and BTLA were required in DC and in the surrounding microenvironment, although DC expression of LTbetaR was necessary to maintain homeostasis. Moreover, enforced activation of the LTbetaR with an agonist Ab drove expansion of CD8alpha- DC subsets, overriding regulation by the HVEM-BTLA pathway. These results indicate the HVEM-BTLA pathway provides an inhibitory checkpoint for DC homeostasis in lymphoid tissue. Together, the LTbetaR and HVEM-BTLA pathways form an integrated signaling network regulating DC homeostasis.


Subject(s)
Dendritic Cells/immunology , Lymphotoxin beta Receptor/metabolism , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Animals , CD8 Antigens/analysis , Cell Proliferation , Homeostasis , Lymphotoxin beta Receptor/genetics , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Lymphotoxin-beta/genetics , Lymphotoxin-beta/metabolism , Mice , Mice, Mutant Strains , Receptors, Immunologic/genetics , Receptors, Tumor Necrosis Factor, Member 14/genetics
20.
Adv Exp Med Biol ; 597: 160-72, 2007.
Article in English | MEDLINE | ID: mdl-17633025

ABSTRACT

The lymphotoxin-beta receptor (LTbetaR, TNFRSF3) signaling pathway activates gene transcription programs and cell death important in immune development and host defense. The TNF receptor associated factors (TRAF)-2, 3 and 5 function as adaptors linking LTbetaR signaling targets. Interestingly, TRAF deficient mice do not phenocopy mice deficient in components of the LTbetaR pathway, presenting a conundrum. Here, an update of our understanding and models of the LTbetaR signaling pathway are reviewed, with a focus on this conundrum.


Subject(s)
Lymphotoxin beta Receptor/physiology , Signal Transduction/physiology , Animals , Humans , Lymphotoxin beta Receptor/deficiency , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...