Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.093
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 17(5): 593-603, 2016 May.
Article in English | MEDLINE | ID: mdl-26950238

ABSTRACT

Persistent viral infections are characterized by the simultaneous presence of chronic inflammation and T cell dysfunction. In prototypic models of chronicity--infection with human immunodeficiency virus (HIV) or lymphocytic choriomeningitis virus (LCMV)--we used transcriptome-based modeling to reveal that CD4(+) T cells were co-exposed not only to multiple inhibitory signals but also to tumor-necrosis factor (TNF). Blockade of TNF during chronic infection with LCMV abrogated the inhibitory gene-expression signature in CD4(+) T cells, including reduced expression of the inhibitory receptor PD-1, and reconstituted virus-specific immunity, which led to control of infection. Preventing signaling via the TNF receptor selectively in T cells sufficed to induce these effects. Targeted immunological interventions to disrupt the TNF-mediated link between chronic inflammation and T cell dysfunction might therefore lead to therapies to overcome persistent viral infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Tumor Necrosis Factor-alpha/immunology , Adolescent , Adult , Aged , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Flow Cytometry , HEK293 Cells , HIV/physiology , HIV Infections/genetics , HIV Infections/virology , Host-Pathogen Interactions/immunology , Humans , Immunoblotting , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Oligonucleotide Array Sequence Analysis , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome/drug effects , Transcriptome/genetics , Transcriptome/immunology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Young Adult
2.
Nat Immunol ; 16(8): 819-828, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26147686

ABSTRACT

Fat-associated lymphoid clusters (FALCs) are a type of lymphoid tissue associated with visceral fat. Here we found that the distribution of FALCs was heterogeneous, with the pericardium containing large numbers of these clusters. FALCs contributed to the retention of B-1 cells in the peritoneal cavity through high expression of the chemokine CXCL13, and they supported B cell proliferation and germinal center differentiation during peritoneal immunological challenges. FALC formation was induced by inflammation, which triggered the recruitment of myeloid cells that expressed tumor-necrosis factor (TNF) necessary for signaling via the TNF receptors in stromal cells. Natural killer T cells (NKT cells) restricted by the antigen-presenting molecule CD1d were likewise required for the inducible formation of FALCs. Thus, FALCs supported and coordinated the activation of innate B cells and T cells during serosal immune responses.


Subject(s)
Inflammation/immunology , Intra-Abdominal Fat/immunology , Lymphocytes/immunology , Lymphoid Tissue/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/immunology , Chemokine CXCL13/metabolism , Flow Cytometry , Gene Expression/immunology , Inflammation/genetics , Inflammation/metabolism , Intra-Abdominal Fat/metabolism , Lymphocytes/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Myeloid Cells/immunology , Myeloid Cells/metabolism , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/immunology , Stromal Cells/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
3.
Cell ; 137(6): 1112-23, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19524513

ABSTRACT

Programmed necrosis is a form of caspase-independent cell death whose molecular regulation is poorly understood. The kinase RIP1 is crucial for programmed necrosis, but also mediates activation of the prosurvival transcription factor NF-kappaB. We postulated that additional molecules are required to specifically activate programmed necrosis. Using a RNA interference screen, we identified the kinase RIP3 as a crucial activator for programmed necrosis induced by TNF and during virus infection. RIP3 regulates necrosis-specific RIP1 phosphorylation. The phosphorylation of RIP1 and RIP3 stabilizes their association within the pronecrotic complex, activates the pronecrotic kinase activity, and triggers downstream reactive oxygen species production. The pronecrotic RIP1-RIP3 complex is induced during vaccinia virus infection. Consequently, RIP3(-/-) mice exhibited severely impaired virus-induced tissue necrosis, inflammation, and control of viral replication. Our findings suggest that RIP3 controls programmed necrosis by initiating the pronecrotic kinase cascade, and that this is necessary for the inflammatory response against virus infections.


Subject(s)
GTPase-Activating Proteins/metabolism , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , Humans , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor/immunology , Tumor Necrosis Factor-alpha/immunology , Vaccinia/immunology , Vaccinia virus/metabolism
4.
Eur J Immunol ; 50(7): 944-948, 2020 07.
Article in English | MEDLINE | ID: mdl-32428262

ABSTRACT

Regulatory T (Treg) cells are crucial mediators of immune tolerance suppressing self-reactive T cells and preventing autoimmune diseases. Besides activation of the T cell receptor (TCR), empowerment of Treg cell functions requires co-accessory signals, such as those released by the TNF receptor superfamily (TNFRSF) that, however, can also promote immunostimulatory responses when engaged by effector T cells. In this issue of European Journal of Immunology, Lubrano di Ricco et al. [Eur. J. Immunol. 2020. 50: 972-985] have taken a closer look at the important question of the functional meaning of TNFRSF-activated signaling pathways in Treg cells. They have demonstrated that costimulation by TNFR2, 4-1BB, GITR, DR3, but not OX40 in mouse Foxp3+ Treg cells activates the same and unique signaling pathway, i.e., canonical NF-κB, which in turn leads to Foxp3 gene upregulation, cell expansion in vitro and in vivo, and suppressive activity in an experimental model of colitis. Moreover, induction of markers of T helper 2 (Th2) and Th17 as well as of genes encoding proteins involved in noncanonical NF-κΒ was also observed. We here discussed how these findings further highlight the emerging concept of Treg cell plasticity in immune tolerance.


Subject(s)
Immune Tolerance , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Mice , T-Lymphocytes, Regulatory/cytology
5.
Eur J Immunol ; 50(7): 972-985, 2020 07.
Article in English | MEDLINE | ID: mdl-32012260

ABSTRACT

Several drugs targeting members of the TNF superfamily or TNF receptor superfamily (TNFRSF) are widely used in medicine or are currently being tested in therapeutic trials. However, their mechanism of action remains poorly understood. Here, we explored the effects of TNFRSF co-stimulation on murine Foxp3+ regulatory T cell (Treg) biology, as they are pivotal modulators of immune responses. We show that engagement of TNFR2, 4-1BB, GITR, and DR3, but not OX40, increases Treg proliferation and survival. Triggering these TNFRSF in Tregs induces similar changes in gene expression patterns, suggesting that they engage common signal transduction pathways. Among them, we identified a major role of canonical NF-κB. Importantly, TNFRSF co-stimulation improves the ability of Tregs to suppress colitis. Our data demonstrate that stimulation of discrete TNFRSF members enhances Treg activation and function through a shared mechanism. Consequently, therapeutic effects of drugs targeting TNFRSF or their ligands may be mediated by their effect on Tregs.


Subject(s)
Lymphocyte Activation , NF-kappa B/immunology , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Mice , Mice, Knockout , NF-kappa B/genetics , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction/genetics , T-Lymphocytes, Regulatory/cytology
6.
Cell Immunol ; 370: 104441, 2021 12.
Article in English | MEDLINE | ID: mdl-34628221

ABSTRACT

Septic arthritis is a destructive joint disease caused by Staphylococcus aureus. Synovial inflammation involved Th17 proliferation and down regulation of Treg population, thus resolution of inflammation targeting IL-17 may be important to control arthritis. Endogenous inhibition of IL-17 to regulate arthritic inflammation correlating with Th17/Treg cells TLR2 and TNFRs are not done. The role of SOD, CAT and GRx in relation to ROS production during arthritis along with expression of TLR2, TNFR1/TNFR2 in Th17/Treg cells of mice treated with IL-17A Ab/ IL-2 were studied. Increased ROS, reduced antioxidant enzyme activity was found in Th17 cells of SA infected mice whereas Treg cells of IL-17A Ab/ IL-2 treated group showed opposite effects. Neutralization of IL-17 after arthritis cause decreased TNFR1 and increased TNFR2 expression in Treg cells. Thus, neutralization of IL-17 or IL-2 treatment regulates septic arthritis by enhancing anti-inflammatory properties of Treg via antioxidant balance and modulating TLR2/TNFR response.


Subject(s)
Arthritis, Infectious/immunology , Interleukin-17/antagonists & inhibitors , Interleukin-2/pharmacology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Antioxidants/metabolism , Arthritis, Experimental/immunology , Interleukin-17/immunology , Male , Mice , Reactive Oxygen Species/immunology , Receptors, Tumor Necrosis Factor/immunology , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Toll-Like Receptor 2/immunology
7.
Nat Immunol ; 10(4): 348-55, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19295631

ABSTRACT

Death receptors (DRs) are members of the tumor necrosis factor receptor superfamily that possess a cytoplasmic death domain (DD). DRs regulate important operational and homeostatic aspects of the immune system. They transmit signals through apical protein complexes, which are nucleated by the DD adaptors FADD and TRADD, to control cellular outcomes that range from apoptosis to gene activation. FADD and TRADD also nucleate several distal signaling complexes, which mediate cross-talk between distinct DR signaling pathways. Moreover, together with other DR signal transducers, FADD and TRADD participate in functional complexes assembled by certain non-DR immune cell receptors, such as pattern-recognition receptors. Thus, DR signal transducers may provide important nodes of coordination in immune signaling networks.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Fas-Associated Death Domain Protein/physiology , Receptors, Death Domain/physiology , Signal Transduction/physiology , TNF Receptor-Associated Death Domain Protein/physiology , Adaptor Proteins, Signal Transducing/immunology , Animals , Apoptosis/physiology , Fas-Associated Death Domain Protein/immunology , Humans , Immunity, Active , Immunity, Innate , Receptors, Death Domain/immunology , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/physiology , TNF Receptor-Associated Death Domain Protein/immunology
8.
Nat Immunol ; 10(4): 340-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19295630

ABSTRACT

An important function of receptors that signal through immunoreceptor tyrosine-based activation motifs (ITAMs) is to regulate signaling by heterologous receptors. This review describes mechanisms by which ITAM-associated receptors modulate signaling by Toll-like receptors (TLRs), tumor necrosis factor receptor family members and cytokine receptors that use the Jak-STAT signaling pathway, and the biological importance of this signal transduction cross-talk. ITAM-mediated cross-regulation can either augment or dampen signaling by other receptors. Conversely, TLRs and cytokines modulate ITAM-mediated signaling, by means including activation of beta2 integrins that are coupled to the ITAM-containing adaptors DAP12 and FcRgamma. Integration of ITAM signaling into signaling networks through cross-talk with other signal transduction pathways results in tight regulation and fine tuning of cellular responses to various extracellular stimuli and contributes to induction of specific activation and differentiation pathways.


Subject(s)
Amino Acid Motifs , Receptors, Cell Surface/physiology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/immunology , Animals , CD18 Antigens/immunology , CD18 Antigens/metabolism , Humans , Immunoglobulin Variable Region/immunology , Janus Kinases/immunology , Janus Kinases/physiology , Membrane Proteins/immunology , Receptor Cross-Talk/immunology , Receptors, Cell Surface/immunology , Receptors, Cytokine/immunology , Receptors, Cytokine/physiology , Receptors, Immunologic/immunology , Receptors, Immunologic/physiology , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/physiology , STAT Transcription Factors/immunology , STAT Transcription Factors/physiology , Toll-Like Receptors/immunology , Toll-Like Receptors/physiology
9.
J Immunol ; 203(7): 1766-1775, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31484730

ABSTRACT

Proinflammatory macrophages and miR-155 are increased in patients with rheumatoid arthritis (RA). We studied membrane TNF (mTNF) expression on blood monocytes, polarization into macrophages, miR-155 expression, and the effect of anti-TNF on these biomarkers in RA patients. Sixty-seven RA patients and 109 controls (55 healthy, 54 with spondyloarthritis and connective tissue diseases) were studied. Monocytes were isolated and differentiated into macrophages with or without anti-TNF. mTNF expression was increased on monocytes from RA patients, but not from other inflammatory diseases, correlated with disease activity. Under human serum AB or M-CSF, only monocytes from RA had a defect of differentiation into M2-like macrophages and had a propensity for preferential maturation toward M1-like macrophages that contributed to synovial inflammation. This defect was correlated to mTNF expression and was partially reversed by monoclonal anti-TNF Abs but not by the TNF soluble receptor. miR-155 was increased in M2-macrophages except in adalimumab-treated patients. Transfection of healthy monocytes with miR-155 induced a decrease in M2-like markers, and transfection of RA monocytes with antagomir-155 allowed restoration of M2-like polarization. Defect in differentiation of monocytes into M2-like-macrophages linked to increased miR-155 and correlated with increased mTNF on monocytes could play a key role in RA pathogenesis. Monoclonal anti-TNF Abs but not the TNF soluble receptor partially restored this defect.


Subject(s)
Arthritis, Rheumatoid/immunology , Cell Differentiation/immunology , Macrophage Activation , Macrophages/immunology , MicroRNAs/immunology , Monocytes/immunology , Adult , Aged , Antirheumatic Agents/administration & dosage , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Female , Humans , Macrophages/pathology , Male , Middle Aged , Monocytes/pathology , Receptors, Tumor Necrosis Factor/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
10.
Int Immunol ; 31(10): 639-648, 2019 09 18.
Article in English | MEDLINE | ID: mdl-30838383

ABSTRACT

Autoinflammatory syndromes are a group of disorders characterized by recurring episodes of inflammation as a result of specific defects in the innate immune system. Patients with autoinflammatory disease present with recurrent outbreaks of chronic systemic inflammation that are mediated by innate immune cells, for the most part. A number of these diseases arise from defects in the tumour necrosis factor receptor (TNFR) signalling pathway leading to elevated levels of inflammatory cytokines. Elucidation of the molecular mechanisms of these recently defined autoinflammatory diseases has led to a greater understanding of the mechanisms of action of key molecules involved in TNFR signalling, particularly those involved in ubiquitination, as found in haploinsufficiency of A20 (HA20), otulipenia/OTULIN-related autoinflammatory syndrome (ORAS) and linear ubiquitin chain assembly complex (LUBAC) deficiency. In this review, we also address other TNFR signalling disorders such as TNFR-associated periodic syndrome (TRAPS), RELA haploinsufficiency, RIPK1-associated immunodeficiency and autoinflammation, X-linked ectodermal dysplasia and immunodeficiency (X-EDA-ID) and we review the most recent advances surrounding these diseases and therapeutic approaches currently used to target these diseases. Finally, we explore therapeutic advances in TNF-related immune-based therapies and explore new approaches to target disease-specific modulation of autoinflammatory diseases.


Subject(s)
Autoimmune Diseases/immunology , Inflammation/immunology , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction , Animals , Autoimmune Diseases/genetics , Humans , Inflammation/genetics , Signal Transduction/immunology
11.
Crit Rev Immunol ; 39(6): 439-479, 2019.
Article in English | MEDLINE | ID: mdl-32421957

ABSTRACT

Tumor necrosis factor (TNF)-α, the most potent proinflammatory cytokine discovered to date, was first isolated in 1984 from human macrophage cells. Initially, it was thought to be a protein that was cytotoxic to tumor cells. But later, it was regarded as an agent that promotes inflammation and other chronic diseases found in humans. Currently, we know that the TNF superfamily (TNFS) has 19 members that perform a wide variety of functions via > 40 TNF receptors. Of TNFS members, TNF-α has been studied extensively and was found to be implicated in numerous autoimmune diseases, such as rheumatoid arthritis, ankylosing spondylitis, inflammatory bowel disease, psoriasis, systemic lupus erythematosus, juvenile idiopathic arthritis, and diabetes. Thus, agents that can inhibit TNF-α have great potential for prevention and treatment of chronic diseases. To date, the U.S. Food and Drug Administration has approved many TNF-α blockers, such as etanercept, infliximab, adalimumab, certolizumab pegol, and golimumab. These agents can block TNF-α actions and be used to treat different diseases. However, the uses of TNF-α blockers are not without serious adverse effects. Therefore, natural TNF-α blockers are best for developing safe, efficacious, and affordable agents for prevention and treatment of chronic diseases. The current review details the TNFS, functions of TNF-α in normal and disease conditions, roles of TNF-α blockers, and advantages and disadvantages.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Certolizumab Pegol/therapeutic use , Etanercept/therapeutic use , Immune System Diseases/therapy , Inflammation/therapy , Receptors, Tumor Necrosis Factor/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Humans , Immune System Diseases/immunology , Inflammation/immunology , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors
12.
Fish Shellfish Immunol ; 98: 147-159, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31923566

ABSTRACT

Proteins in the tumor necrosis factor receptor (TNFR) superfamily play significant roles in many physiological and pathological events, such as inflammation, apoptosis, autoimmunity, and organogenesis. Here, two TNFR gene homologs (PmTNFR1 and PmTNFR5) were identified in the pearl oyster Pinctada fucata martensii. The predicted PmTNFR1 and PmTNFR5 protein sequences were 406 and 533 amino acids long, respectively, and both possessed motifs characteristic of the TNFR family, including a TNFR homology domain (CRD), a transmembrane domain (TM), and death domains. However, the predicted amino acid sequences of PmTNFR1 and PmTNFR5 had low identity (~16-23%) with sequences of vertebrate TNFR family proteins. Furthermore, PmTNFR5 had a death domain at the C-terminal, indicating that this protein may be a novel member of the TNFR superfamily. Constitutive PmTNFR1 and PmTNFR5 mRNA expression was detected in all six pearl oyster tissues tested, with comparatively greater transcript abundance in the hepatopancreas and gill. The gene expression levels of PmTNFR1 and PmTNFR5, as well as those of downstream signaling molecules related to the NF-κB pathway (RIP, TRAF2, TRAF3, IKK, and NF-κB), were quantified in the gill after LPS challenge and in the hemocytes after nucleus insertion surgery using real-time PCR (qRT-PCR). We found that all genes were significantly upregulated at 6 h and 12 h post-injection, as well as at 15 d post-insertion. We used RNAi to inhibit the expression of the PmTNFR1 and PmTNFR5 genes. We then quantified the expression levels of PmTNFR1 and PmTNFR5, as well as downstream genes, using qRT-PCR. We found that RNAi inhibition of PmTNFR1 and PmTNFR5 downregulated the downstream genes (RIP, TRAF2, TRAF3, IKK, and NF-κB). Therefore, our results suggested that PmTNFR1 and PmTNFR5 mediate the NF-κB signaling pathway, and are closely related to immune defense, particularly allograft immunity, in the pearl oyster P. fucata martensii.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate/genetics , Pinctada/genetics , Pinctada/immunology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Amino Acid Sequence , Animals , Base Sequence , Gene Expression Profiling , Phylogeny , Receptors, Tumor Necrosis Factor/chemistry , Sequence Alignment
13.
Fish Shellfish Immunol ; 104: 470-477, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32585357

ABSTRACT

Tumor necrosis factor-α (TNF-α) is a pluripotent mediator of pro-inflammatory and antimicrobial defense mechanisms and a regulator of lymphoid organ development. Although two types of TNF-α have been identified in several teleost species, their functions in pathogen infection remain largely unexplored, especially in pathogen clearance. Herein, we cloned and characterized two types of TNF-α, termed shTNF-α1 and shTNF-α2, and their receptors, shTNFR1 and shTNFR2, from snakehead (Channa argus). These genes were constitutively expressed in all tested tissues, and were induced by Aeromonas schubertii and Nocardia seriolae in head kidney and spleen in vivo, and by lipoteichoic acid (LTA), lipopolysaccharides (LPS), and Polyinosinic-polycytidylic acid [Poly (I:C)] in head kidney leukocytes (HKLs) in vitro. Moreover, recombinant shTNF-α1 and shTNF-α2 upregulated the expression of endogenous shTNF-α1, shTNF-α2, shTNFR1, and shTNFR2, and enhanced intracellular bactericidal activity, with shTNF-α1 having a greater effect than shTNF-α2. These findings suggest important roles of fish TNFα1, TNFα2, and their receptors in bacterial infection and pathogen clearance, and provide a new insight into their function in antibacterial innate immunity.


Subject(s)
Fish Diseases/immunology , Fishes/genetics , Fishes/immunology , Immunity, Innate/genetics , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/genetics , Aeromonas/physiology , Animals , Fish Proteins/genetics , Fish Proteins/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary , Head Kidney/immunology , Leukocytes/immunology , Lipopolysaccharides/pharmacology , Nocardia/physiology , Nocardia Infections/immunology , Nocardia Infections/veterinary , Poly I-C/pharmacology , Receptors, Tumor Necrosis Factor/immunology , Spleen/immunology , Teichoic Acids/pharmacology , Tumor Necrosis Factor-alpha/immunology
14.
Int J Mol Sci ; 21(19)2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32992926

ABSTRACT

Inflammation and an influx of macrophages are common elements in many diseases. Among pro-inflammatory cytokines, tumor necrosis factor α (TNFα) plays a central role by amplifying the cytokine network. Progranulin (PGRN) is a growth factor that binds to TNF receptors and interferes with TNFα-mediated signaling. Extracellular PGRN is processed into granulins by proteases released from immune cells. PGRN exerts anti-inflammatory effects, whereas granulins are pro-inflammatory. The factors coordinating these ambivalent functions remain unclear. In our study, we identify Y-box binding protein-1 (YB-1) as a candidate for this immune-modulating activity. Using a yeast-2-hybrid assay with YB-1 protein as bait, clones encoding for progranulin were selected using stringent criteria for strong interaction. We demonstrate that at physiological concentrations, YB-1 interferes with the binding of TNFα to its receptors in a dose-dependent manner using a flow cytometry-based binding assay. We show that YB-1 in combination with progranulin interferes with TNFα-mediated signaling, supporting the functionality with an NF-κB luciferase reporter assay. Together, we show that YB-1 displays immunomodulating functions by affecting the binding of TNFα to its receptors and influencing TNFα-mediated signaling via its interaction with progranulin.


Subject(s)
Macrophages/immunology , Progranulins/immunology , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction/immunology , Transcription Factors/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Macrophages/pathology , Mice , Progranulins/genetics , RAW 264.7 Cells , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/genetics
15.
Trends Immunol ; 37(3): 170-180, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26830540

ABSTRACT

Inducing sustained, robust CD8(+) T cell responses is necessary for therapeutic intervention in chronic infectious diseases and cancer. Unfortunately, most adjuvant formulations fail to induce substantial cellular immunity in humans. Attenuated acute infectious agents induce strong CD8(+) T cell immunity, and are thought to therefore represent a good road map for guiding the development of subunit vaccines capable of inducing the same. However, recent evidence suggests that this assumption may need reconsideration. Here we provide an overview of subunit vaccine history as it pertains to instigating T cell responses. We argue that in light of evidence demonstrating that T cell responses to vaccination differ from those induced by infectious challenge, research in pursuit of cellular immunity-inducing vaccine adjuvants should no longer follow only the infection paradigm.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Immunotherapy, Adoptive , Infections/immunology , Vaccines, Subunit/immunology , Adjuvants, Immunologic , Animals , Humans , Lymphocyte Activation , Receptors, Tumor Necrosis Factor/immunology
16.
Fish Shellfish Immunol ; 93: 542-550, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31394160

ABSTRACT

In this study, an echinoderm tumor necrosis factor receptor named HLTNFR-16 was first cloned from the tropical sea cucumber Holothuria leucospilota. The full-length cDNA of HLTNFR-16 is 3675 bp in size, containing a 415 bp 5'-untranslated region (UTR), a 2024 bp 3'-UTR and a 1236 bp open reading frame (ORF) encoding a protein of 411 amino acids with a deduced molecular weight of 45.63 kDa. The HLTNFR-16 protein contains a signal peptide, four TNFR domains (the last three were identified as extracellular cysteine-rich domains), a transmembrane region and a death domain. Phylogenetic analysis showed that HLTNFR-16 was clustered into a clade with TNFR-16s in other species, indicating that this echinoderm TNFR may be a new member of the TNFR-16 subfamily. The results of TUNEL assay showed that the over expression of HLTNFR-16 could induce apoptosis in HEK293T cells. When HLTNFR-16 was silenced by siRNA, the apoptosis of sea cucumber coelomocytes induced by inactivated Vibrio harveyi was suppressed significantly, indicating that HLTNFR-16 is important for apoptosis induction. Additionally, luciferase reporter assay exhibited that the over-expressed HLTNFR-16 in HEK293T cells could activate the transcription factors nuclear factor-κB (NF-κB) and activator protein-1 (AP-1). Moreover, the secretion of proinflammatory cytokines interleukin (IL)-1ß, IL-6 and IL-18 in HEK293T cells was increased by the over-expression of HLTNFR-16. This study provides evidences for the potential roles of sea cucumber TNFR in the innate immunity.


Subject(s)
Gene Expression Regulation/immunology , Holothuria/genetics , Holothuria/immunology , Immunity, Innate/genetics , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Amino Acid Sequence , Animals , Base Sequence , Phylogeny , Receptors, Tumor Necrosis Factor/chemistry , Sequence Alignment , Vibrio/physiology
17.
Immunol Invest ; 48(5): 451-465, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30689461

ABSTRACT

Lipopolysaccharide (LPS) induces apoptosis in murine macrophages through the autocrine secretion of tumor necrosis factor (TNF)-α and nitric oxide (NO). LPS-induced inflammation in murine macrophages is associated with hydrogen sulfide (H2S) production. In this present study, we reported the novel role of H2S in LPS-induced apoptosis and its underlying molecular mechanism specifically at late phases in murine macrophage cells. Stimulation of RAW 264.7 macrophages with LPS resulted in a time- and dose-dependent induction of apoptosis. We observed that the LPS-induced early apoptosis (associated with TNF-α secretion) in macrophages was not inhibited in the presence of H2S inhibitor (DL-propargylglycine), whereas early apoptosis was absent in the presence of TNF receptor antibody. Interestingly, LPS-induced late apoptosis paralleled with H2S production was reduced in the presence of H2S inhibitor but not with TNF receptor antibody. The late apoptotic events mediated by H2S and not the TNF-α induced early apoptosis correlated significantly with the induction of p53 and Bax expression in LPS-induced macrophages. Thus, it is possible that RAW 264.7 murine macrophages treated with LPS mediated early apoptosis through TNF-α and the late apoptotic events through the production of H2S.


Subject(s)
Hydrogen Sulfide/metabolism , Inflammation/immunology , Macrophages/immunology , Alkynes/pharmacology , Animals , Antibodies, Blocking/pharmacology , Apoptosis , Cystathionine gamma-Lyase/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Lipopolysaccharides/immunology , Mice , RAW 264.7 Cells , Receptors, Tumor Necrosis Factor/immunology , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
18.
Cytokine ; 101: 4-13, 2018 01.
Article in English | MEDLINE | ID: mdl-27639923

ABSTRACT

TNFα is a principal pro-inflammatory cytokine vital for immunity to infections. However, its excessive production is involved in chronic inflammation and disease pathology in autoimmune diseases. Evidence for its pathogenic role is validated by the fact that its neutralisation by therapeutic agents in vivo is beneficial in ameliorating disease and controlling symptoms. Paradoxically, however, treatment with TNFα inhibitors can either have no clinical effects, or even exacerbate disease in some patients. The explanation for such contradictory outcomes may lay in how and which downstream signalling pathways are activated and drive disease. TNFα causes its effects by binding to either or both of two membrane-bound receptors, TNFR1 and TNFR2. Engagement of the receptors can induce cell death or cell proliferation. T cells both produce and respond to TNFα and depending on whether the cytokine is membrane-bound or soluble and the level of expression of its two receptors, the biological outcome can be distinct. In addition, polymorphisms in genes encoding TNFα and T cell signalling proteins can significantly impact the outcome of TNFα receptor engagement. Early studies revealed that effector T cells in patients with rheumatoid arthritis (RA) are hyporesponsive due to chronic exposure to TNFα. However, recent evidence indicates that the relationship between TNFα and T cell responses is complex and, at times, can be paradoxical. In addition, there is controversy as to the specific effects of TNFα on different T cell subsets. This review will summarise knowledge on how TNFα modulates T cell responses and the effect of engaging either of its two receptors. Furthermore, we discuss how such interactions can dictate the outcome of treatment with TNFα inhibitors.


Subject(s)
Arthritis, Rheumatoid/immunology , Autoimmune Diseases/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Tumor Necrosis Factor-alpha/genetics , Animals , Antirheumatic Agents/therapeutic use , Autoimmune Diseases/drug therapy , Autoimmune Diseases/physiopathology , Gene Expression Regulation/immunology , Humans , Inflammation/immunology , Inflammation/physiopathology , Mice , Polymorphism, Genetic , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
19.
Cytokine ; 101: 48-55, 2018 01.
Article in English | MEDLINE | ID: mdl-27527809

ABSTRACT

Autoimmune disease encompasses an array of conditions with a variety of presentations and the involvement of multiple organs. Though the etiologies of many autoimmune conditions are unclear, uncontrolled inflammatory immune response is believed to be a major cause of disease development and progression. Progranulin (PGRN), an anti-inflammatory molecule with therapeutic effect in inflammatory arthritis, was identified as an endogenous antagonist of TNFα by competitively binding to TNFR. PGRN exerts its anti-inflammatory activity through multiple pathways, including induction of Treg differentiation and IL-10 expression and inhibition of chemokine release from macrophages. In addition, the protective role of PGRN has also been demonstrated in osteoarthritis, inflammatory bowel disease, and psoriasis. Intriguingly, PGRN was reported to contribute to development of insulin resistance in high-fat diet induced diabetes. Emerging evidences indicate that PGRN may also be associated with various autoimmune diseases, including systemic lupus erythematous, systemic sclerosis, multiple sclerosis and Sjogren's syndrome. This review summarizes recent studies of PGRN as a novel target molecule in the field of autoimmune disease, and provides updated information to inspire future studies.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/physiopathology , Intercellular Signaling Peptides and Proteins/immunology , Animals , Arthritis/immunology , Arthritis/physiopathology , Arthritis/therapy , Autoimmune Diseases/complications , Autoimmune Diseases/therapy , Humans , Inflammation/immunology , Inflammation/therapy , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/physiopathology , Inflammatory Bowel Diseases/therapy , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/therapeutic use , Interleukin-10/genetics , Interleukin-10/immunology , Macrophages/immunology , Mice , Osteoarthritis/immunology , Osteoarthritis/physiopathology , Osteoarthritis/therapy , Progranulins , Protein Binding , Psoriasis/immunology , Psoriasis/physiopathology , Psoriasis/therapy , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
20.
Trends Immunol ; 36(11): 697-708, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26481667

ABSTRACT

The control of persistent viral infections requires the immune system to limit the spread of the virus while avoiding immunopathology. Recent studies have revealed that members of the tumor necrosis factor receptor (TNFR) superfamily play unique and pivotal roles in control of chronic lymphocytic choriomeningitis virus (LCMV) infection and in some settings can tip the balance between immune control and immune pathology. We review these findings and discuss how our understanding of the role of TNFRs in the immune response to chronic LCMV infection may shed light on what happens during HIV infection in humans. We discuss preclinical models of TNF/TNFR family-targeted immunotherapy of chronic LCMV infection and evaluate which TNFRs present the most promising targets for immune intervention.


Subject(s)
Arenaviridae Infections/immunology , Arenaviridae Infections/therapy , Lymphocytic choriomeningitis virus/immunology , Receptors, Tumor Necrosis Factor/immunology , Animals , Arenaviridae Infections/virology , Chronic Disease , Humans , Immunotherapy , Lymphocytic choriomeningitis virus/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL