Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Cell Death Dis ; 13(4): 297, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35379781

ABSTRACT

Hepatic stem/progenitor cells are the major cell compartment for tissue repair when hepatocyte proliferation is compromised in chronic liver diseases, but the expansion of these cells increases the risk of carcinogenesis. Therefore, it is essential to explore the pathways restricting their expansion and abnormal transformation. The ligand of glucocorticoid-induced tumour necrosis factor receptor (GITRL) showed the most highly increased expression in hepatic progenitor cells treated with transforming growth factor (TGF)-ß1. If overexpressed by hepatic progenitor cells, GITRL stimulated cell proliferation by activating the epithelial-mesenchymal transition pathway and enhancing ERK1/2 and Akt phosphorylation via GITRL binding to ANXA2. However, GITR, the specific GITRL receptor, suppressed the epithelial-mesenchymal transition pathway of GITRL-expressing cells and decreased their growth by dissociating ANXA2 from GITRL and reducing downstream ERK1/2 and Akt phosphorylation. This study identifies GITR/GITRL reverse signalling as a cross-interaction pathway between immune cells and hepatic stem/progenitor cells that restricts the expansion of hepatic stem/progenitor cells and reduces the possibility of carcinogenesis.


Subject(s)
Annexin A2 , Tumor Necrosis Factors , Annexin A2/metabolism , Carcinogenesis , Cell Proliferation , Glucocorticoid-Induced TNFR-Related Protein/genetics , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Humans , MAP Kinase Signaling System , Proto-Oncogene Proteins c-akt/metabolism , Stem Cells/metabolism , Tumor Necrosis Factors/metabolism
2.
J Immunother ; 45(4): 194-206, 2022 05 01.
Article in English | MEDLINE | ID: mdl-34908008

ABSTRACT

The tumor necrosis factor receptor superfamily members 4 (TNFRSF4, OX40) and 18 (TNFRSF18, GITR, AITR) are under investigation as targets for immunotherapy of various cancers, including head and neck squamous cell carcinomas. Understanding the regulation of OX40 and GITR, particularly on an epigenetic level, might help to develop companion predictive biomarkers. We conducted broad correlation analyses of DNA methylation of 46 CpG sites within the GITR/OX40 gene locus in head and neck squamous cell carcinomas and normal adjacent tissues provided by The Cancer Genome Atlas (TCGA) Research Network. We analyzed methylation levels with regard to transcriptional gene activity (mRNA expression), human papillomavirus (HPV) infection, differential methylation between tumors and normal adjacent tissues, signatures of immune cell infiltrates, an interferon-γ signature, mutational load, and overall survival. Moreover, we investigated methylation levels in HPV-positive and HPV-negative cell lines and in isolated monocytes, granulocytes, CD8+ and CD4+ T cells, and B cells from peripheral blood from healthy donors. Our results revealed a complex and sequence-contextual methylation pattern in accordance with features of epigenetic regulated genes. We detected significant methylation differences between normal adjacent and tumor tissues, between HPV-positive and HPV-negative tumors, between tumor and immune cells, and significant correlations between methylation and mRNA expression. We further found significant correlations of CpG methylation with overall survival, signatures of immune cell infiltrates, an interferon-γ signature, and mutational load. Our study provides a framework to prospectively test specific CpG sites as biomarkers, in particular in the context of immunotherapies.


Subject(s)
Head and Neck Neoplasms , Papillomavirus Infections , DNA Methylation , Glucocorticoid-Induced TNFR-Related Protein/genetics , Head and Neck Neoplasms/genetics , Humans , Interferon-gamma , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Prognosis , RNA, Messenger/genetics , Receptors, OX40/genetics , Squamous Cell Carcinoma of Head and Neck/genetics
3.
Cell Rep ; 36(12): 109734, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34551288

ABSTRACT

Glucocorticoid-induced tumor necrosis factor receptor family-related protein (GITR) is a critical regulatory molecule in modulation of T cell immune responses. Here we report the mouse GITR (mGITR) and mGITR ligand (mGITRL) complex structure and find that the binding interface of mGITR and mGITRL is distinct from the typical tumor necrosis factor superfamily (TNFSF)/TNF receptor superfamily (TNFRSF) members. mGITR binds to its ligand with a single domain, whereas the binding interface on mGITRL is located on the side, which is distal from conserved binding sites of TNFSF molecules. Mutational analysis reveals that the binding interface of GITR/GITRL in humans is conserved with that in the mouse. Substitution of key interacting D93-I94-V95 (DIV) in mGITR with the corresponding K93-F94-S95 (KFS) in human GITR enables cross-recognition with human GITRL and cross-activation of receptor signaling. The findings of this study substantially expand our understanding of the interaction of TNFSF/TNFRSF superfamily molecules and can benefit the future design of biologics by targeting GITR/GITRL.


Subject(s)
Glucocorticoid-Induced TNFR-Related Protein/metabolism , T-Lymphocytes/metabolism , Tumor Necrosis Factors/metabolism , Amino Acid Sequence , Animals , Cell Line , Crystallography, X-Ray , Glucocorticoid-Induced TNFR-Related Protein/chemistry , Glucocorticoid-Induced TNFR-Related Protein/genetics , Humans , Lymphocyte Activation , Mice , Mutagenesis , Protein Binding , Protein Structure, Quaternary , Sequence Alignment , Surface Plasmon Resonance , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tumor Necrosis Factors/chemistry , Tumor Necrosis Factors/genetics
4.
Int J Mol Sci ; 22(13)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34281195

ABSTRACT

Regulatory T cells (Tregs) exert a highly suppressive function in the immune system. Disturbances in their function predispose an individual to autoimmune dysregulation, with a predominance of the pro-inflammatory environment. Besides Foxp3, which is a master regulator of these cells, other genes (e.g., Il2ra, Ctla4, Tnfrsf18, Ikzf2, and Ikzf4) are also involved in Tregs development and function. Multidimensional Tregs suppression is determined by factors that are believed to be crucial in the action of Tregs-related genes. Among them, epigenetic changes, such as DNA methylation, tend to be widely studied over the past few years. DNA methylation acts as a repressive mark, leading to diminished gene expression. Given the role of increased CpG methylation upon Tregs imprinting and functional stability, alterations in the methylation pattern can cause an imbalance in the immune response. Due to the fact that epigenetic changes can be reversible, so-called epigenetic modifiers are broadly used in order to improve Tregs performance. In this review, we place emphasis on the role of DNA methylation of the genes that are key regulators of Tregs function. We also discuss disease settings that have an impact on the methylation status of Tregs and systematize the usefulness of epigenetic drugs as factors able to influence Tregs functions.


Subject(s)
DNA Methylation , T-Lymphocytes, Regulatory/physiology , CTLA-4 Antigen/genetics , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Gene Expression , Gene Expression Regulation , Glucocorticoid-Induced TNFR-Related Protein/genetics , Humans , Ikaros Transcription Factor/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
5.
Commun Biol ; 4(1): 914, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34312483

ABSTRACT

Malignant pleural mesothelioma (MPM) is an aggressive neoplasm originating from the pleura. Non-epithelioid (biphasic and sarcomatoid) MPM are particularly resistant to therapy. We investigated the role of the GITR-GITRL pathway in mediating the resistance to therapy. We found that GITR and GITRL expressions were higher in the sarcomatoid cell line (CRL5946) than in non-sarcomatoid cell lines (CRL5915 and CRL5820), and that cisplatin and Cs-137 irradiation increased GITR and GITRL expressions on tumor cells. Transcriptome analysis demonstrated that the GITR-GITRL pathway was promoting tumor growth and inhibiting cell apoptosis. Furthermore, GITR+ and GITRL+ cells demonstrated increased spheroid formation in vitro and in vivo. Using patient derived xenografts (PDXs), we demonstrated that anti-GITR neutralizing antibodies attenuated tumor growth in sarcomatoid PDX mice. Tumor immunostaining demonstrated higher levels of GITR and GITRL expressions in non-epithelioid compared to epithelioid tumors. Among 73 patients uniformly treated with accelerated radiation therapy followed by surgery, the intensity of GITR expression after radiation negatively correlated with survival in non-epithelioid MPM patients. In conclusion, the GITR-GITRL pathway is an important mechanism of autocrine proliferation in sarcomatoid mesothelioma, associated with tumor stemness and resistance to therapy. Blocking the GITR-GITRL pathway could be a new therapeutic target for non-epithelioid mesothelioma.


Subject(s)
Antineoplastic Agents/pharmacology , Cesium Radioisotopes/pharmacology , Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic , Glucocorticoid-Induced TNFR-Related Protein/genetics , Mesothelioma, Malignant/genetics , Tumor Necrosis Factors/genetics , Animals , Cell Line, Tumor , Female , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Humans , Mesothelioma, Malignant/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Necrosis Factors/metabolism
6.
Vascul Pharmacol ; 139: 106884, 2021 08.
Article in English | MEDLINE | ID: mdl-34102305

ABSTRACT

Atherosclerosis is a lipid-driven, chronic inflammatory disease. In spite of efficient lipid lowering treatments, such as statins and PCSK9 inhibitors, patients, especially those with elevated inflammatory biomarkers, still have a significant residual cardiovascular disease risk. Novel drugs targeting inflammatory mediators are needed to further reduce this residual risk. Agonistic immune checkpoint proteins, including CD86, CD40L and CD40, have been shown to be drivers of atherosclerosis. Recently, glucocorticoid-induced tumour necrosis factor receptor family-related protein (GITR), a co-stimulatory immune checkpoint protein, was identified to be pivotal in cardiovascular disease. Cardiovascular patients have elevated soluble GITR plasma levels compared to healthy controls. Furthermore, in human carotid endarterectomy plaques, GITR expression was higher in plaques from symptomatic compared to asymptomatic patients and correlated with features of plaque vulnerability. Moreover, depleting GITR reduced atherosclerotic plaque development in mice. GITR-deficient monocytes and macrophages exhibited less inflammatory potential and reduced migratory capacity. In this review, we discuss GITR's effects on various immune cells, mechanisms, signalling pathways and finally GITR's potential as a novel drug target in atherosclerosis.


Subject(s)
Atherosclerosis , Glucocorticoid-Induced TNFR-Related Protein/genetics , Proprotein Convertase 9 , Animals , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Glucocorticoids/pharmacology , Humans , Mice
7.
Nat Commun ; 12(1): 2710, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976194

ABSTRACT

Treg cells are critical regulators of immune homeostasis, and environment-driven Treg cell differentiation into effector (e)Treg cells is crucial for optimal functioning. However, human Treg cell programming in inflammation is unclear. Here, we combine transcriptional and epigenetic profiling to identify a human eTreg cell signature. Inflammation-derived functional Treg cells have a transcriptional profile characterized by upregulation of both a core Treg cell (FOXP3, CTLA4, TIGIT) and effector program (GITR, BLIMP-1, BATF). We identify a specific human eTreg cell signature that includes the vitamin D receptor (VDR) as a predicted regulator in eTreg cell differentiation. H3K27ac/H3K4me1 occupancy indicates an altered (super-)enhancer landscape, including enrichment of the VDR and BATF binding motifs. The Treg cell profile has striking overlap with tumor-infiltrating Treg cells. Our data demonstrate that human inflammation-derived Treg cells acquire a conserved and specific eTreg cell profile guided by epigenetic changes, and fine-tuned by environment-specific adaptations.


Subject(s)
Arthritis, Juvenile/genetics , Epigenesis, Genetic , Receptors, Calcitriol/genetics , T-Lymphocytes, Regulatory/immunology , Transcriptome , Adolescent , Arthritis, Juvenile/immunology , Arthritis, Juvenile/pathology , Base Sequence , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Case-Control Studies , Cell Differentiation , Child , Child, Preschool , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Profiling , Gene Regulatory Networks , Glucocorticoid-Induced TNFR-Related Protein/genetics , Glucocorticoid-Induced TNFR-Related Protein/immunology , Histones/genetics , Histones/immunology , Humans , Joints/immunology , Joints/pathology , Male , Metabolic Networks and Pathways/genetics , Metabolic Networks and Pathways/immunology , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , Primary Cell Culture , Receptors, Calcitriol/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , T-Lymphocytes, Regulatory/pathology , Young Adult
8.
Cancer Immunol Immunother ; 70(9): 2483-2496, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33538861

ABSTRACT

Owing to their key role in several diseases including cancer, activating and inhibitory immune checkpoint molecules are increasingly exploited as targets for immunotherapy. Recently, we demonstrated that platelets, which largely influence tumor progression and immune evasion, functionally express the ligand of the checkpoint molecule GITR. This immunoreceptor modulates effector functions of T cells and NK cells with its function varying dependent on cellular context and activation state. Here, we provide a comparative analysis of platelet-derived GITRL (pGITRL) in breast cancer patients and healthy volunteers. The levels of pGITRL were found to be higher on platelets derived from cancer patients and appeared to be specifically regulated during tumor progression as exemplified by several clinical parameters including tumor stage/grade, the occurrence of metastases and tumor proliferation (Ki67) index. In addition, we report that pGITRL is upregulated during platelet maturation and particularly induced upon exposure to tumor-derived soluble factors. Our data indicate that platelets modulate the GITR/GITRL immune checkpoint in the context of malignant disease and provide a rationale to further study the GITR/GITRL axis for exploitation for immunotherapeutic intervention in cancer patients.


Subject(s)
Blood Platelets/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Immune Checkpoint Proteins/genetics , Tumor Necrosis Factors/genetics , Biomarkers, Tumor , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Flow Cytometry , Glucocorticoid-Induced TNFR-Related Protein/genetics , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Humans , Immune Checkpoint Proteins/metabolism , Immunophenotyping , Lymphocytes/immunology , Lymphocytes/metabolism , Odds Ratio , Platelet Activation , Platelet Aggregation , Tumor Necrosis Factors/metabolism
9.
Nat Commun ; 12(1): 832, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547304

ABSTRACT

The two T cell inhibitory receptors PD-1 and TIM-3 are co-expressed during exhausted T cell differentiation, and recent evidence suggests that their crosstalk regulates T cell exhaustion and immunotherapy efficacy; however, the molecular mechanism is unclear. Here we show that PD-1 contributes to the persistence of PD-1+TIM-3+ T cells by binding to the TIM-3 ligand galectin-9 (Gal-9) and attenuates Gal-9/TIM-3-induced cell death. Anti-Gal-9 therapy selectively expands intratumoral TIM-3+ cytotoxic CD8 T cells and immunosuppressive regulatory T cells (Treg cells). The combination of anti-Gal-9 and an agonistic antibody to the co-stimulatory receptor GITR (glucocorticoid-induced tumor necrosis factor receptor-related protein) that depletes Treg cells induces synergistic antitumor activity. Gal-9 expression and secretion are promoted by interferon ß and γ, and high Gal-9 expression correlates with poor prognosis in multiple human cancers. Our work uncovers a function for PD-1 in exhausted T cell survival and suggests Gal-9 as a promising target for immunotherapy.


Subject(s)
Adenocarcinoma/therapy , Colonic Neoplasms/therapy , Galectins/immunology , Gene Expression Regulation, Neoplastic/immunology , Glucocorticoid-Induced TNFR-Related Protein/immunology , Hepatitis A Virus Cellular Receptor 2/immunology , Programmed Cell Death 1 Receptor/immunology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/mortality , Animals , Antibodies/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/mortality , Galectins/antagonists & inhibitors , Galectins/genetics , Glucocorticoid-Induced TNFR-Related Protein/agonists , Glucocorticoid-Induced TNFR-Related Protein/genetics , Hepatitis A Virus Cellular Receptor 2/genetics , Humans , Immunotherapy/methods , Jurkat Cells , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/mortality , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Programmed Cell Death 1 Receptor/genetics , Protein Binding , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Skin Neoplasms/therapy , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
10.
Front Immunol ; 12: 809208, 2021.
Article in English | MEDLINE | ID: mdl-34987524

ABSTRACT

Rationale: Inflammatory monocyte (MC) subset differentiation is a major feature in tissue inflammatory and atherosclerosis. The underlying molecular mechanism remains unclear. Objective: This study aims to explore molecule targets and signaling which determinate immunological features in MC subsets. Methods and Results: Blood Ly6Chigh and Ly6Clow MC subsets from control and ApoE-/- mice were isolated by flow cytometry sorting and subjected for bulk high-throughput RNA-sequencing. Intensive bioinformatic studies were performed by analyzing transcriptome through four pairs of comparisons: A) Ly6Chigh vs Ly6Clow in control mice; B) Ly6Chigh vs Ly6Clow in ApoE-/- mice; C) ApoE-/- Ly6Chigh vs control Ly6Chigh MC; D) ApoE-/- Ly6Clow vs control Ly6Clow MC. A total of 80 canonical pathways and 16 enriched pathways were recognized by top-down analysis using IPA and GSEA software, and further used for overlapping analysis. Immunological features and signaling were assessed on four selected functional groups, including MHCII, immune checkpoint, cytokine, and transcription factor (TF). Among the total 14578 significantly differentially expressed (SDE) genes identified though above four comparison, 1051 TF and 348 immunological genes were discovered. SDE immunological genes were matched with corresponding upstream SDE TF by IPA upstream analysis. Fourteen potential transcriptional axes were recognized to modulate immunological features in the Ly6C MC subset. Based on an intensive literature search, we found that the identified SDE immune checkpoint genes in Ly6Chigh MC are associated with pro-inflammatory/atherogenic balance function. Immune checkpoint genes GITR, CTLA4, and CD96 were upregulated in Ly6Clow MC from all mice and presented anti-inflammatory/atherogenic features. Six cytokine genes, including Ccl2, Tnfsf14, Il1rn, Cxcl10, Ccl9, and Cxcl2, were upregulated in Ly6Chigh MC from all mice and associated with pro-inflammatory/atherogenic feature. Cytokine receptor gene Il12rb2, Il1r1, Il27ra, Il5ra, Ngfr, Ccr7, and Cxcr5 were upregulated in Ly6Clow MC from all mice and presented anti-inflammatory/atherogenic features. MHCII genes (H2-Oa, H2-DMb2, H2-Ob, H2-Eb2, H2-Eb1, H2-Aa, and Cd74) were elevated in Ly6Clow MC from all mice. ApoE-/- augmented pro-atherogenic/inflammatory and antigen-presenting cells (APC) feature in both subsets due to elevated expression of cytokine genes (Cxcl11, Cntf, Il24, Xcl, Ccr5, Mpl, and Acvr2a) and MHCII gene (H2-Aa and H2-Ea-ps). Finally, we modeled immunological gene expression changes and functional implications in MC differentiation and adaptive immune response for MC subsets from control and ApoE-/- mice. Conclusions: Ly6Chigh MC presented pro-inflammatory/atherogenic features and lower APC potential. Ly6Clow MC displayed anti-inflammatory/atherogenic features and higher APC potential. ApoE-/- confers upon both subsets with augmented pro-atherogenic/inflammatory function and APC potential.


Subject(s)
Apolipoproteins E/metabolism , Atherosclerosis/immunology , Inflammation/immunology , Macrophages/immunology , Monocytes/immunology , Adaptive Immunity , Animals , Antigens, Ly/metabolism , Apolipoproteins E/genetics , Cell Differentiation , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation , Glucocorticoid-Induced TNFR-Related Protein/genetics , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Histocompatibility Antigens/genetics , Humans , Mice , Mice, Knockout , Signal Transduction
11.
Mol Ther ; 29(3): 1294-1311, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33279722

ABSTRACT

Tissue-resident macrophages (TRMs) are sentinel cells for maintaining tissue homeostasis and organ function. In this study, we discovered that lipopolysaccharide (LPS) administration dramatically reduced TRM populations and suppressed their self-renewal capacities in multiple organs. Using loss- and gain-of-function approaches, we define Sectm1a as a novel regulator of TRM self-renewal. Specifically, at the earlier stage of endotoxemia, Sectm1a deficiency exaggerated acute inflammation-induced reduction of TRM numbers in multiple organs by suppressing their proliferation, which was associated with more infiltrations of inflammatory monocytes/neutrophils and more serious organ damage. By contrast, administration of recombinant Sectm1a enhanced TRM populations and improved animal survival upon endotoxin challenge. Mechanistically, we identified that Sectm1a-induced upregulation in the self-renewal capacity of TRM is dependent on GITR-activated T helper cell expansion and cytokine production. Meanwhile, we found that TRMs may play an important role in protecting local vascular integrity during endotoxemia. Our study demonstrates that Sectm1a contributes to stabling TRM populations through maintaining their self-renewal capacities, which benefits the host immune response to acute inflammation. Therefore, Sectm1a may serve as a new therapeutic agent for the treatment of inflammatory diseases.


Subject(s)
Glucocorticoid-Induced TNFR-Related Protein/metabolism , Immunologic Memory/immunology , Inflammation/complications , Macrophages/immunology , Membrane Proteins/metabolism , Monocytes/immunology , Multiple Organ Failure/prevention & control , Animals , Glucocorticoid-Induced TNFR-Related Protein/genetics , Homeostasis , Membrane Proteins/genetics , Mice , Multiple Organ Failure/etiology , T-Lymphocytes, Helper-Inducer/immunology
12.
Front Immunol ; 11: 2150, 2020.
Article in English | MEDLINE | ID: mdl-33072079

ABSTRACT

The mechanism(s) underlying endotoxin tolerance in asthma remain elusive. As the endotoxin lipopolysaccharide (LPS) affects the expression of the regulatory T-cell (Treg)-suppressive glucocorticoid-induced tumor necrosis factor receptor ligand (GITRL) on antigen-presenting dendritic cells (DCs), we hypothesized that LPS-induced changes in DC GITRL expression may impact Treg-mediated T-helper (Th) cell suppression and the induction of endotoxin tolerance. Here, we propose a novel mechanism by which low-dose LPS inhalation in neonatal mice induces endotoxin tolerance, thereby offering protection from later asthma development. Three-day old wild-type and Toll-like receptor 4 (TLR4)-deficient neonatal mice were exposed to low-dose LPS (1 µg) intranasally for 10 consecutive days prior to ovalbumin (OVA)-induced asthma to better understand the tolerogenic mechanism(s) of low-dose LPS pre-exposure. In vivo findings were validated using in vitro co-culturing studies of primary CD11c+ DCs and CD4+ T-cells with or without low-dose LPS pre-exposure before OVA stimulation. Low-dose LPS pre-exposure upregulated the Treg response and downregulated pathogenic Th2 and Th17 responses through promoting apoptosis of Th2 and Th17 cells. Low-dose LPS pre-exposure downregulated DC GITRL expression and T-cell GITR expression. Artificial DC GITRL expression abrogated the tolerogenic Treg-skewing effect of low-dose LPS pre-exposure. Low-dose LPS pre-exposure inhibited TRIF/IRF3/IFNß signaling and upregulated expression of tolerogenic TRIF/IRF3/IFNß negative regulators in a TLR4-dependent manner. This tolerogenic DC GITRL downregulation was attributable to TRIF/IRF3/IFNß signaling inhibition. Low-dose LPS pre-exposure produces tolerogenic Treg skewing in neonatal asthmatic mice, a phenomenon attributable to TLR4-dependent TRIF/IRF3/IFNß-mediated DC GITRL downregulation.


Subject(s)
Asthma/immunology , Immune Tolerance/drug effects , Lipopolysaccharides/toxicity , T-Lymphocytes, Regulatory/drug effects , Animals , Apoptosis/drug effects , Asthma/etiology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Disease Models, Animal , Dose-Response Relationship, Immunologic , Gene Expression Regulation/drug effects , Glucocorticoid-Induced TNFR-Related Protein/biosynthesis , Glucocorticoid-Induced TNFR-Related Protein/genetics , Humans , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/pharmacology , Lymphocyte Count , Mice , Mice, Inbred BALB C , Ovalbumin , Signal Transduction/drug effects , Specific Pathogen-Free Organisms , Th17 Cells/drug effects , Th17 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Toll-Like Receptor 4/deficiency , Tumor Necrosis Factors/biosynthesis , Tumor Necrosis Factors/genetics
13.
FASEB J ; 34(11): 14820-14831, 2020 11.
Article in English | MEDLINE | ID: mdl-32910505

ABSTRACT

Glucocorticoid-induced TNFR family related gene (GITR) is a member of the TNFR superfamily that is expressed on cells of the immune system. Although the protective and pathogenic roles of GITR in T cell immunity are well characterized, the role of GITR in innate immunity in the intestinal tissues has not been well clarified. In this study, using a dextran sulfate sodium (DSS)-induced colitis model in mice, we found that GITR-deficiency rendered mice more susceptible to acute intestinal inflammation and that a significantly higher number of activated natural killer (NK) cells was accumulated in the colonic lamina propria of Gitr-/- mice as compared to wild-type mice. Additionally, Rag2-/- Gitr-/- mice, which lack T cells but have NK cells, also displayed more severe colonic inflammation than Rag2-/- mice. In contrast, an anti-GITR agonistic antibody significantly alleviated colitis in Rag2-/- mice. Engagement of GITR inhibited IL-15-mediated activating signaling events in NK cells, which include cell activation and proliferation, and production of cytokines and cytotoxic granules. Taken together, our results provide the first evidence that GITR negatively controls intestinal inflammation through NK cell functions.


Subject(s)
Colitis, Ulcerative/immunology , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Intestinal Mucosa/immunology , Killer Cells, Natural/immunology , Animals , Cells, Cultured , Colitis, Ulcerative/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Glucocorticoid-Induced TNFR-Related Protein/genetics , Interleukin-15/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL
14.
J Immunol ; 205(6): 1633-1643, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32769121

ABSTRACT

The inability to effectively control invading bacteria or other pathogens is a major cause of multiple organ dysfunction and death in sepsis. As the first-line defense of the immune system, macrophages play a crucial role in the removal of pathogens during sepsis. In this study, we define secreted and transmembrane 1A (Sectm1a) as a novel ligand of glucocorticoid-induced TNFR (GITR) that greatly boosts macrophage phagocytosis and bactericidal capacity. Using a global Sectm1a knockout (KO) mouse model, we observed that Sectm1a deficiency significantly suppressed phagocytosis and bactericidal activity in both recruited macrophages and tissue-resident macrophages, which consequently aggravated bacterial burden in the blood and multiple organs and further increased systemic inflammation, leading to multiple organ injury and increased mortality during polymicrobial sepsis. By contrast, treatment of septic mice with recombinant Sectm1a protein (rSectm1a) not only promoted macrophage phagocytosis and bactericidal activity but also significantly improved survival outcome. Mechanistically, we identified that Sectm1a could bind to GITR in the surface of macrophages and thereby activate its downstream PI3K-Akt pathway. Accordingly, rSectm1a-mediated phagocytosis and bacterial killing were abolished in macrophages by either KO of GITR or pharmacological inhibition of the PI3K-Akt pathway. In addition, rSectm1a-induced therapeutic effects on sepsis injury were negated in GITR KO mice. Taken together, these results uncover that Sectm1a may represent a novel target for drug development to control bacterial dissemination during sepsis or other infectious diseases.


Subject(s)
Glucocorticoid-Induced TNFR-Related Protein/metabolism , Macrophages/physiology , Membrane Proteins/metabolism , Multiple Organ Failure/immunology , Sepsis/immunology , Animals , Glucocorticoid-Induced TNFR-Related Protein/genetics , Humans , Immune Tolerance , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncogene Protein v-akt/metabolism , Phagocytosis , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
15.
Iran Biomed J ; 24(5): 295-305, 2020 09.
Article in English | MEDLINE | ID: mdl-32429643

ABSTRACT

Background: Vitamin D insufficiency and deficiency can be associated with adverse effects on fetus and pregnancy outcomes. This study aimed at evaluating the effect of 1,25VitD3 on specific transcription factor and markers of Tregs and T helper 17 (Th17) cells in peripheral blood mononuclear cells (PBMCs) of women with unexplained recurrent pregnancy loss (URPL) as a case group and PBMCs of healthy women as a control group. Methods: Samples from 20 non-pregnant patients with a history of URPL were compared to 20 normal non-pregnant women. PBMCs were divided into three wells for each subject in the presence of 1,25VitD3 (50 nM, for 16 hours), phytohemagglutinin (10 µM; positive control), and without any treatment (negative control). By Real-time PCR (Taqman assay), specific transcription factors of Tregs and Th17 cells, forkhead box P3 (FOXP3), retinoic acid-related orphan receptor γt (ROR-γt), glucocorticoid-induced tumor necrosis factor receptor-related (GITR), and CTLA-4 mRNA expressions in two groups were measured. Results: FOXP3/ROR-γt mRNA expression in PBMCs decreased significantly in women experiencing URPL compared to the control group (p = 0.0001). Although 1,25VitD3 (50 nM) increased FOXP3 gene expression (p = 0.0001), it did not significantly affect ROR-γt gene expression. Besides, 1,25VitD3 treatment significantly increased FOXP3/ROR-γt mRNA expression from baseline in PBMCs of the fetal loss group compared to that of the control group (p = 0.01). The 1,25VitD3 also increased GITR gene expression (p = 0.017) in PBMCs of URPL women compared to the controls. Conclusion: Vitamin D deficiency may be a contributor to recurrent pregnancy loss and suggests that the supplementation of women with Vitamin D pre-pregnancy may be protective against URPL via affecting Tregs signature genes, FOXP3 and GITR.


Subject(s)
Abortion, Habitual/genetics , CTLA-4 Antigen/genetics , Calcitriol/pharmacology , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Glucocorticoid-Induced TNFR-Related Protein/genetics , Leukocytes, Mononuclear/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Abortion, Habitual/blood , Abortion, Habitual/immunology , Biomarkers/blood , CTLA-4 Antigen/metabolism , Case-Control Studies , Female , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Gonadal Steroid Hormones/blood , Humans , Leukocytes, Mononuclear/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Pregnancy , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Young Adult
16.
Immunol Lett ; 222: 73-79, 2020 06.
Article in English | MEDLINE | ID: mdl-32259529

ABSTRACT

Follicular helper CD4+ T-cells (Tfh) control humoral immunity by driving affinity maturation and isotype-switching of activated B-cells. Tfh localize within B-cell follicles and, upon encounter with cognate antigen, drive B-cell selection in germinal centers (GCs) as GC-Tfh. Tfh functionality is controlled by Foxp3-expressing Tfh, which are known as regulatory T follicular cells (Tfr). Thus far, it remains unclear which factors determine the balance between these functionally opposing follicular T-cell subsets. Here, we demonstrate in human and mouse that Tfh and GC-Tfh, as well as their regulatory counterparts, express glucocorticoid-induced TNF receptor related protein (GITR) on their surface. This costimulatory molecule not only helps to identify follicular T-cell subsets, but also increases the ratio of Tfh vs. Tfr, both within and outside the GC. Correspondingly, GITR triggering increases the number of IL-21 producing CD4+ T-cells, which also produce more IFN-γ and IL-10. The latter are known switch factors for IgG2c and IgG1, respectively, which corresponds to a concomitant increase in IgG2c and IgG1 production upon GITR-mediated costimulation. These results demonstrate that GITR can skew the functional balance between Tfh and Tfr, which offers new therapeutic possibilities in steering humoral immunity.


Subject(s)
Glucocorticoid-Induced TNFR-Related Protein/genetics , Immunity, Humoral , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Cytokines/metabolism , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation , Germinal Center/immunology , Germinal Center/metabolism , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Humans , Immunoglobulin Class Switching/genetics , Male , Mice
17.
J Leukoc Biol ; 107(6): 953-970, 2020 06.
Article in English | MEDLINE | ID: mdl-32125017

ABSTRACT

Tissue resident memory T cells (Trm) are critical for local protection against reinfection. The accumulation of T cells in the tissues requires a post-priming signal from TNFR superfamily members, referred to as signal 4. Glucocorticoid-induced TNFR-related protein (GITR; TNFRSF18) signaling is important for this post-priming signal and for Trm formation during respiratory infection with influenza virus. As GITR signaling impacts both effector T cell accumulation and Trm formation, we asked if GITR differentially affects subsets of effector cells with different memory potential. Effector CD4+ T cells can be subdivided into 2 populations based on expression of lymphocyte antigen 6C (Ly6C), whereas effector CD8+ cells can be divided into 3 populations based on Ly6C and CX3CR1. The Ly6Chi and CX3CR1hi T cell populations represent the most differentiated effector T cells. Upon transfer, the Ly6Clo CD4+ effector T cells preferentially enter the lung parenchyma, compared to the Ly6Chi CD4+ T cells. We show that GITR had a similar effect on the accumulation of both the Ly6Chi and Ly6Clo CD4+ T cell subsets. In contrast, whereas GITR increased the accumulation of all three CD8+ T cell subsets defined by CX3CR1 and Ly6C expression, it had a more substantial effect on the least differentiated Ly6Clo CX3CR1lo subset. Moreover, GITR selectively up-regulated CXCR6 on the less differentiated CX3CR1lo CD8+ T cell subsets and induced a small but significant increase in CD127 selectively on the Ly6Clo CD4+ T cell subset. Thus, GITR contributes to accumulation of both differentiated effector cells as well as memory precursors, but with some differences between subsets.


Subject(s)
Antigens, Ly/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Glucocorticoid-Induced TNFR-Related Protein/genetics , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Animals , Antigens, Ly/immunology , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/classification , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/immunology , Cell Differentiation , Cell Movement , Female , Gene Expression Regulation , Glucocorticoid-Induced TNFR-Related Protein/deficiency , Glucocorticoid-Induced TNFR-Related Protein/immunology , Immunologic Memory , Immunophenotyping , Influenza A virus/growth & development , Interleukin-7 Receptor alpha Subunit/genetics , Interleukin-7 Receptor alpha Subunit/immunology , Lung/immunology , Lung/pathology , Lung/virology , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Signal Transduction , Spleen/immunology , Spleen/pathology , Spleen/virology
18.
Diabetes ; 69(5): 965-980, 2020 05.
Article in English | MEDLINE | ID: mdl-32169893

ABSTRACT

As an alternative to lifelong insulin supplementation, potentiation of immune tolerance in patients with type 1 diabetes could prevent the autoimmune destruction of pancreatic islet ß-cells. This study was aimed to assess whether the G3c monoclonal antibody (mAb), which triggers the glucocorticoid-induced TNFR-related (Gitr) costimulatory receptor, promotes the expansion of regulatory T cells (Tregs) in SV129 (wild-type) and diabetic-prone NOD mice. The delivery of the G3c mAb via G3C hybridoma cells enveloped in alginate-based microcapsules (G3C/cps) for 3 weeks induced Foxp3+ Treg-cell expansion in the spleen of wild-type mice but not in Gitr-/- mice. G3C/cps also induced the expansion of nonconventional Cd4+Cd25-/lowFoxp3lowGitrint/high (GITR single-positive [sp]) Tregs. Both Cd4+Cd25+GitrhighFoxp3+ and GITRsp Tregs (including also antigen-specific cells) were expanded in the spleen and pancreas of G3C/cps-treated NOD mice, and the number of intact islets was higher in G3C/cps-treated than in empty cps-treated and untreated animals. Consequently, all but two G3C/cps-treated mice did not develop diabetes and all but one survived until the end of the 24-week study. In conclusion, long-term Gitr triggering induces Treg expansion, thereby delaying/preventing diabetes development in NOD mice. This therapeutic approach may have promising clinical potential for the treatment of inflammatory and autoimmune diseases.


Subject(s)
Antibodies, Monoclonal , Cell Encapsulation , Diabetes Mellitus, Type 1/prevention & control , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Hybridomas , T-Lymphocytes, Regulatory/physiology , Animals , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation , Glucocorticoid-Induced TNFR-Related Protein/genetics , Mice , Mice, Inbred NOD , Mice, Knockout
19.
J Leukoc Biol ; 107(2): 309-321, 2020 02.
Article in English | MEDLINE | ID: mdl-31833599

ABSTRACT

Glucocorticoid-induced tumor necrosis factor receptor family-related protein ligand (GITRL), a member of the tumor necrosis factor superfamily, is expressed in APCs and acts as a costimulatory molecule in the immune system. Although the glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR)/GITRL system has been modulated to promote or decrease T cell-related responses in multiple diseases, studies in macrophages are limited. To address this issue, we compared the expression of GITRL in various types of macrophages and analyzed whether GITRL can affect the fundamental properties and major functions of these cells. Our results demonstrated that M1 polarized macrophages had the highest GITRL levels. Furthermore, GITRL overexpression skewed macrophage polarization toward the M1 phenotype, accelerating proliferation and migration and regulating phagocytosis and killing function. Moreover, GITRL-silenced cells showed a loss of these functions, further confirming its vital role. We also developed an acute peritonitis mouse model, in which macrophages were driven to differentiate into a proinflammatory phenotype with GITRL up-regulation, triggering a positive feedback loop. Our results provide molecular insight into how the GITR/GITRL system modulates innate immune responses, suggesting that manipulation of the GITR/GITRL system to treat diseases depends not only on T cell regulation but also on macrophage participation.


Subject(s)
Bacteria/immunology , Bacterial Infections/immunology , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Immunity, Innate/immunology , Macrophages/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factors/metabolism , Animals , Bacterial Infections/microbiology , Cell Movement , Cell Proliferation , Cells, Cultured , Glucocorticoid-Induced TNFR-Related Protein/genetics , Humans , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Tumor Necrosis Factors/genetics
20.
Eur J Cancer ; 121: 210-223, 2019 11.
Article in English | MEDLINE | ID: mdl-31593831

ABSTRACT

BACKGROUND: Nivolumab and pembrolizumab targeting programmed cell death protein 1 (PD-1) have recently been approved among patients with recurrent and/or metastatic head and neck squamous cell carcinoma (HNSCC) who failed platinum therapy. We aimed to evaluate the prognostic value of selected immune gene expression in HNSCC. PATIENTS AND METHODS: We retrospectively assessed the expression of 46 immune-related genes and immune-cell subpopulation genes including immune checkpoints by real-time polymerase chain reaction among 96 patients with HNSCC who underwent primary surgery at Institut Curie between 1990 and 2006. Univariate and multivariate analyses were performed to assess the prognostic value of dysregulated genes. RESULTS: The Median age of the population was 56 years [range: 35-78]. Primary tumour location was oral cavity (45%), oropharynx (21%), larynx (18%) and hypopharynx (17%). Twelve patients (13%) had an oropharyngeal human papillomavirus-positive tumour. Most significantly overexpressed immune-related genes were TNFRSF9/4-1BB (77%), IDO1 (75%), TNFSF4/OX40L (74%) and TNFRSF18/GITR (74%), and immune-cell subpopulation gene was FOXP3 (62%). Eighty-five percent of tumours analysed overexpressed actionable immunity genes, including PD-1/PD-L1, TIGIT, OX40/OX40L and/or CTLA4. Among the immune-related genes, high OX40L mRNA level (p = 0.0009) and low PD-1 mRNA level (p = 0.004) were associated with the highest risk of recurrence. Among the immune-cell subpopulation genes, patients with high PDGFRB mRNA level (p < 0.0001) and low CD3E (p = 0.0009) or CD8A mRNA levels (p = 0.004) were also at the highest risk of recurrence. CONCLUSIONS: OX40L and PDGFRB overexpression was associated with poor outcomes, whereas PD-1 overexpression was associated with good prognosis in patients with HNSCC treated with primary surgery, suggesting their relevance as potential prognostic biomarkers and major therapeutic targets.


Subject(s)
Biomarkers, Tumor/genetics , Head and Neck Neoplasms/genetics , Immune System Phenomena/genetics , Neoplasm Recurrence, Local/diagnosis , Squamous Cell Carcinoma of Head and Neck/genetics , Adult , Aged , B7-H1 Antigen/genetics , Biomarkers, Tumor/metabolism , CD3 Complex/genetics , CD8 Antigens/genetics , CTLA-4 Antigen/genetics , Female , Gene Expression Regulation, Neoplastic , Glucocorticoid-Induced TNFR-Related Protein/genetics , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/pathology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , OX40 Ligand/genetics , Prognosis , Programmed Cell Death 1 Receptor/genetics , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck/diagnosis , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...