Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 199
Filter
Add more filters

Publication year range
1.
J Cardiovasc Pharmacol ; 83(5): 446-456, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38416872

ABSTRACT

ABSTRACT: Myocardial fibrosis, a common complication of myocardial infarction (MI), is characterized by excessive collagen deposition and can result in impaired cardiac function. The specific role of CD137 in the development of post-MI myocardial fibrosis remains unclear. Thus, this study aimed to elucidate the effects of CD137 signaling using CD137 knockout mice and in vitro experiments. CD137 expression levels progressively increased in the heart after MI, particularly in myofibroblast, which play a key role in fibrosis. Remarkably, CD137 knockout mice exhibited improved cardiac function and reduced fibrosis compared with wild-type mice at day 28 post-MI. The use of Masson's trichrome and picrosirius red staining demonstrated a reduction in the infarct area and collagen volume fraction in CD137 knockout mice. Furthermore, the expression of alpha-smooth muscle actin and collagen I, key markers of fibrosis, was decreased in heart tissues lacking CD137. In vitro experiments supported these findings because CD137 depletion attenuated cardiac fibroblast differentiation, and migration, and collagen I synthesis. In addition, the administration of CD137L recombinant protein further promoted alpha-smooth muscle actin expression and collagen I synthesis, suggesting a profibrotic effect. Notably, the application of an inhibitor targeting the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway attenuated the profibrotic effects of CD137L. To conclude, this study provides evidence that CD137 plays a significant role in promoting myocardial fibrosis after MI. Inhibition of CD137 signaling pathways may hold therapeutic potential for mitigating pathological cardiac remodeling and improving post-MI cardiac function.


Subject(s)
Disease Models, Animal , Fibrosis , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Ventricular Remodeling , Animals , Myocardial Infarction/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/enzymology , Myocardial Infarction/physiopathology , Ventricular Remodeling/drug effects , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Male , Collagen Type I/metabolism , Collagen Type I/genetics , Myofibroblasts/metabolism , Myofibroblasts/pathology , Myofibroblasts/enzymology , MAP Kinase Signaling System , Myocardium/pathology , Myocardium/metabolism , Myocardium/enzymology , 4-1BB Ligand/metabolism , 4-1BB Ligand/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Actins/metabolism , Cells, Cultured , Signal Transduction , Cell Movement , Mice , Ventricular Function, Left , Cell Differentiation , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/drug effects
2.
Immunol Cell Biol ; 101(5): 458-464, 2023 05.
Article in English | MEDLINE | ID: mdl-36811384

ABSTRACT

Since NKG2D ligands (NKG2DLs) are primarily overexpressed on multiple types of solid tumors but absent on most normal tissues, NKG2DLs could be optimal antigens for CAR-T cells. To date, there have been two types of NKG2DL CARs: (i) the extracellular domain of NKG2D fused to the CD8a transmembrane domain, signaling domains of 4-1BB and CD3ζ (NKBz) and (ii) full-length NKG2D fused to the CD3ζ signaling domain (chNKz). Although NKBz- and chNKz-engineered T cells both showed antitumor activities, a comparison of their functions has not been reported. In addition, use of the 4-1BB signaling domain into the CAR construct could prolong the persistence and resistance to antitumor activities of CAR-T cells, we designed a new NKG2DL CAR, full-length NKG2D fused to the signaling domains of 4-1BB and CD3ζ (chNKBz). Among the two types of NKG2DL CAR-T cells reported in previous studies, we found that chNKz T cells had stronger antitumor ability than NKBz T cells in vitro, but their antitumor activity in vivo is similar. The chNKBz T cells showed antitumor activity superior to that of chNKz T cells and NKBz T cells in vitro and in vivo, providing a new option for the immunotherapy of NKG2DL-positive tumor patients.


Subject(s)
Neoplasms , T-Lymphocytes , Humans , Cell Line, Tumor , Immunotherapy , Immunotherapy, Adoptive , NK Cell Lectin-Like Receptor Subfamily K , Signal Transduction , Xenograft Model Antitumor Assays , 4-1BB Ligand/metabolism
3.
Immunol Cell Biol ; 100(9): 705-717, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35916066

ABSTRACT

Aberrant expression of the proto-oncogene BCL6 is a driver of tumorigenesis in diffuse large B cell lymphoma (DLBCL). Mice overexpressing BCL6 from the B cell-specific immunoglobulin heavy chain µ intron promoter (Iµ-Bcl6Tg/+ ) develop B cell lymphomas with features typical of human DLBCL. While the development of B cell lymphoma in these mice is tightly controlled by T cells, the mechanisms of this immune surveillance are poorly understood. Here we show that CD4 T cells contribute to the control of lymphoproliferative disease in lymphoma-prone Iµ-Bcl6Tg/+ mice. We reveal that this CD4 T cell immuno-surveillance requires signaling by the co-stimulatory molecule CD137 ligand (CD137L; also known as 4-1BBL), which may promote the transition of pre-malignant B cells with an activated phenotype into the germinal center stage via reverse signaling, preventing their hazardous accumulation. Thus, CD137L-mediated CD4 T cell immuno-surveillance adds another layer of protection against B cell malignancy to that provided by CD8 T cell cytotoxicity.


Subject(s)
4-1BB Ligand , Lymphoma, Large B-Cell, Diffuse , 4-1BB Ligand/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Germinal Center/metabolism , Humans , Immunoglobulin Heavy Chains , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism
4.
J Immunol ; 204(11): 2887-2899, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32295876

ABSTRACT

CD137 modulates type 1 diabetes (T1D) progression in NOD mice. We previously showed that CD137 expression in CD4 T cells inhibits T1D, but its expression in CD8 T cells promotes disease development by intrinsically enhancing the accumulation of ß-cell-autoreactive CD8 T cells. CD137 is expressed on a subset of FOXP3+ regulatory CD4 T cells (Tregs), and CD137+ Tregs are the main source of soluble CD137. Soluble CD137 suppresses T cells in vitro by binding to the CD137 ligand (CD137L) upregulated on activated T cells. To further study how the opposing functions of CD137 are regulated, we successfully targeted Tnfsf9 (encoding CD137L) in NOD mice using the CRISPR/Cas9 system (designated NOD.Tnfsf9 -/-). Relative to wild-type NOD mice, T1D development in the NOD.Tnfsf9 -/- strain was significantly delayed, and mice developed less insulitis and had reduced frequencies of ß-cell-autoreactive CD8 T cells. Bone marrow chimera experiments showed that CD137L-deficient hematopoietic cells were able to confer T1D resistance. Adoptive T cell transfer experiments showed that CD137L deficiency on myeloid APCs was associated with T1D suppression. Conversely, lack of CD137L on T cells enhanced their diabetogenic activity. Furthermore, neither CD137 nor CD137L was required for the development and homeostasis of FOXP3+ Tregs. However, CD137 was critical for the in vivo T1D-suppressive activity of FOXP3+ Tregs, suggesting that the interaction between CD137 and CD137L regulates their function. Collectively, our results provide new insights into the complex roles of CD137-CD137L interaction in T1D.


Subject(s)
4-1BB Ligand/metabolism , Diabetes Mellitus, Type 1/immunology , T-Lymphocytes, Regulatory/metabolism , 4-1BB Ligand/genetics , Animals , CD4 Antigens/metabolism , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats , Forkhead Transcription Factors/metabolism , Homeostasis , Humans , Immune Tolerance , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, Knockout , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Transplantation Chimera , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
5.
J Immunol ; 204(7): 1892-1903, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32041783

ABSTRACT

4-1BBL, a member of the TNF superfamily, regulates the sustained production of inflammatory cytokines in macrophages triggered by TLR signaling. In this study, we have investigated the role of 4-1BBL in macrophage metabolism and polarization and in skin inflammation using a model of imiquimod-induced psoriasis in mice. Genetic ablation or blocking of 4-1BBL signaling by Ab or 4-1BB-Fc alleviated the pathology of psoriasis by regulating the expression of inflammatory cytokines associated with macrophage activation and regulated the polarization of macrophages in vitro. We further linked this result with macrophage by finding that 4-1BBL expression during the immediate TLR response was dependent on glycolysis, mitochondrial oxidative phosphorylation, and fatty acid metabolism, whereas the late-phase 4-1BBL-mediated sustained inflammatory response was dependent on glycolysis and fatty acid synthesis. Correlating with this, administration of a fatty acid synthase inhibitor, cerulenin, also alleviated the pathology of psoriasis. We further found that 4-1BBL-mediated psoriasis development is independent of its receptor 4-1BB, as a deficiency of 4-1BB augmented the severity of psoriasis linked to a reduced regulatory T cell population and increased IL-17A expression in γδ T cells. Additionally, coblocking of 4-1BBL signaling and IL-17A activity additively ameliorated psoriasis. Taken together, 4-1BBL signaling regulates macrophage polarization and contributes to imiquimod-induced psoriasis by sustaining inflammation, providing a possible avenue for psoriasis treatment in patients.


Subject(s)
4-1BB Ligand/metabolism , Imiquimod/pharmacology , Macrophages/metabolism , Psoriasis/chemically induced , Psoriasis/metabolism , Signal Transduction/physiology , Animals , Cell Line , Female , Inflammation/metabolism , Interleukin-17/metabolism , Macrophage Activation/drug effects , Macrophage Activation/physiology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Signal Transduction/drug effects , Skin/drug effects , Skin/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism
6.
J Immunol ; 205(10): 2893-2904, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33020146

ABSTRACT

The activation of 41BB costimulatory signals by agonistic Abs enhances the expansion and function of tumor-infiltrating lymphocytes (TILs) for treating cancer patients with adoptive cell therapy. However, the impact of 41BB agonism is not limited to enhancing the activity of T cells, and the mechanism by which additional activation of this costimulatory axis in tumor-associated myeloid cells is poorly understood. In this study, we describe that the intratumoral administration of 41BB agonistic Abs led to increases in CD8 T cell infiltration followed by tumor regression in murine models. We found that granulocytes and monocytes rapidly replaced macrophages and dendritic cells in tumors following administration of anti-41BB Abs. Overall, myeloid cells from anti-41BB-treated tumors had an improved capacity to stimulate T cells in comparison with control-treated tumors. In human coculture systems, we demonstrated that the agonism of the 41BB-41BBL axis enhanced costimulatory signals and effector functions among APC and autologous TILs. Overall, these findings suggest that the effect of 41BB agonistic Abs are supported by additional costimulatory signals from tumor-associated myeloid cells,v leading to enhanced TIL expansion and function.


Subject(s)
4-1BB Ligand/agonists , Antineoplastic Agents, Immunological/administration & dosage , CD8-Positive T-Lymphocytes/drug effects , Immunotherapy, Adoptive/methods , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists , 4-1BB Ligand/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Coculture Techniques , Disease Models, Animal , Female , Granulocytes/drug effects , Granulocytes/immunology , Humans , Injections, Intralesional , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Monocytes/drug effects , Monocytes/immunology , Neoplasms/immunology , Neoplasms/pathology , Primary Cell Culture , Tumor Cells, Cultured , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
7.
Eur J Immunol ; 50(9): 1374-1385, 2020 09.
Article in English | MEDLINE | ID: mdl-32357256

ABSTRACT

BACKGROUND: Adoptive NK cell infusion is a promising immunotherapy for acute myeloid leukemia (AML) patients. The aim of this study was to test the activity of clinical-grade membrane-bound IL-21/4-1BBL-expanded NK cell products against AML in vivo. METHODS: Fresh peripheral blood mononuclear cells (PBMCs) were incubated with equal numbers of irradiated membrane-bound IL-21/4-1BBL-expressing K562 cells for 2-3 weeks to induce clinical-grade NK cell expansion. RESULTS: Expansion for 2 and 3 weeks produced ∼4 and 8 × 109 NK cells from 2 × 107 PBMCs. The production of CD107a and TNF-α in NK cell products in response to AML cell lines and primary blasts was higher than that observed in resting NK cells. The 2-week expanded NK cell products were xenografted into immunodeficient mice with leukemia and were persistently found in the BM, spleen, liver, lung, and peripheral blood for at least 13 days; furthermore, these expanded products reduced the AML burden in vivo. Compared with matched AML patients with persistent or relapsed minimal residual disease (MRD+ ) who underwent regular consolidation therapy, MRD+ patients who underwent NK treatment had better overall survival and showed no major adverse events. CONCLUSIONS: Clinical-grade mbIL-21/4-1BBL-expanded NK cells exhibited antileukemic activity against AML in vitro and in vivo.


Subject(s)
Killer Cells, Natural/transplantation , Leukemia, Myeloid, Acute/therapy , 4-1BB Ligand/immunology , 4-1BB Ligand/metabolism , Adult , Aged , Animals , Female , Humans , Interleukins/immunology , Interleukins/metabolism , K562 Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Mice , Middle Aged , Xenograft Model Antitumor Assays
8.
Cancer Immunol Immunother ; 70(9): 2701-2719, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34244816

ABSTRACT

Recombinant agonists that activate co-stimulatory and cytokine receptors have shown limited clinical anticancer utility, potentially due to narrow therapeutic windows, the need for coordinated activation of co-stimulatory and cytokine pathways and the failure of agonistic antibodies to recapitulate signaling by endogenous ligands. RTX-240 is a genetically engineered red blood cell expressing 4-1BBL and IL-15/IL-15Rα fusion (IL-15TP). RTX-240 is designed to potently and simultaneously stimulate the 4-1BB and IL-15 pathways, thereby activating and expanding T cells and NK cells, while potentially offering an improved safety profile through restricted biodistribution. We assessed the ability of RTX-240 to expand and activate T cells and NK cells and evaluated the in vivo efficacy, pharmacodynamics and tolerability using murine models. Treatment of PBMCs with RTX-240 induced T cell and NK cell activation and proliferation. In vivo studies using mRBC-240, a mouse surrogate for RTX-240, revealed biodistribution predominantly to the red pulp of the spleen, leading to CD8 + T cell and NK cell expansion. mRBC-240 was efficacious in a B16-F10 melanoma model and led to increased NK cell infiltration into the lungs. mRBC-240 significantly inhibited CT26 tumor growth, in association with an increase in tumor-infiltrating proliferating and cytotoxic CD8 + T cells. mRBC-240 was tolerated and showed no evidence of hepatic injury at the highest feasible dose, compared with a 4-1BB agonistic antibody. RTX-240 promotes T cell and NK cell activity in preclinical models and shows efficacy and an improved safety profile. Based on these data, RTX-240 is now being evaluated in a clinical trial.


Subject(s)
4-1BB Ligand/genetics , Cell- and Tissue-Based Therapy , Erythrocytes/metabolism , Gene Expression , Genetic Therapy , Interleukin-15/genetics , 4-1BB Ligand/metabolism , Animals , Cell- and Tissue-Based Therapy/methods , Erythroid Precursor Cells/metabolism , Female , Flow Cytometry , Genes, Reporter , Genetic Engineering , Genetic Therapy/methods , Humans , Interleukin-15/metabolism , Mice , Models, Animal , Protein Binding , Treatment Outcome , Xenograft Model Antitumor Assays
9.
J Immunol ; 203(3): 676-685, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31201235

ABSTRACT

To understand and dissect the mechanisms driving human NK cell proliferation, we exploited the methodology used in cell therapy to numerically expand NK cells in the presence of K562-derived artificial APC (aAPCs) and cytokines. For four consecutive weeks, high expression of CD137L by a K562-derived aAPC cell line could sustain NK cell expansion by 3 × 105-fold, whereas low expression of CD137L by the parental K562 cell line only supported the expansion by 2 × 103-fold. The level of expression of CD137L, however, did not modulate the sensitivity of K562 cells to the intrinsic cytotoxicity of NK cells. Similarly, the low NK cell proliferation in the presence of the parental K562 cell line and cytokines was increased by adding agonistic anti-CD137 Abs to levels similar to CD137L-expressing K562-derived aAPCs. Finally, synergy between IL-15 and IL-21 was observed only upon CD137 engagement and the presence of aAPCs. Therefore, we conclude that NK cell proliferation requires cell-to-cell contact, activation of the CD137 axis, and presence of IL-15 (or its membranous form) and IL-21. By analogy with the three-signal model required to activate T cells, we speculate that the cell-to-cell contact represents "signal 1," CD137 represents "signal 2," and cytokines represent "signal 3." The precise nature of signal 1 remains to be defined.


Subject(s)
4-1BB Ligand/metabolism , Interleukin-15/immunology , Interleukins/immunology , Killer Cells, Natural/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Antibodies/immunology , Cell Line, Tumor , Cell Proliferation , Humans , Immunotherapy , Lymphocyte Activation/immunology , Signal Transduction/immunology
10.
Mol Ther ; 28(12): 2577-2592, 2020 12 02.
Article in English | MEDLINE | ID: mdl-32755564

ABSTRACT

T cells engineered to express chimeric antigen receptors (CARs) targeting CD19 have produced impressive outcomes for the treatment of B cell malignancies, but different products vary in kinetics, persistence, and toxicity profiles based on the co-stimulatory domains included in the CAR. In this study, we performed transcriptional profiling of bulk CAR T cell populations and single cells to characterize the transcriptional states of human T cells transduced with CD3ζ, 4-1BB-CD3ζ (BBζ), or CD28-CD3ζ (28ζ) co-stimulatory domains at rest and after activation by triggering their CAR or their endogenous T cell receptor (TCR). We identified a transcriptional signature common across CARs with the CD3ζ signaling domain, as well as a distinct program associated with the 4-1BB co-stimulatory domain at rest and after activation. CAR T cells bearing BBζ had increased expression of human leukocyte antigen (HLA) class II genes, ENPP2, and interleukin (IL)-21 axis genes, and decreased PD1 compared to 28ζ CAR T cells. Similar to previous studies, we also found BBζ CAR CD8 T cells to be enriched in a central memory cell phenotype and fatty acid metabolism genes. Our data uncovered transcriptional signatures related to costimulatory domains and demonstrated that signaling domains included in CARs uniquely shape the transcriptional programs of T cells.


Subject(s)
4-1BB Ligand/chemistry , 4-1BB Ligand/metabolism , Cell Engineering/methods , Protein Domains/genetics , RNA, Small Cytoplasmic/genetics , Receptors, Chimeric Antigen/genetics , Signal Transduction/genetics , T-Lymphocytes/metabolism , Transcriptome , HEK293 Cells , Humans , K562 Cells , RNA-Seq/methods , Single-Cell Analysis , Transduction, Genetic
11.
Proc Natl Acad Sci U S A ; 115(4): E686-E695, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29311309

ABSTRACT

The B-lymphotropic Epstein-Barr virus (EBV), pandemic in humans, is rapidly controlled on initial infection by T cell surveillance; thereafter, the virus establishes a lifelong latent infection in the host. If surveillance fails, fatal lymphoproliferation and lymphomagenesis ensue. The initial T cell response consists of predominantly CD8+ cytotoxic T cells and a smaller expansion of CD4+ cells. A major approach to treating EBV-associated lymphomas is adoptive transfer of autologous or allogeneic T cells that are stimulated/expanded on EBV-transformed B cells. Strikingly, the clinical response correlates with the frequency of CD4 cells in the infused T cells. Although in vitro studies suggested that EBV-specific CD4 cells develop cytotoxicity, they have not been comprehensively characterized and the molecular mechanism underlying their formation remains unknown. Our recent work, using a transgenic approach in mice, has revealed a central role for the EBV signaling molecule LMP1 in immune surveillance and transformation of EBV-infected B cells. The mouse model offers a unique tool for uncovering basic features of EBV immunity. Here, we show that LMP1 expression in B cells induces potent cytotoxic CD4 and CD8 T cell responses, by enhancing antigen presentation and costimulation by CD70, OX40 ligand, and 4-1BB ligand. Our data further suggest that cytotoxic CD4 cells hold superior therapeutic value for LMP1 (EBV)-driven lymphomas. These findings provide insights into EBV immunity, demonstrating that LMP1 signaling alone is sufficient to induce a prominent cytotoxic CD4 response, and suggest strategies for immunotherapy in EBV-related and other cancers.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Herpesvirus 4, Human/immunology , Lymphoma/immunology , Viral Matrix Proteins/immunology , 4-1BB Ligand/metabolism , Animals , B-Lymphocytes/metabolism , CD27 Ligand/metabolism , Lymphoma/virology , Mice, Inbred BALB C , Mice, Inbred C57BL , OX40 Ligand/metabolism , T-Box Domain Proteins/metabolism
12.
Int J Mol Sci ; 22(4)2021 Feb 16.
Article in English | MEDLINE | ID: mdl-33669419

ABSTRACT

We have established an immune cell therapy with immortalized induced pluripotent stem-cell-derived myeloid lines (iPS-ML). The benefits of using iPS-ML are the infinite proliferative capacity and ease of genetic modification. In this study, we introduced 4-1BBL gene to iPS-ML (iPS-ML-41BBL). The analysis of the cell-surface molecules showed that the expression of CD86 was upregulated in iPS-ML-41BBL more than that in control iPS-ML. Cytokine array analysis was performed using supernatants of the spleen cells that were cocultured with iPS-ML or iPS-ML-41BBL. Multiple cytokines that are beneficial to cancer immunotherapy were upregulated. Peritoneal injections of iPS-ML-41BBL inhibited tumor growth of peritoneally disseminated mouse melanoma and prolonged survival of mice compared to that of iPS-ML. Furthermore, the numbers of antigen-specific CD8+ T cells were significantly increased in the spleen and tumor tissues treated with epitope peptide-pulsed iPS-ML-41BBL compared to those treated with control iPS-ML. The number of CXCR6-positive T cells were increased in the tumor tissues after treatment with iPS-ML-41BBL compared to that with control iPS-ML. These results suggest that iPS-ML-41BBL could activate antigen-specific T cells and promote their infiltration into the tumor tissues. Thus, iPS-ML-41BBL may be a candidate for future immune cell therapy aiming to change immunological "cold tumor" to "hot tumor".


Subject(s)
4-1BB Ligand/metabolism , CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Induced Pluripotent Stem Cells/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/therapy , Myeloid Cells/metabolism , Myeloid Cells/transplantation , Skin Neoplasms/therapy , Animals , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , Female , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CXCR6/metabolism , Skin Neoplasms/pathology , Treatment Outcome
13.
Expert Rev Proteomics ; 17(2): 175-185, 2020 02.
Article in English | MEDLINE | ID: mdl-32125181

ABSTRACT

Background: The immune-inducing effect of the quorum sensing (QS) molecule autoinducer-2 (AI-2) on macrophages has not been previously comprehensively studied.Methods: We performed proteomic analysis on macrophages cocultured with purified Fusobacterium nucleatum (F. nucleatum) AI-2 and performed western blot analysis to verify the differential protein expression. We further used the Gene Expression Profiling Interactive Analysis and Tumor Immune Estimation Resource databases to analyze the expression of differentially expressed proteins in microbial-associated digestive tract tumors.Results: Based on proteomic analysis, we identified 46 upregulated proteins and 11 downregulated proteins. The upregulated proteins were mostly inflammatory factors such as tumor necrosis factor ligand superfamily member 9 (TNFSF9). These proteins have a range of biological functions associated with the regulation of inflammatory responses, apoptosis and tumorigenesis. TNFSF9 is highly expressed in pancreatic adenocarcinoma (PAAD) tissues and is associated with M1 polarization of macrophages.Conclusions: Our data indicated that F. nucleatum AI-2 induced inflammatory responses and activated multiple signaling pathways in macrophages. TNFSF9 is the most significantly differentially expressed protein induced by F. nucleatum AI-2 and is involved in regulating immune cell infiltration in PAAD. Thus, AI-2 may become a new focus for studying the relationship between bacteria and cancer.


Subject(s)
Homoserine/analogs & derivatives , Lactones/pharmacology , Macrophage Activation , Macrophages/metabolism , Proteome/metabolism , 4-1BB Ligand/genetics , 4-1BB Ligand/metabolism , Cell Line, Tumor , Fusobacterium nucleatum/chemistry , Homoserine/pharmacology , Humans , Macrophages/drug effects , Proteome/genetics , Proteomics/methods , Tandem Mass Spectrometry/methods , Up-Regulation
14.
Nat Immunol ; 9(8): 917-26, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18604213

ABSTRACT

The costimulatory molecule 4-1BB and its ligand 4-1BBL can control adaptive immunity, but here we show that their interaction also suppressed myelopoiesis. We found that 4-1BBL was expressed on hematopoietic stem cells, differentiating common myeloid progenitors and granulocyte-macrophage progenitors, and 4-1BB was inducible on activated myeloid progenitors. Steady-state numbers of granulocyte-macrophage progenitors, myeloid-lineage cells and mature dendritic cells were higher in 4-1BB- and 4-1BBL-deficient mice, indicative of a negative function, and we confirmed that result with bone marrow chimeras and in vitro, where the absence of interactions between 4-1BB and 4-1BBL led to enhanced differentiation into dendritic cell lineages. The regulatory activity was mediated by 4-1BBL, with binding by 4-1BB inhibiting differentiation of myeloid progenitors. Thus, 4-1BB and 4-1BBL have a previously unknown function in limiting myelopoiesis and the development of dendritic cells.


Subject(s)
4-1BB Ligand/physiology , Dendritic Cells/immunology , Myelopoiesis , Receptors, Nerve Growth Factor/physiology , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/physiology , 4-1BB Ligand/metabolism , Animals , Mice , Receptors, Nerve Growth Factor/genetics , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/genetics
15.
J Autoimmun ; 112: 102499, 2020 08.
Article in English | MEDLINE | ID: mdl-32505443

ABSTRACT

CD137 (TNFRSF9, 4-1BB) is a potent co-stimulatory molecule of the tumour necrosis factor receptor superfamily (TNFRSF) that is expressed by activated T cells. CD137/CD137 ligand (CD137L) signalling primarily induces a potent cell-mediated immune response, while signalling of cell surface-expressed CD137L into antigen presenting cells enhances their activation, differentiation and migratory capacity. Studies have shown that bidirectional CD137/CD137L signalling plays an important role in the pathogenesis of autoimmune diseases. This review discusses the mechanisms how CD137/CD137L signalling contributes to immune deviation of helper T cell pathways in various murine models, and the potential of developing immunotherapies targeting CD137/CD137L signalling for the treatment of autoimmune diseases.


Subject(s)
4-1BB Ligand/metabolism , Autoimmune Diseases/drug therapy , Immunologic Factors/pharmacology , Signal Transduction/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , 4-1BB Ligand/antagonists & inhibitors , Animals , Autoimmune Diseases/immunology , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Humans , Immunologic Factors/therapeutic use , Mice , Molecular Targeted Therapy/methods , Signal Transduction/drug effects , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors
16.
J Cardiovasc Pharmacol ; 75(2): 148-154, 2020 02.
Article in English | MEDLINE | ID: mdl-31651672

ABSTRACT

BACKGROUND: Angiogenesis in atherosclerotic plaque is an important factor causing plaque hemorrhage, vulnerability, and rupture, and different phenotypes of macrophages have different effects on angiogenesis. Our previous study has demonstrated CD137-CD137L signaling, a pair of inflammatory costimulatory molecules, can promote angiogenesis in atherosclerotic plaque. Therefore, we aimed to investigate whether this signaling could affect angiogenesis by regulating phenotypic transition of macrophages. METHODS: Male mouse primary peritoneal macrophages were extracted by intraperitoneal injection of thioglycollate, and then flow cytometry was used to detect the expression of CD137. Flow cytometry, Western blotting, quantitative real-time PCR, and enzyme-linked immunosorbent assay (ELISA) were used to assess the phenotypic changes of macrophages after different treatment. Mouse brain microvascular endothelial cells (bEnd.3) were cocultured with macrophages, and tube formation was assessed to examine angiogenesis. RESULTS: We found that the number of junctions and branches of bEnd.3 were increased when CD137-CD137L signaling was activated, while such number was further increased when bEnd.3 were cocultured with macrophages. Flow cytometry showed that CD137 was expressed on almost all primary peritoneal macrophages. The expression of CD86 was decreased in the agonist CD137L group and increased in the agonist CD137L + inhibitory anti-CD137 antibody group after adding the CD137 inhibitor. The expression of CD206 in each group exhibited opposite trend compared with CD86. Moreover, the expression of inducible nitric oxide synthase at the mRNA, and protein levels were decreased after stimulating CD137-CD137L signaling, and such downward trend was reversed when CD137-CD137L signaling was inhibited. Furthermore, the expression of arginase-1 was opposite to that of inducible nitric oxide synthase. Enzyme-linked immunosorbent assay indicated that the content of interleukin-12 (IL-12) in the supernatant of macrophages in the agonist CD137L group was lower than that in the control group, and its content in the inhibited group was higher than that in the activated group. The change of interleukin-10 (IL-10) content in macrophage supernatant was opposite to that of IL-12. When AKT serine/threonine kinase 1 (Akt1) inhibitor was used to inhibit the phenotypic transformation of macrophages induced by CD137-CD137L, the number of junctions and branches formed by bEnd.3 was decreased compared with the coculture group. CONCLUSIONS: These results indicated that CD137-CD137L signaling could promote angiogenesis by regulating phenotypic transition of macrophages of male mice.


Subject(s)
4-1BB Ligand/metabolism , Cell Plasticity , Endothelial Cells/metabolism , Macrophages, Peritoneal/metabolism , Neovascularization, Physiologic , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Animals , Arginase/metabolism , B7-2 Antigen/metabolism , Cell Line , Coculture Techniques , Endothelial Cells/immunology , Intercellular Junctions/immunology , Intercellular Junctions/metabolism , Interleukin-10/metabolism , Interleukin-12/metabolism , Lectins, C-Type/metabolism , Macrophages, Peritoneal/immunology , Male , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction
17.
Mol Ther ; 27(11): 1906-1918, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31466933

ABSTRACT

Oncolytic viruses are an excellent platform for developing effective strategies in cancer immunotherapy. Several challenges remain in the use of viro-immunotherapy for cancer, such as the lack of costimulatory signals and negative regulation of immune checkpoints. In this study, we designed a novel adenovirus expressing a soluble fusion protein, programmed cell death protein 1 (PD-1)/CD137L, which contains the extracellular domains of PD-1 and CD137L at each terminus (Ad5-PC). Ad5-PC preserved the costimulatory activity of CD137L and facilitated the persistence of activated CD8+ T cells. Ad5-PC induced strikingly increased antitumor activity in both ascitic and subcutaneous hepatocellular carcinoma (HCC) tumor models, with 70% and 60% long-term cure rates, respectively. The improved antitumor effect of Ad5-PC was attributed to the sustained high-level lymphocyte activation and interferon (IFN)-γ production in the tumor microenvironment, and was essentially dependent on CD8+ T cells rather than natural killer (NK) cells. Moreover, Ad5-huPC-expressing human soluble PD-1/CD137L fusion protein was effective in suppressing tumor growth and improving survival in a humanized mouse model. We confirmed that Ad5-PC induced tumor-specific and systematic protection against tumor rechallenges at both in situ and distant sites. Thus, Ad5-PC harnesses several distinct functions to efficiently overcome several major hurdles of viro-immunotherapy.


Subject(s)
4-1BB Ligand/genetics , Adenoviridae/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Genetic Vectors/genetics , Programmed Cell Death 1 Receptor/genetics , Recombinant Fusion Proteins/genetics , 4-1BB Ligand/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Line, Tumor , Disease Models, Animal , Gene Expression , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Immunomodulation/genetics , Immunomodulation/immunology , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Lymphocyte Activation/immunology , Mice , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction , Treatment Outcome , Xenograft Model Antitumor Assays
18.
J Biol Chem ; 293(26): 9958-9969, 2018 06 29.
Article in English | MEDLINE | ID: mdl-29720398

ABSTRACT

Human (h)4-1BB (TNFRSF9 or CD137) is an inducible tumor necrosis factor receptor (TNFR) superfamily member that interacts with its cognate ligand h4-1BBL to promote T lymphocyte activation and proliferation. h4-1BB is currently being targeted with agonists in cancer immunotherapy. Here, we determined the crystal structures of unbound h4-1BBL and both WT h4-1BB and a dimerization-deficient h4-1BB mutant (C121S) in complex with h4-1BBL at resolutions between 2.7 and 3.2 Å. We observed that the structural arrangement of 4-1BBL, both unbound and in the complex, represents the canonical bell shape as seen in other similar TNF proteins and differs from the previously reported three-bladed propeller structure of 4-1BBL. We also found that the binding site for the receptor is at the crevice formed between two protomers of h4-1BBL, but that h4-1BB interacts predominantly with only one ligand protomer. Moreover, h4-1BBL lacked the conserved tyrosine residue in the DE loop that forms canonical interactions between other TNFR family molecules and their ligands, suggesting h4-1BBL engages h4-1BB through a distinct mechanism. Of note, we discovered that h4-1BB forms a disulfide-linked dimer because of the presence of an additional cysteine residue found in its cysteine-rich domain 4 (CRD4). As a result, h4-1BB dimerization, in addition to trimerization via h4-1BBL binding, could result in cross-linking of individual ligand-receptor complexes to form a 2D network that stimulates strong h4-1BB signaling. This work provides critical insights into the structural and functional properties of both h4-1BB and h4-1BBL and reveals that covalent receptor dimerization amplifies h4-1BB signaling.


Subject(s)
4-1BB Ligand/metabolism , Protein Multimerization , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 9/chemistry , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , 4-1BB Ligand/chemistry , Crystallography, X-Ray , Humans , Ligands , Protein Binding , Protein Structure, Quaternary
19.
J Biol Chem ; 293(25): 9880-9891, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29720399

ABSTRACT

4-1BBL is a member of the tumor necrosis factor (TNF) superfamily and is the ligand for the TNFR superfamily receptor, 4-1BB. 4-1BB plays an immunomodulatory role in T cells and NK cells, and agonists of this receptor have garnered strong attention as potential immunotherapy agents. Broadly speaking, the structural features of TNF superfamily members, their receptors, and ligand-receptor complexes are similar. However, a published crystal structure of human 4-1BBL suggests that it may be unique in this regard, exhibiting a three-bladed propeller-like trimer assembly that is distinctly different from that observed in other family members. This unusual structure also suggests that the human 4-1BB/4-1BBL complex may be structurally unique within the TNF/TNFR superfamily, but to date no structural data have been reported. Here we report the crystal structure of the human 4-1BB/4-1BBL complex at 2.4-Å resolution. In this structure, 4-1BBL does not adopt the unusual trimer assembly previously reported, but instead forms a canonical bell-shaped trimer typical of other TNF superfamily members. The structure of 4-1BB is also largely canonical as is the 4-1BB/4-1BBL complex. Mutational data support the 4-1BBL structure reported here as being biologically relevant, suggesting that the previously reported structure is not. Together, the data presented here offer insight into structure/function relationships in the 4-1BB/4-1BBL system and improve our structural understanding of the TNF/TNFR superfamily more broadly.


Subject(s)
4-1BB Ligand/chemistry , 4-1BB Ligand/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/chemistry , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Binding Sites , Crystallography, X-Ray , HEK293 Cells , Humans , Ligands , Protein Binding , Protein Conformation , Protein Multimerization
20.
J Biol Chem ; 293(4): 1317-1329, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29242193

ABSTRACT

4-1BB (CD137) is a TNF receptor superfamily (TNFRSF) member that is thought to undergo receptor trimerization upon binding to its trimeric TNF superfamily ligand (4-1BBL) to stimulate immune responses. 4-1BB also can bind to the tandem repeat-type lectin galectin-9 (Gal-9), and signaling through mouse (m)4-1BB is reduced in galectin-9 (Gal-9)-deficient mice, suggesting a pivotal role of Gal-9 in m4-1BB activation. Here, using sulfur-SAD phasing, we determined the crystal structure of m4-1BB to 2.2-Å resolution. We found that similar to other TNFRSFs, m4-1BB has four cysteine-rich domains (CRDs). However, the organization of CRD1 and the orientation of CRD3 and CRD4 with respect to CRD2 in the m4-1BB structure distinctly differed from those of other TNFRSFs. Moreover, we mapped two Asn residues within CRD4 that are N-linked glycosylated and mediate m4-1BB binding to Gal-9. Kinetics studies of m4-1BB disclosed a very tight nanomolar binding affinity to m4-1BBL with an unexpectedly strong avidity effect. Both N- and C-terminal domains of Gal-9 bound m4-1BB, but with lower affinity compared with m4-1BBL. Although the TNF homology domain (THD) of human (h)4-1BBL forms non-covalent trimers, we found that m4-1BBL formed a covalent dimer via 2 cysteines absent in h4-1BBL. As multimerization and clustering is a prerequisite for TNFR intracellular signaling, and as m4-1BBL can only recruit two m4-1BB monomers, we hypothesize that m4-1BBL and Gal-9 act together to aid aggregation of m4-1BB monomers to efficiently initiate m4-1BB signaling.


Subject(s)
4-1BB Ligand/chemistry , Galectins/chemistry , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 9/chemistry , 4-1BB Ligand/genetics , 4-1BB Ligand/metabolism , Animals , Crystallography, X-Ray , Galectins/genetics , Galectins/metabolism , HEK293 Cells , Humans , Mice , Protein Domains , Protein Structure, Quaternary , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL