Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 189
Filter
1.
Cancer Immunol Immunother ; 72(9): 3029-3043, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37310433

ABSTRACT

Targeting co-stimulatory receptors promotes the activation and effector functions of anti-tumor lymphocytes. 4-1BB (CD137/TNFSF9), a member of the tumor necrosis factor receptor superfamily (TNFR-SF), is a potent co-stimulatory receptor that plays a prominent role in augmenting effector functions of CD8+ T cells, but also CD4+ T cells and NK cells. Agonistic antibodies against 4-1BB have entered clinical trials and shown signs of therapeutic efficacy. Here, we have used a T cell reporter system to evaluate various formats of 4-1BBL regarding their capacity to functionally engage its receptor. We found that a secreted 4-1BBL ectodomain harboring a trimerization domain derived from human collagen (s4-1BBL-TriXVIII) is a strong inducer of 4-1BB co-stimulation. Similar to the 4-1BB agonistic antibody urelumab, s4-1BBL-TriXVIII is very potent in inducing CD8+ and CD4+ T cell proliferation. We provide first evidence that s4-1BBL-TriXVIII can be used as an effective immunomodulatory payload in therapeutic viral vectors. Oncolytic measles viruses encoding s4-1BBL-TriXVIII significantly reduced tumor burden in a CD34+ humanized mouse model, whereas measles viruses lacking s4-1BBL-TriXVIII were not effective. Natural soluble 4-1BB ligand harboring a trimerization domain might have utility in tumor therapy especially when delivered to tumor tissue as systemic administration might induce liver toxicity.


Subject(s)
4-1BB Ligand , CD8-Positive T-Lymphocytes , Mice , Animals , Humans , 4-1BB Ligand/genetics , Immunomodulating Agents , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Receptors, Tumor Necrosis Factor , Killer Cells, Natural
2.
Life Sci ; 307: 120899, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-35988753

ABSTRACT

AIMS: 4-1BB is a member of the tumor necrosis factor receptor superfamily that mainly expressed on activated T-cells and plays important roles in cell proliferation and survival of T-cells and natural killer cells. The roles of 4-1BB in immune cells have been intensively studied, whereas little is known about the expression and roles of 4-1BB in cancer cells. MAIN METHODS: In the present study, we investigated 4-1BB expression in colorectal cancer tissues from human patients and established colorectal cancer cells, using mRNA expression, FACS, and immunostaining. Cancer cell proliferation and metastasis regulated by transfected 4-1BB was evaluated by cell growth rate, colony forming assay, cell migration, and Western blot with antibodies which are involved in epithelial-mesenchymal transition and anti-apoptosis. Expression of 4-1BB was knockdown by 4-1BB shRNA to prove that 4-1BB was involved in the cell proliferation. In vivo, 4-1BB transfected cancer cells were injected into mice, to induce tumor local region or lung. KEY FINDINGS: We found that colorectal cancer tissues from human patients and established colorectal cancer cells expressed 4-1BB at the high level. The higher expression of 4-1BB proliferated faster. In addition, we identified two forms of 4-1BB detected in colorectal cancer cells: full length form that was located on the plasma membrane and a short soluble form in the cytosol. The soluble form was also detected in the plasma from the mice with tumor xenografts expressed 4-1BB. SIGNIFICANCE: Tumor-mediated 4-1BB expression in the colorectal cancer cells showed effects on cancer cell proliferation, invasion, and metastasis.


Subject(s)
Colorectal Neoplasms , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Animals , Cell Proliferation , Colorectal Neoplasms/pathology , Humans , Mice , RNA, Messenger , RNA, Small Interfering/genetics , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
3.
Nat Commun ; 12(1): 7264, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34907171

ABSTRACT

Antibodies targeting costimulatory receptors of T cells have been developed for the activation of T cell immunity in cancer immunotherapy. However, costimulatory molecule expression is often lacking in tumor-infiltrating immune cells, which can impede antibody-mediated immunotherapy. Here, we hypothesize that delivery of costimulatory receptor mRNA to tumor-infiltrating T cells will enhance the antitumor effects of antibodies. We first design a library of biomimetic nanoparticles and find that phospholipid nanoparticles (PL1) effectively deliver costimulatory receptor mRNA (CD137 or OX40) to T cells. Then, we demonstrate that the combination of PL1-OX40 mRNA and anti-OX40 antibody exhibits significantly improved antitumor activity compared to anti-OX40 antibody alone in multiple tumor models. This treatment regimen results in a 60% complete response rate in the A20 tumor model, with these mice being resistant to rechallenge by A20 tumor cells. Additionally, the combination of PL1-OX40 mRNA and anti-OX40 antibody significantly boosts the antitumor immune response to anti-PD-1 + anti-CTLA-4 antibodies in the B16F10 tumor model. This study supports the concept of delivering mRNA encoding costimulatory receptors in combination with the corresponding agonistic antibody as a strategy to enhance cancer immunotherapy.


Subject(s)
Biomimetic Materials/administration & dosage , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Nanoparticles/administration & dosage , RNA, Messenger/administration & dosage , T-Lymphocytes/immunology , Animals , Biomimetic Materials/chemistry , Drug Delivery Systems , Glycolipids/administration & dosage , Glycolipids/chemistry , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Nanoparticles/chemistry , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Phospholipids/administration & dosage , Phospholipids/chemistry , RNA, Messenger/chemistry , Receptors, OX40/antagonists & inhibitors , Receptors, OX40/genetics , Receptors, OX40/immunology , Receptors, OX40/metabolism , T-Lymphocytes/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
4.
Nat Commun ; 12(1): 7296, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911975

ABSTRACT

CD137 (4-1BB; TNFSR9) is an activation-induced surface receptor that through costimulation effects provide antigen-primed T cells with augmented survival, proliferation and effector functions as well as metabolic advantages. These immunobiological mechanisms are being utilised for cancer immunotherapy with agonist CD137-binding and crosslinking-inducing agents that elicit CD137 intracellular signaling. In this study, side-by-side comparisons show that provision of CD137 costimulation in-cis with regard to the TCR-CD3-ligating cell is superior to that provided in-trans in terms of T cell activation, proliferation, survival, cytokine secretion and mitochondrial fitness in mouse and human. Cis ligation of CD137 relative to the TCR-CD3 complex results in more intense canonical and non-canonical NF-κB signaling and provides a more robust induction of cell cycle and DNA damage repair gene expression programs. Here we report that the superiority of cis versus trans CD137-costimulation is readily observed in vivo and is relevant for understanding the immunotherapeutic effects of CAR T cells and CD137 agonistic therapies currently undergoing clinical trials, which may provide costimulation either in cis or in trans.


Subject(s)
CD3 Complex/immunology , CD8-Positive T-Lymphocytes/immunology , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Animals , CD3 Complex/genetics , Cell Proliferation , Cytokines/genetics , Cytokines/immunology , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Receptor-CD3 Complex, Antigen, T-Cell/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
5.
Mov Disord ; 36(12): 2973-2976, 2021 12.
Article in English | MEDLINE | ID: mdl-34605055

ABSTRACT

Patients with mutations in DJ-1 have early-onset Parkinson's disease and slow progression. Here we describe a Turkish family with a large deletion in the neighboring genes DJ-1 (del exons 1-5) and TNFRSF9 (del exons 1-6), raising the question if TNFRSF9 is a possible disease modifier.


Subject(s)
Parkinson Disease , Protein Deglycase DJ-1 , Sequence Deletion , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Age of Onset , Exons , Humans , Parkinson Disease/genetics , Protein Deglycase DJ-1/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
6.
Int J Biochem Cell Biol ; 136: 106003, 2021 07.
Article in English | MEDLINE | ID: mdl-33971320

ABSTRACT

BACKGROUND: CD137 signaling is an essential factor in cell fate and atherosclerosis. An increase in the number of apoptotic macrophages accelerates atherosclerotic development involving mitochondrial dynamics.However, the role of CD137 signaling in macrophage apoptosis and changes in mitochondria has not been demonstrated clearly. METHODS AND RESULTS: Here, we used ApoE-/- mice as a model of atherosclerotic plaques. Mouse agonist anti-CD137 L and inhibitory anti-CD137 antibody were used to activate or block the CD137 signaling, respectively. Treatment of ApoE-/- mice with agonist anti-CD137 L promoted the formation of necrotic cores and macrophage apoptosis in plaques. Further, activation of CD137 signaling caused macrophage apoptosis in vitro, with upregulation of caspase-9 and caspase-3 expression and an increase in the Bax/Bcl-2 ratio. Meanwhile, CD137 signaling promoted mitochondrial fission observed by mitochondrial fragmentation. Interestingly, inhibition of mitochondrial dynamin-related protein 1 (Drp1) using Mdivi-1 reduced the expression of pro-apoptotic proteins and the amounts of apoptotic macrophages induced by CD137 signaling. Finally, we also found that the p38 MAPK pathway activated by CD137 signaling increased the expression of Drp1 expression and number of mitochondrial fragmented structures. Inhibition of the p38 MAPK pathway by SB203580 attenuated mitochondrial dysfunction through reducing mitochondrial membrane potential loss, cytochrome c release, and mitochondrial reactive oxygen species (ROS) generation. CONCLUSIONS: Overall, we identify a novel mechanism whereby CD137 signaling induces macrophage apoptosis through promoting mitochondrial fission dependent on the p38 MAPK pathway.


Subject(s)
Apoptosis , Macrophages/pathology , Mitochondria/pathology , Plaque, Atherosclerotic/pathology , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Macrophages/metabolism , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Mitochondria/metabolism , Mitochondrial Dynamics , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/metabolism , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , p38 Mitogen-Activated Protein Kinases/genetics
8.
Methods Mol Biol ; 2248: 221-229, 2021.
Article in English | MEDLINE | ID: mdl-33185879

ABSTRACT

The tumor microenvironment (TME) contains noncancerous cells such as various types of immune cells and fibroblasts. Cancer cells direct these stromal cells to create a microenvironment favorable for tumor growth and intercellular interactions have a critical role in this process. In established tumors, interactions between CD137 and its ligand (CD137L) contribute to tumor immune evasion and tumor growth. Therefore, it is important to identify cells expressing CD137 and CD137L within tumors. In this chapter, we will introduce a simple, powerful method of analyzing CD137- and CD137L-expressing tumor cells using Fluorescence-activated cell sorting.


Subject(s)
4-1BB Ligand/metabolism , Neoplasms/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , 4-1BB Ligand/genetics , Biomarkers , Cell Line , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression , Humans , Immunophenotyping/methods , Macrophages/immunology , Macrophages/metabolism , Neoplasms/genetics , Neoplasms/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
9.
Front Immunol ; 11: 539654, 2020.
Article in English | MEDLINE | ID: mdl-33281809

ABSTRACT

While chimeric antigen receptor-modified T (CAR-T) cells have shown great success for the treatment of B cell leukemia, their efficacy appears to be compromised in B cell derived lymphoma and solid tumors. Optimization of the CAR design to improve persistence and cytotoxicity is a focus of the current CAR-T study. Herein, we established a novel CAR structure by adding a full length 4-1BB co-stimulatory receptor to a 28Z-based second generation CAR that targets CD20. Our data indicated that this new 2028Z-4-1BB CAR-T cell showed improved proliferation and cytotoxic ability. To further understand the mechanism of action, we found that constitutive 4-1BB sensing significantly reduced the apoptosis of CAR-T cells, enhanced proliferation, and increased NF-κB pathway activation. Consistent with the enhanced proliferation and cytotoxicity in vitro, this new structure of CAR-T cells exhibited robust persistence and anti-tumor activity in a mouse xenograft lymphoma model. This work provides evidence for a new strategy to optimize the function of CAR-T against lymphoma.


Subject(s)
CD28 Antigens/immunology , Immunotherapy, Adoptive , Lymphoma/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Animals , CD28 Antigens/genetics , Cell Line, Tumor , Humans , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/transplantation , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Xenograft Model Antitumor Assays
10.
Int Immunopharmacol ; 89(Pt B): 107069, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33242709

ABSTRACT

Current treatment of cholangiocarcinoma (CCA) - a lethal bile duct cancer - is ineffective because the disease is usually diagnosed at late and advanced stage. Thus, a novel therapeutic modality is urgently required. Fourth-generation chimeric antigen receptor (CAR4) T cells was created to target CD133, a well-known cancer stem cell marker, that is highly expressed and associates with cancer progression. The anti-CD133-CAR4 T cells showed high efficacy against CD133-expressing CCA cells. Tumour cell lysis occurred in a dose- and CD133 antigen-dependent manner, and significantly higher, up to 57.59% ± 9.62 at effector to target ratio of 5:1 in a CCA cell line - KKU-213A cells, compared to mock control (p = 0.008). Similarly, significant IFN-γ (p = 0.011) and TNF-α (p = 0.002) upregulation was observed upon tumour treatment. The effectiveness of our anti-CD133-CAR4 T cells will be beneficial not only for CD133-expressing CCA, but also for other CD133-expressing tumours. This study may guide future in vivo study and clinical trials.


Subject(s)
AC133 Antigen/metabolism , Bile Duct Neoplasms/therapy , Cholangiocarcinoma/therapy , Immunotherapy, Adoptive , Neoplastic Stem Cells/metabolism , Single-Chain Antibodies/metabolism , T-Lymphocytes/metabolism , AC133 Antigen/immunology , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , CD28 Antigens/genetics , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD3 Complex/genetics , CD3 Complex/immunology , CD3 Complex/metabolism , Cell Line, Tumor , Cholangiocarcinoma/immunology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Coculture Techniques , Cytotoxicity, Immunologic , Humans , Interferon-gamma/metabolism , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Spheroids, Cellular , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
Oncoimmunology ; 9(1): 1838141, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33178496

ABSTRACT

Objectives: Tumor necrosis receptor super family (TNFRSF) plays an important role in regulating the function of CD8+ T cells. In this study, we explored the clinical significance and immune profile of TNFRSF9+ CD8+ T cells in clear cell renal cell carcinoma (ccRCC). Methods: The infiltration of immune cells was determined by immunohistochemistry in ZS cohort from our hospital and their prognostic value was further determined by Cox regression. Functional status of CD8+ T cells in ccRCC was determined by flow cytometry in 29 fresh tumor samples. In silico analysis on a TCGA cohort and other datasets was performed to further demonstrate our findings. Results: High TNFRSF9+ CD8+ T cells infiltration was associated with inferior overall survival in ZS cohort (p = .0016) and TCGA-KIRC cohort (p = .018). TNFRSF9+ CD8+ T cells expressed higher exhaustion markers (PD-1, TIM-3, CTLA-4, and TIGIT), and effector markers (IFN-γ, GZMB, CD107a, and Ki-67), than their TNFRSF9 negative counterparts. In silico analysis indicated the expression of TNFRSF9 was significantly correlated with IFNG, GZMK, MKI-67, PDCD1, HAVCR2, TIGIT, and CTLA-4 in CD8+ T cells. However, higher TNFRSF9 signature was correlated with larger tumor size shrinkage (p = .003) and better progression-free survival (p = .012) in patients treated with nivolumab but not everolimus. Conclusion: TNFRSF9+ CD8+ T cells, which possessed both exhaustion and effector phenotype, were identified as an adverse prognosticator in ccRCC. These cells enrichment was associated with better immunotherapy response which indicated these cells potentially be crucial in immunotherapy.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , CD8-Positive T-Lymphocytes , Carcinoma, Renal Cell/drug therapy , Humans , Immunotherapy , Kidney Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating , Prognosis , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
12.
Biomed Res Int ; 2020: 8765028, 2020.
Article in English | MEDLINE | ID: mdl-33150182

ABSTRACT

Immunotherapy, especially based on chimeric antigen receptor (CAR) T cells, has achieved prominent success in the treatment of hematological malignancies. However, approximately 30-50% of patients will have disease relapse following remission after receiving CD19-targeting CAR-T cells, with failure of maintaining a long-term effect. Mechanisms underlying CAR-T therapy inefficiency consist of loss or modulation of target antigen and CAR-T cell poor persistence which mostly results from T cell exhaustion. The unique features and restoration strategies of exhausted T cells (Tex) have been well described in solid tumors. However, the overview associated with CAR-T cell exhaustion is relatively rare in hematological malignancies. In this review, we summarize the characteristics, cellular, and molecular mechanisms of Tex cells as well as approaches to reverse CAR-T cell exhaustion in hematological malignancies, providing novel strategies for immunotherapies.


Subject(s)
Antigens, CD19/genetics , Clonal Anergy , Hematologic Neoplasms/therapy , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , Antigens, CD19/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CD28 Antigens/genetics , CD28 Antigens/immunology , Gene Expression , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Humans , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Recurrence , T-Lymphocytes/pathology , Treatment Failure , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
13.
J Exp Med ; 217(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-33147322

ABSTRACT

4-1BB (CD137, TNFRSF9) mediates costimulatory signals important for activation and persistence of cytotoxic T lymphocytes. In this issue of JEM, Oda et al. (https://doi.org/10.1084/jem.20191166) report on a chimeric construction encompassing extracellular Fas and intracellular 4-1BB to dramatically improve adoptive T cell therapy.


Subject(s)
Chimera , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Cell- and Tissue-Based Therapy , Immunotherapy, Adoptive , T-Lymphocytes, Cytotoxic , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
14.
BMC Cancer ; 20(1): 1017, 2020 Oct 21.
Article in English | MEDLINE | ID: mdl-33087075

ABSTRACT

BACKGROUND: Thirty to forty percent of patients with Diffuse Large B-cell Lymphoma (DLBCL) have an adverse clinical evolution. The increased understanding of DLBCL biology has shed light on the clinical evolution of this pathology, leading to the discovery of prognostic factors based on gene expression data, genomic rearrangements and mutational subgroups. Nevertheless, additional efforts are needed in order to enable survival predictions at the patient level. In this study we investigated new machine learning-based models of survival using transcriptomic and clinical data. METHODS: Gene expression profiling (GEP) of in 2 different publicly available retrospective DLBCL cohorts were analyzed. Cox regression and unsupervised clustering were performed in order to identify probes associated with overall survival on the largest cohort. Random forests were created to model survival using combinations of GEP data, COO classification and clinical information. Cross-validation was used to compare model results in the training set, and Harrel's concordance index (c-index) was used to assess model's predictability. Results were validated in an independent test set. RESULTS: Two hundred thirty-three and sixty-four patients were included in the training and test set, respectively. Initially we derived and validated a 4-gene expression clusterization that was independently associated with lower survival in 20% of patients. This pattern included the following genes: TNFRSF9, BIRC3, BCL2L1 and G3BP2. Thereafter, we applied machine-learning models to predict survival. A set of 102 genes was highly predictive of disease outcome, outperforming available clinical information and COO classification. The final best model integrated clinical information, COO classification, 4-gene-based clusterization and the expression levels of 50 individual genes (training set c-index, 0.8404, test set c-index, 0.7942). CONCLUSION: Our results indicate that DLBCL survival models based on the application of machine learning algorithms to gene expression and clinical data can largely outperform other important prognostic variables such as disease stage and COO. Head-to-head comparisons with other risk stratification models are needed to compare its usefulness.


Subject(s)
Biomarkers, Tumor/genetics , Computational Biology/methods , Gene Expression Profiling/methods , Lymphoma, Large B-Cell, Diffuse/mortality , Adaptor Proteins, Signal Transducing/genetics , Baculoviral IAP Repeat-Containing 3 Protein/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Male , Microarray Analysis , Middle Aged , Prognosis , RNA-Binding Proteins/genetics , Retrospective Studies , Survival Analysis , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Unsupervised Machine Learning , bcl-X Protein/genetics
15.
Signal Transduct Target Ther ; 5(1): 192, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32895366

Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/immunology , Cytokine Release Syndrome/immunology , Gene Expression Regulation/immunology , Lymphopenia/immunology , Pneumonia, Viral/immunology , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Betacoronavirus/immunology , Biomarkers/blood , COVID-19 , Coronavirus Infections/diagnosis , Coronavirus Infections/genetics , Coronavirus Infections/mortality , Cytokine Release Syndrome/diagnosis , Cytokine Release Syndrome/genetics , Cytokine Release Syndrome/mortality , Disease Progression , Female , Hepatitis A Virus Cellular Receptor 2/blood , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/immunology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/blood , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Lymphocyte Count , Lymphopenia/diagnosis , Lymphopenia/genetics , Lymphopenia/mortality , Male , Middle Aged , Pandemics , Pneumonia, Viral/diagnosis , Pneumonia, Viral/genetics , Pneumonia, Viral/mortality , Retrospective Studies , SARS-CoV-2 , Severity of Illness Index , Survival Analysis , T-Lymphocytes/virology , Tumor Necrosis Factor Receptor Superfamily, Member 7/blood , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/blood , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
17.
Sci Signal ; 13(625)2020 03 31.
Article in English | MEDLINE | ID: mdl-32234960

ABSTRACT

Clinical response to chimeric antigen receptor (CAR) T cell therapy is correlated with CAR T cell persistence, especially for CAR T cells that target CD19+ hematologic malignancies. 4-1BB-costimulated CAR (BBζ) T cells exhibit longer persistence after adoptive transfer than do CD28-costimulated CAR (28ζ) T cells. 4-1BB signaling improves T cell persistence even in the context of 28ζ CAR activation, which indicates distinct prosurvival signals mediated by the 4-1BB cytoplasmic domain. To specifically study signal transduction by CARs, we developed a cell-free, ligand-based activation and ex vivo culture system for CD19-specific CAR T cells. We observed greater ex vivo survival and subsequent expansion of BBζ CAR T cells when compared to 28ζ CAR T cells. We showed that only BBζ CARs activated noncanonical nuclear factor κB (ncNF-κB) signaling in T cells basally and that the anti-CD19 BBζ CAR further enhanced ncNF-κB signaling after ligand engagement. Reducing ncNF-κB signaling reduced the expansion and survival of anti-CD19 BBζ T cells and was associated with a substantial increase in the abundance of the most pro-apoptotic isoforms of Bim. Although our findings do not exclude the importance of other signaling differences between BBζ and 28ζ CARs, they demonstrate the necessary and nonredundant role of ncNF-κB signaling in promoting the survival of BBζ CAR T cells, which likely underlies the engraftment persistence observed with this CAR design.


Subject(s)
NF-kappa B/immunology , Receptors, Chimeric Antigen/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Cell Line , Humans , Receptors, Chimeric Antigen/genetics , Signal Transduction/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
18.
J Oleo Sci ; 69(5): 487-493, 2020 May 02.
Article in English | MEDLINE | ID: mdl-32281564

ABSTRACT

Obesity, a lifestyle disease resulting from excessive caloric intake and insufficient physical activity, results in a state of chronic inflammation. A food ingredient that suppresses chronic inflammation could help prevent associated diseases. Sweet basil (Ocimum basilicum L.) is a herb from the Lamiaceae family with some reported anti-inflammatory effects. Via this in vitro study, we aimed to investigate whether sweet basil exerts anti-inflammatory effects in obese patients. Fresh sweet basil leaves were freeze-dried and powered. After that, this was extracted with 80% methanol. After 3T3-L1 adipocytes were cultured with sweet basil extracts at final concentrations of either 5 or 25 µg/mL for 24h, RAW264.7 macrophages were seeded onto this adipocytes and co-cultured for 12h. We determined the effects of sweet basil extracts on inflammatory cytokine expression by real-time PCR or western blotting. Sweet basil extracts reduced the expression of inflammatory cytokine mRNA induced by co-culture, including that of IL-6 (Il6), IL-1ß (Il1b), TNF-α (Tnf), and CCL2 (Ccl2). In addition, sweet basil extracts suppressed the mRNA expression of NF-κB (Nfκb1), a transcription factor of inflammatory cytokines. In an investigation of costimulatory CD137 (Tnfrsf9)/CD137L inflammatory signaling, a member of the TNF super-family, sweet basil extracts inhibited Tnfrsf9 expression induced by the co-culture. Therefore, the results of this study indicated that sweet basil extracts have an anti-inflammatory effect against adipocyte-induced inflammation, possibly through suppression of Tnfrsf9 expression.


Subject(s)
Adipocytes/metabolism , Anti-Inflammatory Agents , Coculture Techniques , Cytokines/metabolism , Inflammation Mediators/metabolism , Ocimum/chemistry , Plant Extracts/pharmacology , 3T3 Cells , Animals , Cytokines/genetics , Gene Expression/drug effects , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , RAW 264.7 Cells , RNA, Messenger/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
19.
J Cell Mol Med ; 24(8): 4659-4667, 2020 04.
Article in English | MEDLINE | ID: mdl-32149463

ABSTRACT

The role of exosomes derived from endothelial cells (ECs) in the progression of atherosclerosis (AS) and inflammation remains largely unexplored. We aimed to investigate whether exosome derived from CD137-modified ECs (CD137-Exo) played a major role in AS and to elucidate the potential mechanism underlying the inflammatory effect. Exosomes derived from mouse brain microvascular ECs treated with agonist anti-CD137 antibody were used to explore the effect of CD137 signalling in AS and inflammation in vitro and vivo. CD137-Exo efficiently induced the progression of AS in ApoE-/- mice. CD137-Exo increased the proportion of Th17 cells both in vitro and vivo. The IL-6 contained in CD137-Exo which is regulated by Akt and NF-КB pathway was verified to activate Th17 cell differentiation. IL-17 increased apoptosis, inhibited cell viability and improved lactate dehydrogenase (LDH) release in ECs subjected to inflammation induced by lipopolysaccharide (LPS). The expression of soluble intercellular adhesion molecule1 (sICAM-1), monocyte chemoattractant protein-1 (MCP-1) and E-selectin in the supernatants of ECs after IL-17 treatment was dramatically increased. CD137-Exo promoted the progression of AS and Th17 cell differentiation via NF-КB pathway mediated IL-6 expression. This finding provided a potential method to prevent local and peripheral inflammation in AS.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/genetics , Exosomes/genetics , Inflammation/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Animals , Atherosclerosis/chemically induced , Atherosclerosis/pathology , Brain/metabolism , Brain/pathology , Cell Differentiation/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation/genetics , Humans , Inflammation/chemically induced , Inflammation/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , NF-kappa B/genetics , Signal Transduction/genetics , Th17 Cells/metabolism , Th17 Cells/pathology
20.
EBioMedicine ; 52: 102647, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32028068

ABSTRACT

BACKGROUND: Immunotherapy, including checkpoint inhibition, has remarkably improved prognosis in advanced melanoma. Despite this success, acquired resistance is still a major challenge. The T cell costimulatory receptor TNFRSF9 (also known as 4-1BB and CD137) is a promising new target for immunotherapy and two agonistic antibodies are currently tested in clinical trials. However, little is known about epigenetic regulation of the encoding gene. In this study we investigate a possible correlation of TNFRSF9 DNA methylation with gene expression, clinicopathological parameters, molecular and immune correlates, and response to anti-PD-1 immunotherapy to assess the validity of TNFRSF9 methylation to serve as a biomarker. METHODS: We performed a correlation analyses of methylation at twelve CpG sites within TNFRSF9 with regard to transcriptional activity, immune cell infiltration, mutation status, and survival in a cohort of N = 470 melanoma patients obtained from The Cancer Genome Atlas. Furthermore, we used quantitative methylation-specific PCR to confirm correlations in a cohort of N = 115 melanoma patients' samples (UHB validation cohort). Finally, we tested the ability of TNFRSF9 methylation and expression to predict progression-free survival (PFS) and response to anti-PD-1 immunotherapy in a cohort comprised of N = 121 patients (mRNA transcription), (mRNA ICB cohort) and a case-control study including N = 48 patients (DNA methylation, UHB ICB cohort). FINDINGS: We found a significant inverse correlation between TNFRSF9 DNA methylation and mRNA expression levels at six of twelve analyzed CpG sites (P ≤ 0.005), predominately located in the promoter flank region. Consistent with its role as costimulatory receptor in immune cells, TNFRSF9 mRNA expression and hypomethylation positively correlated with immune cell infiltrates and an interferon-γ signature. Furthermore, elevated TNFRSF9 mRNA expression and TNFRSF9 hypomethylation correlated with superior overall survival. In patients receiving anti-PD-1 immunotherapy (mRNA ICB cohort), we found that TNFRSF9 hypermethylation and reduced mRNA expression correlated with poor PFS and response. INTERPRETATION: Our study suggests that TNFRSF9 mRNA expression is regulated via DNA methylation. The observed correlations between TNFRSF9 DNA methylation or mRNA expression with known features of response to immune checkpoint blockage suggest TNFRSF9 methylation could serve as a biomarker in the context of immunotherapies. Concordantly, we identified a correlation between TNFRSF9 DNA methylation and mRNA expression with disease progression in patients under immunotherapy. Our study provides rationale for further investigating TNFRSF9 DNA methylation as a predictive biomarker for response to immunotherapy. FUNDING: AF was partly funded by the Mildred Scheel Foundation. SF received funding from the University Hospital Bonn BONFOR program (O-105.0069). DN was funded in part by DFG Cluster of Excellence ImmunoSensation (EXC 1023). The funders had no role in study design, data collection and analysis, interpretation, decision to publish, or preparation of the manuscript; or any aspect pertinent to the study.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Melanoma/etiology , Melanoma/pathology , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Biomarkers, Tumor , Case-Control Studies , Cytokines/metabolism , Humans , Interferon-gamma , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma/mortality , Melanoma/therapy , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Prognosis , RNA, Messenger
SELECTION OF CITATIONS
SEARCH DETAIL
...