Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
2.
J Hematol Oncol ; 15(1): 55, 2022 05 07.
Article in English | MEDLINE | ID: mdl-35526043

ABSTRACT

Lysophosphatidic acid (LPA) is a naturally occurring phospholipid that regulates cell proliferation, survival, and migration. However, its role on human multiple myeloma (MM) cells is largely unknown. In this study, we show that LPA, which is highly elevated in MM patients, plays an important role in protecting human MM cells against proteasome inhibitor (PI)-induced apoptosis. LPA bound to its receptor LPAR2 activated its downstream MEK1/2-ERK1/2 signaling pathway and enhanced oxidative phosphorylation (OXPHOS) in mitochondria in MM cells. Increased OXPHOS activity produced more NAD+ and ATP, reduced proteasome activity, and enhanced protein folding and refolding in endoplasmic reticulum (ER), leading to induction of MM resistance to PIs. Importantly, inhibiting LPAR2 activity or knocking out LPAR2 in MM cells significantly enhanced MM sensitivity to PI-induced apoptosis in vitro and in vivo. Interestingly, primary MM cells from LPA-high patients were more resistant to PI-induced apoptosis than MM cells from LPA-low patients. Thus, our study indicates that LPA-LPAR2-mediated signaling pathways play an important role in MM sensitivity to PIs and targeting LPA or LPAR2 may potentially be used to (re)sensitize patients to PI-based therapy.


Subject(s)
Multiple Myeloma , Proteasome Inhibitors , Apoptosis , Humans , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism
4.
J Clin Invest ; 132(7)2022 04 01.
Article in English | MEDLINE | ID: mdl-35192544

ABSTRACT

CD8+ T cell longevity regulated by metabolic activity plays important roles in cancer immunotherapy. Although in vitro-polarized, transferred IL-9-secreting CD8+ Tc9 (cytotoxic T lymphocyte subset 9) cells exert greater persistence and antitumor efficacy than Tc1 cells, the underlying mechanism remains unclear. Here, we show that tumor-infiltrating Tc9 cells display significantly lower lipid peroxidation than Tc1 cells in several mouse models, which is strongly correlated with their persistence. Using RNA-sequence and functional validation, we found that Tc9 cells exhibited unique lipid metabolic programs. Tc9 cell-derived IL-9 activated STAT3, upregulated fatty acid oxidation and mitochondrial activity, and rendered Tc9 cells with reduced lipid peroxidation and resistance to tumor- or ROS-induced ferroptosis in the tumor microenvironment. IL-9 signaling deficiency, inhibiting STAT3, or fatty acid oxidation increased lipid peroxidation and ferroptosis of Tc9 cells, resulting in impaired longevity and antitumor ability. Similarly, human Tc9 cells also exhibited lower lipid peroxidation than Tc1 cells and tumor-infiltrating CD8+ T cells expressed lower IL9 and higher lipid peroxidation- and ferroptosis-related genes than circulating CD8+ T cells in patients with melanoma. This study indicates that lipid peroxidation regulates Tc9 cell longevity and antitumor effects via the IL-9/STAT3/fatty acid oxidation pathway and regulating T cell lipid peroxidation can be used to enhance T cell-based immunotherapy in human cancer.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-9 , Animals , CD8-Positive T-Lymphocytes/metabolism , Fatty Acids/metabolism , Humans , Immunotherapy/methods , Interleukin-9/genetics , Lipid Peroxidation , Mice , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
5.
Blood ; 139(1): 59-72, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34411225

ABSTRACT

Proteasome inhibitors (PIs) such as bortezomib (Btz) and carfilzomib (Cfz) are highly efficacious for patients with multiple myeloma (MM). However, relapses are frequent, and acquired resistance to PI treatment emerges in most patients. Here, we performed a high-throughput screen of 1855 Food and Drug Administration (FDA)-approved drugs and identified all-trans retinoic acid (ATRA), which alone has no antimyeloma effect, as a potent drug that enhanced MM sensitivity to Cfz-induced cytotoxicity and resensitized Cfz-resistant MM cells to Cfz in vitro. ATRA activated retinoic acid receptor (RAR)γ and interferon-ß response pathway, leading to upregulated expression of IRF1. IRF1 in turn initiated the transcription of OAS1, which synthesized 2-5A upon binding to double-stranded RNA (dsRNA) induced by Cfz and resulted in cellular RNA degradation by RNase L and cell death. Similar to ATRA, BMS961, a selective RARγ agonist, could also (re)sensitize MM cells to Cfz in vitro, and both ATRA and BMS961 significantly enhanced the therapeutic effects of Cfz in established MM in vivo. In support of these findings, analyses of large datasets of patients' gene profiling showed a strong and positive correlation between RARγ and OAS1 expression and patient's response to PI treatment. Thus, this study highlights the potential for RARγ agonists to sensitize and overcome MM resistance to Cfz treatment in patients.


Subject(s)
Antineoplastic Agents/pharmacology , Immunity, Innate/drug effects , Multiple Myeloma/drug therapy , Oligopeptides/pharmacology , Receptors, Retinoic Acid/agonists , 2',5'-Oligoadenylate Synthetase/immunology , Cell Line, Tumor , Endoribonucleases/immunology , Humans , Receptors, Retinoic Acid/immunology , Tumor Cells, Cultured , Retinoic Acid Receptor gamma
6.
Cell Metab ; 33(5): 1001-1012.e5, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33691090

ABSTRACT

Understanding the mechanisms underlying how T cells become dysfunctional in a tumor microenvironment (TME) will greatly benefit cancer immunotherapy. We found that increased CD36 expression in tumor-infiltrating CD8+ T cells, which was induced by TME cholesterol, was associated with tumor progression and poor survival in human and murine cancers. Genetic ablation of Cd36 in effector CD8+ T cells exhibited increased cytotoxic cytokine production and enhanced tumor eradication. CD36 mediated uptake of fatty acids by tumor-infiltrating CD8+ T cells in TME, induced lipid peroxidation and ferroptosis, and led to reduced cytotoxic cytokine production and impaired antitumor ability. Blocking CD36 or inhibiting ferroptosis in CD8+ T cells effectively restored their antitumor activity and, more importantly, possessed greater antitumor efficacy in combination with anti-PD-1 antibodies. This study reveals a new mechanism of CD36 regulating the function of CD8+ effector T cells and therapeutic potential of targeting CD36 or inhibiting ferroptosis to restore T cell function.


Subject(s)
CD36 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Ferroptosis , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , CD36 Antigens/antagonists & inhibitors , CD36 Antigens/genetics , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cytokines/metabolism , Fatty Acids/metabolism , Ferroptosis/drug effects , Humans , Immunotherapy , Lipid Peroxidation , Melanoma, Experimental/drug therapy , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Multiple Myeloma/therapy , Reactive Oxygen Species/metabolism , Survival Rate , Tumor Microenvironment
7.
Nat Commun ; 11(1): 5902, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33214555

ABSTRACT

CAR-T cell therapy is effective for hematologic malignancies. However, considerable numbers of patients relapse after the treatment, partially due to poor expansion and limited persistence of CAR-T cells in vivo. Here, we demonstrate that human CAR-T cells polarized and expanded under a Th9-culture condition (T9 CAR-T) have an enhanced antitumor activity against established tumors. Compared to IL2-polarized (T1) cells, T9 CAR-T cells secrete IL9 but little IFN-γ, express central memory phenotype and lower levels of exhaustion markers, and display robust proliferative capacity. Consequently, T9 CAR-T cells mediate a greater antitumor activity than T1 CAR-T cells against established hematologic and solid tumors in vivo. After transfer, T9 CAR-T cells migrate effectively to tumors, differentiate to IFN-γ and granzyme-B secreting effector memory T cells but remain as long-lived and hyperproliferative T cells. Our findings are important for the improvement of CAR-T cell-based immunotherapy for human cancers.


Subject(s)
Cytotoxicity, Immunologic , Immunotherapy, Adoptive/methods , Interleukin-9/metabolism , T-Lymphocytes/immunology , Animals , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cytokines/metabolism , Humans , Immunologic Memory , Interferon-gamma/metabolism , Mice , Phenotype , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/transplantation , Xenograft Model Antitumor Assays
8.
Blood ; 136(22): 2557-2573, 2020 11 26.
Article in English | MEDLINE | ID: mdl-32582913

ABSTRACT

Multiple myeloma (MM) remains largely incurable despite significant advances in biotherapy and chemotherapy. The development of drug resistance is a major problem in MM management. Macrophage migration inhibitory factor (MIF) expression was significantly higher in purified MM cells from relapsed patients than those with sustained response, and MM patients with high MIF had significantly shorter progression-free survival (PFS) and overall survival (OS). MM cell lines also express high levels of MIF, and knocking out MIF made them more sensitive to proteasome inhibitor (PI)-induced apoptosis not observed with other chemotherapy drugs. Mechanistic studies showed that MIF protects MM cells from PI-induced apoptosis by maintaining mitochondrial function via suppression of superoxide production in response to PIs. Specifically, MIF, in the form of a homotrimer, acts as a chaperone for superoxide dismutase 1 (SOD1) to suppress PI-induced SOD1 misfolding and to maintain SOD1 activity. MIF inhibitor 4-iodo-6-phenylpyrimidine and homotrimer disrupter ebselen, which do not kill MM cells, enhanced PI-induced SOD1 misfolding and loss of function, resulting in significantly more cell death in both cell lines and primary MM cells. More importantly, inhibiting MIF activity in vivo displayed synergistic antitumor activity with PIs and resensitized PI-resistant MM cells to treatment. In support of these findings, gene-profiling data showed a significantly negative correlation between MIF and SOD1 expression and response to PI treatment in patients with MM. This study shows that MIF plays a crucial role in MM sensitivity to PIs and suggests that targeting MIF may be a promising strategy to (re)sensitize MM to the treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Multiple Myeloma , Neoplasm Proteins/metabolism , Proteasome Inhibitors/pharmacology , Animals , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Cancer Res ; 80(7): 1438-1450, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32015091

ABSTRACT

Tumor-associated macrophages (TAM) are important tumor-promoting cells. However, the mechanisms underlying how the tumor and its microenvironment reprogram these cells remain elusive. Here we report that lipids play a crucial role in generating TAMs in the tumor microenvironment (TME). Macrophages from both human and murine tumor tissues were enriched with lipids due to increased lipid uptake by macrophages. TAMs expressed elevated levels of the scavenger receptor CD36, accumulated lipids, and used fatty acid oxidation (FAO) instead of glycolysis for energy. High levels of FAO promoted mitochondrial oxidative phosphorylation, production of reactive oxygen species, phosphorylation of JAK1, and dephosphorylation of SHP1, leading to STAT6 activation and transcription of genes that regulate TAM generation and function. These processes were critical for TAM polarization and activity, both in vitro and in vivo. In summary, we highlight the importance of lipid metabolism in the differentiation and function of protumor TAMs in the TME. SIGNIFICANCE: This study highlights the role of lipid metabolism in the differentiation and function of TAMs and suggests targeting TAM fatty acid oxidation as a potential therapeutic modality for human cancers.


Subject(s)
Cell Differentiation/immunology , Lipid Metabolism/immunology , Macrophages/immunology , Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor/transplantation , Datasets as Topic , Disease Models, Animal , Fatty Acids/metabolism , Female , Humans , Macrophages/metabolism , Male , Mice , Mitochondria/metabolism , Neoplasms/pathology , Oxidation-Reduction , Oxidative Phosphorylation , Primary Cell Culture , Reactive Oxygen Species/metabolism
11.
Cell Metab ; 30(1): 143-156.e5, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31031094

ABSTRACT

Tumor-infiltrating T cells often lose their effector function; however, the mechanisms are incompletely understood. We report that cholesterol in the tumor microenvironment induces CD8+ T cell expression of immune checkpoints and exhaustion. Tumor tissues enriched with cholesterol and cholesterol content in tumor-infiltrating CD8+ T cells were positively and progressively associated with upregulated T cell expression of PD-1, 2B4, TIM-3, and LAG-3. Adoptively transferred CD8+ T cells acquired cholesterol, expressed high levels of immune checkpoints, and became exhausted upon entering a tumor. Tumor culture supernatant or cholesterol induced immune checkpoint expression by increasing endoplasmic reticulum (ER) stress in CD8+ T cells. Consequently, the ER stress sensor XBP1 was activated and regulated PD-1 and 2B4 transcription. Inhibiting XBP1 or reducing cholesterol in CD8+ T cells effectively restored antitumor activity. This study reveals a mechanism underlying T cell exhaustion and suggests a new strategy for restoring T cell function by reducing cholesterol to enhance T cell-based immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Cholesterol/blood , Tumor Microenvironment/physiology , Animals , Blotting, Western , Flow Cytometry , Humans , Immunoprecipitation , Immunotherapy , Melanoma, Experimental/blood , Mice , Programmed Cell Death 1 Receptor/metabolism , Real-Time Polymerase Chain Reaction
12.
J Clin Invest ; 128(11): 4821-4831, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30277474

ABSTRACT

Plasmacytoid dendritic cells (pDCs) play a key role in antiviral responses by producing type-1 IFNs. However, recent studies showed that pDCs induce immune suppression and promote tumor growth in human ovarian cancer and myeloma. The molecular mechanisms underlying pDC acquisition of these properties are unknown. Here we show that human pDCs activated by CpG inhibited growth and induced apoptosis in myeloma cells via secreted IFN-α, but direct contact with myeloma cells converted pDCs into tumor-promoting cells by suppressing pDC IFN-α production. E-cadherin, expressed on both myeloma cells and pDCs, mediated these effects via a homophilic interaction - activation of E-cadherin signaling upregulated and activated TNFAIP3 to interact with TLR9, resulting in TLR9 ubiquitination and degradation, and inhibition of IFN-α production in pDCs. These findings were supported by an in vivo study in which pDC depletion induced tumor regression and better survival in the Vk*MYC myeloma mouse model. Furthermore, IFNAR1 expression level positively correlated to overall survival of patients with multiple myeloma (MM), and the IFN-α level in patient bone marrow was significantly lower than that in marrow of healthy individuals. This study reveals a novel mechanism underlying how MM tumors educate pDCs in their microenvironment and provides new targets for improving the treatment of MM.


Subject(s)
Antigens, CD/immunology , Cadherins/immunology , Dendritic Cells/immunology , Gene Expression Regulation, Neoplastic/immunology , Immune Tolerance , Multiple Myeloma/immunology , Neoplasm Proteins/immunology , Animals , Antigens, CD/genetics , Bone Marrow/immunology , Bone Marrow/pathology , Cadherins/genetics , Dendritic Cells/pathology , Female , Humans , Interferon-alpha/genetics , Interferon-alpha/immunology , Male , Mice , Mice, Transgenic , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplasm Proteins/genetics , Oligodeoxyribonucleotides/pharmacology , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/immunology
13.
J Lipid Res ; 59(12): 2287-2296, 2018 12.
Article in English | MEDLINE | ID: mdl-30309895

ABSTRACT

Production of 25-hydroxycholesterol (25HC), a potent inhibitor of viral infection, is catalyzed by cholesterol 25-hydroxylase (CH25H). We previously reported that 25HC induced CH25H expression in a liver X receptor (LXR)-dependent manner, implying that LXR can play an important role in antiviral infection. In this study, we determined that activation of LXR by 25HC or synthetic ligands [T0901317 (T317) or GW3965] inhibited infection of herpes simplex virus type 1 (HSV-1) or MLV-(VSV)-GFP in HepG2 cells or RAW 264.7 macrophages. Genetic deletion of LXRα, LXRß, or CH25H expression in HepG2 cells by CRISPR/Cas9 method increased cell susceptibility to HSV-1 infection and attenuated the inhibition of LXR on viral infection. Lack of interferon (IFN)-γ expression also increased cell susceptibility to viral infection. However, it attenuated, but did not block, the inhibition of LXR on HSV-1 infection. In addition, expression of CH25H, but not IFN-γ, was inversely correlated to cell susceptibility to viral infection and the antiviral actions of LXR. Metabolism of 25HC into 25HC-3-sulfate (25HC3S) by cholesterol sulfotransferase-2B1b moderately reduced the antiviral actions of 25HC because 25HC3S is a weaker inhibitor of HSV-1 infection than 25HC. Furthermore, administration of T317 to BALB/c mice reduced HSV-1 growth in mouse tissues. Taken together, we demonstrate an antiviral system of 25HC with involvement of LXR activation, interaction between CH25H and IFN-γ, and 25HC metabolism.


Subject(s)
Hydroxycholesterols/metabolism , Liver X Receptors/metabolism , Animals , Blotting, Western , CRISPR-Cas Systems/genetics , Hep G2 Cells , Herpesvirus 1, Human/metabolism , Humans , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RAW 264.7 Cells , Real-Time Polymerase Chain Reaction , Sulfotransferases/metabolism
14.
Cancer Cell ; 33(6): 1048-1060.e7, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29894691

ABSTRACT

The antitumor effector T helper 1 (Th1) and Th17 cells represent two T cell paradigms: short-lived cytolytic Th1 cells and "stem cell-like" memory Th17 cells. We report that Th9 cells represent a third paradigm-they are less-exhausted, fully cytolytic, and hyperproliferative. Only tumor-specific Th9 cells completely eradicated advanced tumors, maintained a mature effector cell signature with cytolytic activity as strong as Th1 cells, and persisted as long as Th17 cells in vivo. Th9 cells displayed a unique Pu.1-Traf6-NF-κB activation-driven hyperproliferative feature, suggesting a persistence mechanism rather than an antiapoptotic strategy. Th9 antitumor efficacy depended on interleukin-9 and upregulated expression of Eomes and Traf6. Thus, tumor-specific Th9 cells are a more effective CD4+ T cell subset for adoptive cancer therapy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interleukin-9/immunology , Melanoma, Experimental/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/immunology , Immunotherapy, Adoptive/methods , Interleukin-9/genetics , Interleukin-9/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/immunology , TNF Receptor-Associated Factor 6/metabolism , Trans-Activators/genetics , Trans-Activators/immunology , Trans-Activators/metabolism
15.
J Exp Med ; 215(6): 1555-1569, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29743292

ABSTRACT

CD8+ T cells can be polarized into IL-9-secreting (Tc9) cells. We previously showed that adoptive therapy using tumor-specific Tc9 cells generated stronger antitumor responses in mouse melanoma than classical Tc1 cells. To understand why Tc9 cells exert stronger antitumor responses, we used gene profiling to compare Tc9 and Tc1 cells. Tc9 cells expressed different levels of cholesterol synthesis and efflux genes and possessed significantly lower cholesterol content than Tc1 cells. Unique to Tc9, but not other CD8+ or CD4+ T cell subsets, manipulating cholesterol content in polarizing Tc9 cells significantly affected IL-9 expression and Tc9 differentiation and antitumor response in vivo. Mechanistic studies showed that IL-9 was indispensable for Tc9 cell persistence and antitumor effects, and cholesterol or its derivatives inhibited IL-9 expression by activating liver X receptors (LXRs), leading to LXR Sumoylation and reduced p65 binding to Il9 promoter. Our study identifies cholesterol as a critical regulator of Tc9 cell differentiation and function.


Subject(s)
Antineoplastic Agents/pharmacology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cholesterol/pharmacology , Interleukin-9/biosynthesis , Animals , CD8-Positive T-Lymphocytes/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Humans , Liver X Receptors/metabolism , Mice, Inbred C57BL , Oxidation-Reduction , Oxysterols/pharmacology , Sumoylation/drug effects
16.
J Lipid Res ; 59(3): 439-451, 2018 03.
Article in English | MEDLINE | ID: mdl-29298812

ABSTRACT

Cholesterol 25-hydroxylase (CH25H) catalyzes the production of 25-hydroxycholesterol (25-HC), an oxysterol that can play an important role in different biological processes. However, the mechanisms regulating CH25H expression have not been fully elucidated. In this study, we determined that CH25H is highly expressed in mouse liver and peritoneal macrophages. We identified several liver X receptor (LXR) response elements (LXREs) in the human CH25H promoter. In HepG2 cells, activation of LXR by 25-HC or other oxysterols and synthetic ligands [T0901317 (T317) and GW3965] induced CH25H protein expression, which was associated with increased CH25H mRNA expression. 25-HC or T317 activated CH25H transcription in an LXRE-dependent manner. Thus, high-expressing LXRα or LXRß activated CH25H expression, and the activation was further enhanced by LXR ligands. In contrast, inhibition of LXRα/ß expression attenuated 25-HC or T317-induced CH25H expression. Deficiency of interferon γ expression reduced, but did not block, LXR ligand-induced hepatic CH25H expression. Activation of LXR also substantially induced macrophage CH25H expression. In vivo, administration of GW3965 to mice increased CH25H expression in both liver and peritoneal macrophages. Taken together, our study demonstrates that 25-HC can activate CH25H expression in an LXR-dependent manner, which may be an important mechanism to exert the biological actions of 25-HC.


Subject(s)
Hydroxycholesterols/pharmacology , Liver X Receptors/antagonists & inhibitors , Steroid Hydroxylases/biosynthesis , Animals , Dose-Response Relationship, Drug , Gene Expression Profiling , Hep G2 Cells , Humans , Hydroxycholesterols/blood , Interferon-gamma/deficiency , Interferon-gamma/metabolism , Ligands , Lipopolysaccharides/pharmacology , Liver X Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells , Steroid Hydroxylases/metabolism , Toll-Like Receptors/metabolism
17.
Sci Signal ; 10(500)2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29018172

ABSTRACT

Tumor-specific CD4+ T helper 9 (TH9) cells, so-called because of their production of the cytokine interleukin-9 (IL-9), are a powerful effector T cell subset for cancer immunotherapy. We found that pretreatment of naïve CD4+ T cells with IL-7 further enhanced their differentiation into TH9 cells and augmented their antitumor activity. IL-7 markedly increased the abundance of the histone acetyltransferase p300 by activating the STAT5 and PI3K-AKT-mTOR signaling pathways and promoting the acetylation of histones at the Il9 promoter. As a result, the transcriptional regulator Foxo1 was dephosphorylated and translocated to the nucleus, bound to the Il9 promoter, and induced the production of IL-9 protein. In contrast, Foxp1, which bound to the Il9 promoter in naïve CD4+ T cells and inhibited Il9 expression, was outcompeted for binding to the Il9 promoter by Foxo1 and translocated to the cytoplasm. Furthermore, forced expression of Foxo1 or a deficiency in Foxp1 in CD4+ T cells markedly increased the production of IL-9, whereas a deficiency in Foxo1 inhibited the ability of IL-7 to enhance the differentiation and antitumor activity of TH9 cells. Thus, we identified the roles of Foxo1 as a positive regulator and Foxp1 as a negative regulator of TH9 cell differentiation and antitumor activity, which may provide potential targets for cancer immunotherapy.


Subject(s)
Cell Differentiation/drug effects , Forkhead Box Protein O1/physiology , Forkhead Transcription Factors/physiology , Interleukin-7/pharmacology , Repressor Proteins/physiology , T-Lymphocytes, Helper-Inducer/immunology , Acetylation , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/metabolism , Gene Expression Regulation , Histones/metabolism , Humans , Interleukin-9/genetics , Interleukin-9/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lymphocyte Activation , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/metabolism
18.
Nat Commun ; 7: 12368, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27492902

ABSTRACT

Dectin-1 signalling in dendritic cells (DCs) has an important role in triggering protective antifungal Th17 responses. However, whether dectin-1 directs DCs to prime antitumour Th9 cells remains unclear. Here, we show that DCs activated by dectin-1 agonists potently promote naive CD4(+) T cells to differentiate into Th9 cells. Abrogation of dectin-1 in DCs completely abolishes their Th9-polarizing capability in response to dectin-1 agonist curdlan. Notably, dectin-1 stimulation of DCs upregulates TNFSF15 and OX40L, which are essential for dectin-1-activated DC-induced Th9 cell priming. Mechanistically, dectin-1 activates Syk, Raf1 and NF-κB signalling pathways, resulting in increased p50 and RelB nuclear translocation and TNFSF15 and OX40L expression. Furthermore, immunization of tumour-bearing mice with dectin-1-activated DCs induces potent antitumour response that depends on Th9 cells and IL-9 induced by dectin-1-activated DCs in vivo. Our results identify dectin-1-activated DCs as a powerful inducer of Th9 cells and antitumour immunity and may have important clinical implications.


Subject(s)
Dendritic Cells/metabolism , Immunity , Lectins, C-Type/metabolism , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Differentiation/drug effects , Chemokines/metabolism , Cross-Priming/drug effects , Dendritic Cells/drug effects , Humans , Immunity/drug effects , Mice, Inbred C57BL , NF-kappa B/metabolism , OX40 Ligand/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction/drug effects , Syk Kinase/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , beta-Glucans/pharmacology
19.
Sci Rep ; 6: 27295, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27250582

ABSTRACT

We previously reported that LXR ligand, T0901317, inhibited the growth of inoculated Lewis lung carcinoma in C57BL/6 mice by activating IFN-γ production. However, the effects of T0901317 on carcinogen-induced pulmonary carcinomas remain unknown. In this study, we initially conducted a statistical analysis on the data of human lung cancer samples extracted from the TCGA database, and determined that survival rate/time of lung cancer patients and grade of lung adenocarcinoma were positively and negatively related to lung IFN-γ levels, respectively. We then determined the inhibitory effects of T0901317 on mouse pulmonary carcinomas induced by 3-methylcholanthrene (MCA) and butylated hydroxytoluene (BHT) or urethane. We found that T0901317 reduced morbidity and mortality in MCA/BHT-injected BALB/c mice by inhibiting lung adenocarcinoma. T0901317 also protected C57BL/6 mice, but not IFN-γ deficient (IFN-γ(-/-), C57BL/6 background) mice, against MCA/BHT-induced lung hyperplasia/inflammation. In addition, we determined that T0901317 inhibited urethane-induced lung tumors in BABL/c mice. Furthermore, we determined that T0901317 prevented metastasis of 4T1 breast cancer cells in BALB/c mice. Administration of T0901317 substantially increased serum IFN-γ levels and lung IFN-γ expression in BABL/c and C57BL/6 mice. Taken together, our study demonstrates that LXR inhibits MCA/BHT-induced pulmonary carcinomas in BABL/c mice and the inhibition is associated with induction of IFN-γ production.


Subject(s)
Butylated Hydroxytoluene/administration & dosage , Carcinogens/administration & dosage , Carcinoma/prevention & control , Liver X Receptors/metabolism , Lung Neoplasms/prevention & control , Methylcholanthrene/administration & dosage , Animals , Carcinoma/chemically induced , Carcinoma/pathology , Humans , Interferon-gamma/analysis , Interferon-gamma/blood , Lung/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Survival Analysis
20.
J Biol Chem ; 290(36): 21788-99, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26187465

ABSTRACT

The glutathione (GSH)-dependent antioxidant system has been demonstrated to inhibit atherosclerosis. Macrophage CD36 uptakes oxidized low density lipoprotein (oxLDL) thereby facilitating foam cell formation and development of atherosclerosis. It remains unknown if GSH can influence macrophage CD36 expression and cellular oxLDL uptake directly. Herein we report that treatment of macrophages with l-buthionine-S,R-sulfoximine (BSO) decreased cellular GSH production and ratios of GSH to glutathione disulfide (GSH/GSSG) while increasing production of reactive oxygen species. Associated with decreased GSH levels, macrophage CD36 expression was increased, which resulted in enhanced cellular oxLDL uptake. In contrast, N-acetyl cysteine and antioxidant enzyme (catalase or superoxide dismutase) blocked BSO-induced CD36 expression as well as oxLDL uptake. In vivo, administration of mice with BSO increased CD36 expression in peritoneal macrophages and kidneys. BSO had no effect on CD36 mRNA expression and promoter activity but still induced CD36 protein expression in macrophages lacking peroxisome proliferator-activated receptor γ expression, suggesting it induced CD36 expression at the translational level. Indeed, we determined that BSO enhanced CD36 translational efficiency. Taken together, our study demonstrates that cellular GSH levels and GSH/GSSG status can regulate macrophage CD36 expression and cellular oxLDL uptake and demonstrate an important anti-atherogenic function of the GSH-dependent antioxidant system by providing a novel molecular mechanism.


Subject(s)
CD36 Antigens/metabolism , Glutathione/biosynthesis , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Acetylcysteine/pharmacology , Animals , Antimetabolites/pharmacology , Blotting, Western , Buthionine Sulfoximine/pharmacology , CD36 Antigens/genetics , Cell Line , Free Radical Scavengers/pharmacology , Gene Expression/drug effects , Glutathione/antagonists & inhibitors , Glutathione/metabolism , Glutathione Disulfide/metabolism , Lipoproteins, LDL/pharmacokinetics , Macrophages/drug effects , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/genetics , PPAR gamma/metabolism , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , RNA Interference , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...