Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 11(11)2023 11.
Article in English | MEDLINE | ID: mdl-37963637

ABSTRACT

BACKGROUND: The metabolism of tryptophan to kynurenines (KYN) by indoleamine-2,3-dioxygenase or tryptophan-2,3-dioxygenase is a key pathway of constitutive and adaptive tumor immune resistance. The immunosuppressive effects of KYN in the tumor microenvironment are predominantly mediated by the aryl hydrocarbon receptor (AhR), a cytosolic transcription factor that broadly suppresses immune cell function. Inhibition of AhR thus offers an antitumor therapy opportunity via restoration of immune system functions. METHODS: The expression of AhR was evaluated in tissue microarrays of head and neck squamous cell carcinoma (HNSCC), non-small cell lung cancer (NSCLC) and colorectal cancer (CRC). A structure class of inhibitors that block AhR activation by exogenous and endogenous ligands was identified, and further optimized, using a cellular screening cascade. The antagonistic properties of the selected AhR inhibitor candidate BAY 2416964 were determined using transactivation assays. Nuclear translocation, target engagement and the effect of BAY 2416964 on agonist-induced AhR activation were assessed in human and mouse cancer cells. The immunostimulatory properties on gene and cytokine expression were examined in human immune cell subsets. The in vivo efficacy of BAY 2416964 was tested in the syngeneic ovalbumin-expressing B16F10 melanoma model in mice. Coculture of human H1299 NSCLC cells, primary peripheral blood mononuclear cells and fibroblasts mimicking the human stromal-tumor microenvironment was used to assess the effects of AhR inhibition on human immune cells. Furthermore, tumor spheroids cocultured with tumor antigen-specific MART-1 T cells were used to study the antigen-specific cytotoxic T cell responses. The data were analyzed statistically using linear models. RESULTS: AhR expression was observed in tumor cells and tumor-infiltrating immune cells in HNSCC, NSCLC and CRC. BAY 2416964 potently and selectively inhibited AhR activation induced by either exogenous or endogenous AhR ligands. In vitro, BAY 2416964 restored immune cell function in human and mouse cells, and furthermore enhanced antigen-specific cytotoxic T cell responses and killing of tumor spheroids. In vivo, oral application with BAY 2416964 was well tolerated, induced a proinflammatory tumor microenvironment, and demonstrated antitumor efficacy in a syngeneic cancer model in mice. CONCLUSIONS: These findings identify AhR inhibition as a novel therapeutic approach to overcome immune resistance in various types of cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Dioxygenases , Head and Neck Neoplasms , Lung Neoplasms , Humans , Mice , Animals , Tryptophan , Receptors, Aryl Hydrocarbon/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Leukocytes, Mononuclear/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Lung Neoplasms/drug therapy , Kynurenine/metabolism , Immunotherapy , Immunologic Factors , Head and Neck Neoplasms/drug therapy , Tumor Microenvironment
2.
Cancer Res ; 82(8): 1617-1632, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35425959

ABSTRACT

An altered lipidome in tumors may affect not only tumor cells themselves but also their microenvironment. In this study, a lipidomics screen reveals increased amounts of phosphatidylserine (PS), particularly ether-PS (ePS), in murine mammary tumors compared with normal tissue. PS was produced by phosphatidylserine synthase 1 (PTDSS1), and depletion of Ptdss1 from tumor cells in mice reduced ePS levels accompanied by stunted tumor growth and decreased tumor-associated macrophage (TAM) abundance. Ptdss1-deficient tumor cells exposed less PS during apoptosis, which was recognized by the PS receptor MERTK. Mammary tumors in macrophage-specific Mertk-/- mice showed similarly suppressed growth and reduced TAM infiltration. Transcriptomic profiles of TAMs from Ptdss1-knockdown tumors and Mertk-/- TAMs revealed that macrophage proliferation was reduced when the Ptdss1/Mertk pathway was targeted. Moreover, PTDSS1 expression correlated positively with TAM abundance but negatively with breast carcinoma patient survival. PTDSS1 thus may be a target to modify tumor-promoting inflammation. SIGNIFICANCE: This study shows that inhibiting the production of ether-phosphatidylserine by targeting phosphatidylserine synthase PTDSS1 limits tumor-associated macrophage expansion and breast tumor growth.


Subject(s)
Lipidomics , Neoplasms , Animals , CDPdiacylglycerol-Serine O-Phosphatidyltransferase , Ether , Humans , Inflammation/metabolism , Mice , Neoplasms/metabolism , Phosphatidylserines/metabolism , Tumor Microenvironment , c-Mer Tyrosine Kinase/metabolism
3.
Front Immunol ; 13: 827719, 2022.
Article in English | MEDLINE | ID: mdl-35145525

ABSTRACT

The lung tumor microenvironment plays a critical role in the tumorigenesis and metastasis of lung cancer, resulting from the crosstalk between cancer cells and microenvironmental cells. Therefore, comprehensive identification and characterization of cell populations in the complex lung structure is crucial for development of novel targeted anti-cancer therapies. Here, a hierarchical clustering approach with multispectral flow cytometry was established to delineate the cellular landscape of murine lungs under steady-state and cancer conditions. Fluorochromes were used multiple times to be able to measure 24 cell surface markers with only 13 detectors, yielding a broad picture for whole-lung phenotyping. Primary and metastatic murine lung tumor models were included to detect major cell populations in the lung, and to identify alterations to the distribution patterns in these models. In the primary tumor models, major altered populations included CD324+ epithelial cells, alveolar macrophages, dendritic cells, and blood and lymph endothelial cells. The number of fibroblasts, vascular smooth muscle cells, monocytes (Ly6C+ and Ly6C-) and neutrophils were elevated in metastatic models of lung cancer. Thus, the proposed clustering approach is a promising method to resolve cell populations from complex organs in detail even with basic flow cytometers.


Subject(s)
Flow Cytometry/methods , Fluorescent Dyes/chemistry , Lung Neoplasms/pathology , Staining and Labeling/methods , Animals , Antigens, Ly/genetics , Cell Line, Tumor , Dendritic Cells/cytology , Dendritic Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry/instrumentation , Genetic Heterogeneity , Humans , Macrophages, Alveolar/cytology , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Primary Cell Culture , Tumor Microenvironment
4.
Int J Mol Sci ; 23(3)2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35163211

ABSTRACT

The sphingolipid sphingosine-1-phosphate (S1P) promotes tumor development through a variety of mechanisms including promoting proliferation, survival, and migration of cancer cells. Moreover, S1P emerged as an important regulator of tumor microenvironmental cell function by modulating, among other mechanisms, tumor angiogenesis. Therefore, S1P was proposed as a target for anti-tumor therapy. The clinical success of current cancer immunotherapy suggests that future anti-tumor therapy needs to consider its impact on the tumor-associated immune system. Hereby, S1P may have divergent effects. On the one hand, S1P gradients control leukocyte trafficking throughout the body, which is clinically exploited to suppress auto-immune reactions. On the other hand, S1P promotes pro-tumor activation of a diverse range of immune cells. In this review, we summarize the current literature describing the role of S1P in tumor-associated immunity, and we discuss strategies for how to target S1P for anti-tumor therapy without causing immune paralysis.


Subject(s)
Lysophospholipids/immunology , Lysophospholipids/metabolism , Neoplasms/immunology , Sphingosine/analogs & derivatives , Animals , Humans , Immune System/metabolism , Inflammation/immunology , Neoplasms/metabolism , Neovascularization, Pathologic/immunology , Signal Transduction/physiology , Sphingolipids/metabolism , Sphingolipids/physiology , Sphingosine/immunology , Sphingosine/metabolism , Tumor Microenvironment/physiology
5.
Cell Mol Life Sci ; 78(21-22): 7025-7041, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34626204

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most difficult cancer types to treat. Liver cancer is often diagnosed at late stages and therapeutic treatment is frequently accompanied by development of multidrug resistance. This leads to poor outcomes for cancer patients. Understanding the fundamental molecular mechanisms leading to liver cancer development is crucial for developing new therapeutic approaches, which are more efficient in treating cancer. Mice with a liver specific UDP-glucose ceramide glucosyltransferase (UGCG) knockout (KO) show delayed diethylnitrosamine (DEN)-induced liver tumor growth. Accordingly, the rationale for our study was to determine whether UGCG overexpression is sufficient to drive cancer phenotypes in liver cells. We investigated the effect of UGCG overexpression (OE) on normal murine liver (NMuLi) cells. Increased UGCG expression results in decreased mitochondrial respiration and glycolysis, which is reversible by treatment with EtDO-P4, an UGCG inhibitor. Furthermore, tumor markers such as FGF21 and EPCAM are lowered following UGCG OE, which could be related to glucosylceramide (GlcCer) and lactosylceramide (LacCer) accumulation in glycosphingolipid-enriched microdomains (GEMs) and subsequently altered signaling protein phosphorylation. These cellular processes lead to decreased proliferation in NMuLi/UGCG OE cells. Our data show that increased UGCG expression itself does not induce pro-cancerous processes in normal liver cells, which indicates that increased GlcCer expression leads to different outcomes in different cancer types.


Subject(s)
Biomarkers, Tumor/metabolism , Energy Metabolism/physiology , Glucosylceramides/metabolism , Liver/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line , Drug Resistance, Multiple/physiology , Glucosyltransferases/metabolism , Glycolysis/physiology , Glycosphingolipids/metabolism , Liver Neoplasms/metabolism , Mice , Mitochondria/metabolism , Signal Transduction/physiology
6.
Front Oncol ; 10: 1771, 2020.
Article in English | MEDLINE | ID: mdl-33014872

ABSTRACT

Despite the success of immune checkpoint blockade in cancer, the number of patients that benefit from this revolutionary treatment option remains low. Therefore, efforts are being undertaken to sensitize tumors for immune checkpoint blockade, which includes combining immune checkpoint blocking agents such as anti-PD-1 antibodies with standard of care treatments. Here we report that a combination of chemotherapy (doxorubicin) and immune checkpoint blockade (anti-PD-1 antibodies) induces superior tumor control compared to chemotherapy and immune checkpoint blockade alone in the murine autochthonous polyoma middle T oncogene-driven (PyMT) mammary tumor model. Using whole transcriptome analysis, we identified a set of genes that were upregulated specifically upon chemoimmunotherapy. This gene signature and, more specifically, a condensed four-gene signature predicted favorable survival of human mammary carcinoma patients in the METABRIC cohort. Moreover, PyMT tumors treated with chemoimmunotherapy contained higher levels of cytotoxic lymphocytes, particularly natural killer cells (NK cells). Gene set enrichment analysis and bead-based ELISA measurements revealed increased IL-27 production and signaling in PyMT tumors upon chemoimmunotherapy. Moreover, IL-27 signaling improved NK cell cytotoxicity against PyMT cells in vitro. Taken together, our data support recent clinical observations indicating a benefit of chemoimmunotherapy compared to monotherapy in breast cancer and suggest potential underlying mechanisms.

7.
J Clin Invest ; 130(10): 5461-5476, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32663191

ABSTRACT

Tumor immunosuppression is a limiting factor for successful cancer therapy. The lipid sphingosine-1-phosphate (S1P), which signals through 5 distinct G protein-coupled receptors (S1PR1-5), has emerged as an important regulator of carcinogenesis. However, the utility of targeting S1P in tumors is hindered by S1P's impact on immune cell trafficking. Here, we report that ablation of the immune cell-specific receptor S1PR4, which plays a minor role in immune cell trafficking, delayed tumor development and improved therapy success in murine models of mammary and colitis-associated colorectal cancer through increased CD8+ T cell abundance. Transcriptome analysis revealed that S1PR4 affected proliferation and survival of CD8+ T cells in a cell-intrinsic manner via the expression of Pik3ap1 and Lta4h. Accordingly, PIK3AP1 expression was connected to increased CD8+ T cell proliferation and clinical parameters in human breast and colon cancer. Our data indicate a so-far-unappreciated tumor-promoting role of S1P by restricting CD8+ T cell expansion via S1PR4.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Mammary Neoplasms, Experimental/therapy , Sphingosine-1-Phosphate Receptors/deficiency , Sphingosine-1-Phosphate Receptors/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation/genetics , Cell Survival/genetics , Cell Survival/immunology , Colitis/complications , Colitis/immunology , Colitis/pathology , Colonic Neoplasms/etiology , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Disease Progression , Female , Gene Expression Profiling , Gene Knockout Techniques , Humans , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Sphingosine-1-Phosphate Receptors/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
8.
Mol Carcinog ; 58(11): 2127-2138, 2019 11.
Article in English | MEDLINE | ID: mdl-31436357

ABSTRACT

In solid tumors, tumor-associated macrophages (TAMs) commonly accumulate within hypoxic areas. Adaptations to such environments evoke transcriptional changes by the hypoxia-inducible factors (HIFs). While HIF-1α is ubiquitously expressed, HIF-2α appears tissue-specific with consequences of HIF-2α expression in TAMs only being poorly characterized. An E0771 allograft breast tumor model revealed faster tumor growth in myeloid HIF-2α knockout (HIF-2αLysM-/- ) compared with wildtype (wt) mice. In an RNA-sequencing approach of FACS sorted wt and HIF-2α LysM-/- TAMs, serine protease inhibitor, Kunitz type-1 ( Spint1) emerged as a promising candidate for HIF-2α-dependent regulation. We validated reduced Spint1 messenger RNA expression and concomitant Spint1 protein secretion under hypoxia in HIF-2α-deficient bone marrow-derived macrophages (BMDMs) compared with wt BMDMs. In line with the physiological function of Spint1 as an inhibitor of hepatocyte growth factor (HGF) activation, supernatants of hypoxic HIF-2α knockout BMDMs, not containing Spint1, were able to release proliferative properties of inactive pro-HGF on breast tumor cells. In contrast, hypoxic wt BMDM supernatants containing abundant Spint1 amounts failed to do so. We propose that Spint1 contributes to the tumor-suppressive function of HIF-2α in TAMs in breast tumor development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Neoplasms/genetics , Proteinase Inhibitory Proteins, Secretory/genetics , Tumor Microenvironment/genetics , Allografts , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Hepatocyte Growth Factor/genetics , Humans , Macrophages/metabolism , Macrophages/pathology , Membrane Glycoproteins/genetics , Mice , Neoplasms/pathology , RNA, Messenger
9.
Front Immunol ; 10: 1706, 2019.
Article in English | MEDLINE | ID: mdl-31379883

ABSTRACT

The sphingolipid sphingosine-1-phosphate (S1P) is produced by sphingosine kinases to either signal through intracellular targets or to activate a family of specific G-protein-coupled receptors (S1PR). S1P levels are usually low in peripheral tissues compared to the vasculature, forming a gradient that mediates lymphocyte trafficking. However, S1P levels rise during inflammation in peripheral tissues, thereby affecting resident or recruited immune cells, including macrophages. As macrophages orchestrate initiation and resolution of inflammation, the sphingosine kinase/S1P/S1P-receptor axis emerges as an important determinant of macrophage function in the pathogenesis of inflammatory diseases such as cancer, atherosclerosis, and infection. In this review, we therefore summarize the current knowledge how S1P affects macrophage biology.


Subject(s)
Lysophospholipids/immunology , Macrophages/immunology , Sphingosine/analogs & derivatives , Animals , Humans , Phagocytosis , Sphingosine/immunology , Sphingosine-1-Phosphate Receptors/immunology
10.
Int J Biochem Cell Biol ; 112: 95-106, 2019 07.
Article in English | MEDLINE | ID: mdl-31082617

ABSTRACT

The G protein-coupled estrogen receptor 1 (GPER1) is involved in the regulation of physiological processes such as cellular growth and proliferation, but also in pathophysiological processes such as tumor development. The role of GPER1 in breast cancer is contradictory. Therefore, we investigated the influence of GPER1 overexpression on cellular processes in MCF-7 breast cancer cells. GPER1 overexpression leads to a cell cycle arrest in the G1 phase, induction of autophagy and reduced proliferation. Reduced proliferation was accompanied by a reduced basal respiration and reduced glycolysis rate in GPER1 overexpressing cells. This is presumably ascribable to mitophagy induction following GPER1 overexpression. However, GPER1 overexpressing cells were less sensitive against doxorubicin as compared to control cells. In previous work we showed the effect of transient GPER1 overexpression on the synthesis of several ceramide synthases (CerS) thereby influencing the sphingolipid pathway. Therefore, we investigated CerS expression and sphingolipid level in stable GPER1 overexpressing and control cells. Stable GPER1 overexpression strongly reduced CerS4, CerS5 and CerS6 promoter activity and CerS5 and CerS6 mRNA expression, whereas CerS2 mRNA expression was upregulated. The GPER1 effect on CerS5 promoter is mediated by GSK-3ß signaling. In addition, other enzymes of the sphingolipid pathway were upregulated. Our study provides new insights into the role of GPER1 and the activated sphingolipid pathways and how GPER1 may influence cellular processes such as cancer cell survival following chemotherapy. Further studies are needed to investigate the molecular mechanisms leading to these cellular effects. Finding new therapeutic targets for modulating specifically GPER1 in breast tumors may improve endocrine breast cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Ceramides/biosynthesis , Cytostatic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Homeostasis/drug effects , Neoplasm Proteins/metabolism , Oxidoreductases/metabolism , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Ceramides/genetics , Female , G1 Phase Cell Cycle Checkpoints , Humans , MCF-7 Cells , Neoplasm Proteins/genetics , Oxidoreductases/genetics , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics
11.
Cells ; 8(2)2019 02 18.
Article in English | MEDLINE | ID: mdl-30781698

ABSTRACT

Hypoxia poses a stress to cells and decreases mitochondrial respiration, in part by electron transport chain (ETC) complex reorganization. While metabolism under acute hypoxia is well characterized, alterations under chronic hypoxia largely remain unexplored. We followed oxygen consumption rates in THP-1 monocytes during acute (16 h) and chronic (72 h) hypoxia, compared to normoxia, to analyze the electron flows associated with glycolysis, glutamine, and fatty acid oxidation. Oxygen consumption under acute hypoxia predominantly demanded pyruvate, while under chronic hypoxia, fatty acid- and glutamine-oxidation dominated. Chronic hypoxia also elevated electron-transferring flavoproteins (ETF), and the knockdown of ETF⁻ubiquinone oxidoreductase lowered mitochondrial respiration under chronic hypoxia. Metabolomics revealed an increase in citrate under chronic hypoxia, which implied glutamine processing to α-ketoglutarate and citrate. Expression regulation of enzymes involved in this metabolic shunting corroborated this assumption. Moreover, the expression of acetyl-CoA carboxylase 1 increased, thus pointing to fatty acid synthesis under chronic hypoxia. Cells lacking complex I, which experienced a markedly impaired respiration under normoxia, also shifted their metabolism to fatty acid-dependent synthesis and usage. Taken together, we provide evidence that chronic hypoxia fuels the ETC via ETFs, increasing fatty acid production and consumption via the glutamine-citrate-fatty acid axis.


Subject(s)
Electron-Transferring Flavoproteins/metabolism , Cell Hypoxia , Cell Respiration , Electron Transport , Electron Transport Complex I/metabolism , Fatty Acids/metabolism , Glutamine/metabolism , Humans , Iron-Sulfur Proteins/metabolism , Membrane Potential, Mitochondrial , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mitochondria/metabolism , Models, Biological , Oxidation-Reduction , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Oxygen Consumption , Phenotype , THP-1 Cells
12.
Front Immunol ; 9: 973, 2018.
Article in English | MEDLINE | ID: mdl-29867954

ABSTRACT

Aim: Reactive oxygen species (ROS) produced by enzymes of the NADPH oxidase family serve as second messengers for cellular signaling. Processes such as differentiation and proliferation are regulated by NADPH oxidases. In the intestine, due to the exceedingly fast and constant renewal of the epithelium both processes have to be highly controlled and balanced. Nox1 is the major NADPH oxidase expressed in the gut, and its function is regulated by cytosolic subunits such as NoxO1. We hypothesize that the NoxO1-controlled activity of Nox1 contributes to a proper epithelial homeostasis and renewal in the gut. Results: NoxO1 is highly expressed in the colon. Knockout of NoxO1 reduces the production of superoxide in colon crypts and is not subsidized by an elevated expression of its homolog p47phox. Knockout of NoxO1 increases the proliferative capacity and prevents apoptosis of colon epithelial cells. In mouse models of dextran sulfate sodium (DSS)-induced colitis and azoxymethane/DSS induced colon cancer, NoxO1 has a protective role and may influence the population of natural killer cells. Conclusion: NoxO1 affects colon epithelium homeostasis and prevents inflammation.


Subject(s)
Cell Proliferation , Colon/cytology , Epithelial Cells/cytology , Homeostasis , Proteins/genetics , Reactive Oxygen Species/metabolism , Adaptor Proteins, Signal Transducing , Animals , Apoptosis , Colitis/chemically induced , Colonic Neoplasms/chemically induced , Disease Models, Animal , Inflammation , Mice , Mice, Knockout , NADPH Oxidase 1/genetics , NADPH Oxidase 1/immunology , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Proteins/immunology
13.
Mediators Inflamm ; 2017: 6059203, 2017.
Article in English | MEDLINE | ID: mdl-28848247

ABSTRACT

The sphingolipid sphingosine-1-phosphate (S1P) emerges as an important regulator of immunity, mainly by signaling through a family of five specific G protein-coupled receptors (S1PR1-5). While S1P signaling generally has the potential to affect not only trafficking but also differentiation, activation, and survival of a diverse range of immune cells, the specific outcome depends on the S1P receptor repertoire expressed on a given cell. Among the S1PRs, S1PR4 is specifically abundant in immune cells, suggesting a major role of the S1P/S1PR4 axis in immunity. Recent studies indeed highlight its role in activation of immune cells, differentiation, and, potentially, trafficking. In this review, we summarize the emerging data that support a major role of S1PR4 in modulating immunity in humans and mice and discuss therapeutic implications.


Subject(s)
Cell Movement/physiology , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Cell Differentiation/physiology , Cell Movement/genetics , Humans , Signal Transduction/physiology , Sphingosine/metabolism
14.
J Exp Med ; 214(9): 2695-2713, 2017 Sep 04.
Article in English | MEDLINE | ID: mdl-28739604

ABSTRACT

Metastasis is the primary cause of cancer death. The inflammatory tumor microenvironment contributes to metastasis, for instance, by recruiting blood and lymph vessels. Among tumor-infiltrating immune cells, tumor-associated macrophages (TAMs) take a center stage in promoting both tumor angiogenesis and metastatic spread. We found that genetic deletion of the S1P receptor 1 (S1pr1) alone in CD11bhi CD206+ TAMs infiltrating mouse breast tumors prevents pulmonary metastasis and tumor lymphangiogenesis. Reduced lymphangiogenesis was also observed in the nonrelated methylcholanthrene-induced fibrosarcoma model. Transcriptome analysis of isolated TAMs from both entities revealed reduced expression of the inflammasome component Nlrp3 in S1PR1-deficient TAMs. Macrophage-dependent lymphangiogenesis in vitro was triggered upon inflammasome activation and required both S1PR1 signaling and IL-1ß production. Finally, NLRP3 expression in tumor-infiltrating macrophages correlated with survival, lymph node invasion, and metastasis of mammary carcinoma patients. Conceptually, our study indicates an unappreciated role of the NLRP3 inflammasome in promoting metastasis via the lymphatics downstream of S1PR1 signaling in macrophages.


Subject(s)
Interleukin-1beta/physiology , Lymphangiogenesis/physiology , Macrophages/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Neoplasm Metastasis/physiopathology , Receptors, Lysosphingolipid/physiology , Animals , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Female , Fibrosarcoma/physiopathology , Humans , Lymphatic Metastasis , Mammary Neoplasms, Experimental/physiopathology , Mice , Mice, Inbred C57BL , Sphingosine-1-Phosphate Receptors
15.
J Immunol ; 196(4): 1579-90, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26783340

ABSTRACT

Plasmacytoid dendritic cells (pDCs) produce large amounts of type I IFN in response to TLR7/9 ligands. This conveys antiviral effects, activates other immune cells (NK cells, conventional DCs, B, and T cells), and causes the induction and expansion of a strong inflammatory response. pDCs are key players in various type I IFN-driven autoimmune diseases such as systemic lupus erythematosus or psoriasis, but pDCs are also involved in (anti-)tumor immunity. The sphingolipid sphingosine-1-phosphate (S1P) signals through five G-protein-coupled receptors (S1PR1-5) to regulate, among other activities, immune cell migration and activation. The present study shows that S1P stimulation of human, primary pDCs substantially decreases IFN-α production after TLR7/9 activation with different types of CpG oligodeoxynucleotides or tick-borne encephalitis vaccine, which occurred in an S1PR4-dependent manner. Mechanistically, S1PR4 activation preserves the surface expression of the human pDC-specific inhibitory receptor Ig-like transcript 7. We provide novel information that Ig-like transcript 7 is rapidly internalized upon receptor-mediated endocytosis of TLR7/9 ligands to allow high IFN-α production. This is antagonized by S1PR4 signaling, thus decreasing TLR-induced IFN-α secretion. At a functional level, attenuated IFN-α production failed to alter Ag-driven T cell proliferation in pDC-dependent T cell activation assays, but shifted cytokine production of T cells from a Th1 (IFN-γ) to a regulatory (IL-10) profile. In conclusion, S1PR4 agonists block human pDC activation and may therefore be a promising tool to restrict pathogenic IFN-α production.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Interferon-alpha/biosynthesis , Receptors, Immunologic/physiology , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Cell Differentiation , Cell Movement , Cells, Cultured , Cytokines/biosynthesis , Dendritic Cells/drug effects , Humans , Interferon-alpha/immunology , Interleukin-10/biosynthesis , Lymphocyte Activation , Lysophospholipids/immunology , Lysophospholipids/pharmacology , Oligodeoxyribonucleotides/pharmacology , Receptors, Immunologic/immunology , Sphingosine/analogs & derivatives , Sphingosine/immunology , Sphingosine/pharmacology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/immunology
16.
PLoS One ; 10(9): e0139073, 2015.
Article in English | MEDLINE | ID: mdl-26413839

ABSTRACT

Recent studies indicate that the abnormal microenvironment of tumors may play a critical role in carcinogenesis, including lung cancer. We comprehensively assessed the number of stromal cells, especially immune/inflammatory cells, in lung cancer and evaluated their infiltration in cancers of different stages, types and metastatic characteristics potential. Immunohistochemical analysis of lung cancer tissue arrays containing normal and lung cancer sections was performed. This analysis was combined with cyto-/histomorphological assessment and quantification of cells to classify/subclassify tumors accurately and to perform a high throughput analysis of stromal cell composition in different types of lung cancer. In human lung cancer sections we observed a significant elevation/infiltration of total-T lymphocytes (CD3+), cytotoxic-T cells (CD8+), T-helper cells (CD4+), B cells (CD20+), macrophages (CD68+), mast cells (CD117+), mononuclear cells (CD11c+), plasma cells, activated-T cells (MUM1+), B cells, myeloid cells (PD1+) and neutrophilic granulocytes (myeloperoxidase+) compared with healthy donor specimens. We observed all of these immune cell markers in different types of lung cancers including squamous cell carcinoma, adenocarcinoma, adenosquamous cell carcinoma, small cell carcinoma, papillary adenocarcinoma, metastatic adenocarcinoma, and bronchioloalveolar carcinoma. The numbers of all tumor-associated immune cells (except MUM1+ cells) in stage III cancer specimens was significantly greater than those in stage I samples. We observed substantial stage-dependent immune cell infiltration in human lung tumors suggesting that the tumor microenvironment plays a critical role during lung carcinogenesis. Strategies for therapeutic interference with lung cancer microenvironment should consider the complexity of its immune cell composition.


Subject(s)
Inflammation/immunology , Inflammation/pathology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Immunohistochemistry , Peroxidase/metabolism , Stromal Cells/pathology , Tumor Microenvironment
17.
Oncotarget ; 6(12): 10284-96, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25871398

ABSTRACT

Prostaglandin E2 (PGE2) favors multiple aspects of tumor development and immune evasion. Therefore, microsomal prostaglandin E synthase (mPGES-1/-2), is a potential target for cancer therapy. We explored whether inhibiting mPGES-1 in human and mouse models of breast cancer affects tumor-associated immunity. A new model of breast tumor spheroid killing by human PBMCs was developed. In this model, tumor killing required CD80 expression by tumor-associated phagocytes to trigger cytotoxic T cell activation. Pharmacological mPGES-1 inhibition increased CD80 expression, whereas addition of PGE2, a prostaglandin E2 receptor 2 (EP2) agonist, or activation of signaling downstream of EP2 reduced CD80 expression. Genetic ablation of mPGES-1 resulted in markedly reduced tumor growth in PyMT mice. Macrophages of mPGES-1(-/-) PyMT mice indeed expressed elevated levels of CD80 compared to their wildtype counterparts. CD80 expression in tumor-spheroid infiltrating mPGES-1(-/-) macrophages translated into antigen-specific cytotoxic T cell activation. In conclusion, mPGES-1 inhibition elevates CD80 expression by tumor-associated phagocytes to restrict tumor growth. We propose that mPGES-1 inhibition in combination with immune cell activation might be part of a therapeutic strategy to overcome the immunosuppressive tumor microenvironment.


Subject(s)
B7-1 Antigen/antagonists & inhibitors , Breast Neoplasms/metabolism , Intramolecular Oxidoreductases/antagonists & inhibitors , Macrophages/metabolism , Animals , B7-1 Antigen/biosynthesis , B7-1 Antigen/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Dinoprostone/metabolism , Female , Humans , Intramolecular Oxidoreductases/metabolism , MCF-7 Cells , Macrophages/immunology , Mice , Mice, Knockout , Prostaglandin-E Synthases , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Microenvironment
18.
Biol Chem ; 396(6-7): 775-82, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25720060

ABSTRACT

The sphingolipid sphingosine-1-phosphate (S1P) has various functions in immune cell biology, regulating survival, proliferation, and, most prominently, migration. S1P couples to five G protein-coupled receptors (S1PR1-5) to transduce its effects on immune cell function. Expression of S1PR4 is restricted to immune cells. However, its impact on immune cell biology is largely elusive. In the current study, we intended to answer the question of whether S1P might affect plasmacytoid dendritic cell (pDC) migration, which dominantly express S1PR4. pDC are highly specialized cells producing large amounts of type I interferon in response to TLR7/9 ligands after viral infection or during autoimmunity. Surprisingly, we noticed a reduced abundance of pDC, particularly CD4- pDC, in all organs of S1PR4-deficient vs. wildtype mice. This effect was not caused by altered migration of mature pDC, but rather a reduced potential of pDC progenitors, especially common DC progenitors, to differentiate into pDCs. In vitro studies suggested that reduced S1PR4-deficient pDC progenitor differentiation into mature pDC might be explained by both migration and differentiation of pDC progenitors in the bone marrow. As S1PR4 also affected the differentiation of CD34+ human hematopoietic stem cells into pDC, interfering with S1PR4 might be useful to reduce pDC numbers during autoimmunity.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/metabolism , Receptors, Lysosphingolipid/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Cell Movement/physiology , Cells, Cultured , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Receptors, Lysosphingolipid/genetics
19.
Int J Cancer ; 133(7): 1578-88, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23536473

ABSTRACT

Cyclooxygenase (COX)-2-derived prostaglandin E2 (PGE2 ) supports the growth of a spectrum of cancers. The potential benefit of COX-2-inhibiting non-steroidal anti-inflammatory drugs (NSAIDs) for cancer treatment is however limited by their well-known cardiovascular side-effects. Therefore, targeting microsomal PGE synthase 1 (mPGES-1), the downstream enzyme in the COX-2-dependent pathway of PGE2 production might be attractive, although conflicting data regarding a potential tumor-supporting function of mPGES-1 were reported. We determined the impact of mPGES-1 in human DU145 prostate cancer cell growth. Surprisingly, knockdown of mPGES-1 did not alter growth of DU145 monolayer cells, but efficiently inhibited the growth of DU145 multicellular tumor spheroids (MCTS). Opposed to MCTS, monolayer cells did not secrete PGE2 due to a lack of COX-2 expression, which was induced during spheroid formation. Pharmacological inhibition of COX-2 and mPGES-1 supported the crucial role of PGE2 for growth of MCTS. The functionality of spheroid-derived PGE2 was demonstrated by its ability to inhibit cytotoxic T cell activation. When investigating mechanisms of spheroid-induced COX-2 induction, we observed that among microenvironmental factors neither glucose deprivation, hypoxia nor tumor cell apoptosis enhanced COX-2 expression. Interestingly, interfering with apoptosis in spheroids triggered a shift towards necrosis, thus augmenting COX-2 expression. We went on to demonstrate that necrotic cells induced COX-2 mRNA expression and PGE2 secretion from live tumor cells. In conclusion, necrosis-dependent COX-2 upregulation in MCTS promoted PGE2 -dependent tumor growth and inhibited activated cytotoxic T cells. Hence, blocking mPGES-1 as a therapeutic option may be considered for COX-2/mPGES-1-positive solid cancers.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Spheroids, Cellular/pathology , Cell Line, Tumor , Cell Proliferation , Humans , Intramolecular Oxidoreductases/genetics , Lymphocyte Activation , Male , Necrosis , Prostaglandin-E Synthases , Prostatic Neoplasms/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spheroids, Cellular/metabolism , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...