Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Oncoimmunology ; 7(3): e1408751, 2018.
Article in English | MEDLINE | ID: mdl-29399416

ABSTRACT

While the importance of iron for tumor development is widely appreciated, the exact sources of tumor-supporting iron largely remain elusive. The possibility that iron might be provided by stromal cells in the tumor microenvironment was not taken into account so far. In the present study, we show that tumor-associated macrophages (TAM) acquire an iron-release phenotype upon their interaction with tumor cells, thereby increasing the availability of iron in the tumor microenvironment. Mechanistically, TAM expressed elevated levels of the high-affinity iron-binding protein lipocalin-2 (LCN-2), which appeared to be critical for the export of iron from TAM, and in turn enhanced tumor cell proliferation. Moreover, in PyMT-mouse tumors as well as in primary human breast tumors LCN-2 was predominantly expressed in the tumor stroma as compared to tumor cells. LCN-2 expression in the stroma further correlated with enhanced tumor proliferation in vivo. Our data suggest a dominant role of TAM in the tumor iron-management and identify LCN-2 as a critical iron transporter in this context. Targeting the LCN-2 iron export mechanism selectively in stromal cells might open for future iron-targeted tumor therapeutic approaches.

2.
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
3.
Sci Signal ; 9(434): ra64, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27353364

ABSTRACT

Tumor cell-derived factors skew macrophages toward a tumor-supporting phenotype associated with the secretion of protumorigenic mediators. Apoptosing tumor cells release sphingosine 1-phosphate (S1P), which stimulates the production of lipocalin 2 (LCN2) in tumor-associated macrophages and is associated with tumor metastasis. We explored the mechanism by which S1P induces LCN2 in macrophages and investigated how this contributed to tumor growth and metastasis. Knockdown of S1P receptor 1 (S1PR1) in primary human macrophages and experiments with bone marrow-derived macrophages from S1PR1-deficient mice showed that S1P signaled through S1PR1 to induce LCN2 expression. The LCN2 promoter contains a consensus sequence for signal transducer and activator of transcription 3 (STAT3), and deletion of the STAT3 recognition sequence reduced expression of an LCN2-controlled reporter gene. Conditioned medium from coculture experiments indicated that the release of LCN2 from macrophages induced tube formation and proliferation in cultures of primary human lymphatic endothelial cells in a manner dependent on the kinase PI3K and subsequent induction of the growth factor VEGFC, which functioned as an autocrine signal stimulating the receptor VEGFR3. Knockout of Lcn2 attenuated tumor-associated lymphangiogenesis and breast tumor metastasis both in the breast cancer model MMTV-PyMT mice and in mice bearing orthotopic wild-type tumors. Our findings indicate that macrophages respond to dying tumor cells by producing signals that promote lymphangiogenesis, which enables metastasis.


Subject(s)
Breast Neoplasms/metabolism , Lipocalin-2/metabolism , Lymphangiogenesis , Macrophages/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Lipocalin-2/genetics , Lysophospholipids , MCF-7 Cells , Macrophages/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Sphingosine/analogs & derivatives
4.
J Pathol ; 239(3): 274-85, 2016 07.
Article in English | MEDLINE | ID: mdl-27038000

ABSTRACT

Tumour cell-secreted factors skew infiltrating immune cells towards a tumour-supporting phenotype, expressing pro-tumourigenic mediators. However, the influence of lipocalin-2 (Lcn2) on the metastatic cascade in the tumour micro-environment is still not clearly defined. Here, we explored the role of stroma-derived, especially macrophage-released, Lcn2 in breast cancer progression. Knockdown studies and neutralizing antibody approaches showed that Lcn2 contributes to the early events of metastasis in vitro. The release of Lcn2 from macrophages induced an epithelial-mesenchymal transition programme in MCF-7 breast cancer cells and enhanced local migration as well as invasion into the extracellular matrix, using a three-dimensioanl (3D) spheroid model. Moreover, a global Lcn2 deficiency attenuated breast cancer metastasis in both the MMTV-PyMT breast cancer model and a xenograft model inoculating MCF-7 cells pretreated with supernatants from wild-type and Lcn2-knockdown macrophages. To dissect the role of stroma-derived Lcn2, we employed an orthotopic mammary tumour mouse model. Implantation of wild-type PyMT tumour cells into Lcn2-deficient mice left primary mammary tumour formation unaltered, but specifically reduced tumour cell dissemination into the lung. We conclude that stroma-secreted Lcn2 promotes metastasis in vitro and in vivo, thereby contributing to tumour progression. Our study highlights the tumourigenic potential of stroma-released Lcn2 and suggests Lcn2 as a putative therapeutic target. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Breast Neoplasms/genetics , Lipocalin-2/metabolism , Lung Neoplasms/secondary , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Transformation, Neoplastic , Disease Progression , Epithelial-Mesenchymal Transition , Female , Humans , Lipocalin-2/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , RNA, Small Interfering , Stromal Cells/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
5.
Biol Chem ; 394(6): 773-81, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23399635

ABSTRACT

Betulinic acid (BA) exhibits antitumoral activity by blocking proliferation, invasion, and angiogenesis. However, the impact of BA on epithelial-to-mesenchymal transition (EMT), a hallmark of cancer metastasis induced among others by neutrophil gelatinase-associated lipocalin (NGAL), remains unknown. The present study aimed at determining the effect of BA on NGAL-induced EMT. In A375 melanoma cells, BA downregulated mesenchymal markers, increased epithelial markers, and inhibited cytoskeletal reorganization. In addition, BA limited endogenous NGAL production and further suppressed EMT induced by exogenously added NGAL and the corresponding invasive cellular phenotype. In conclusion, BA interferes with EMT-associated changes, a mechanism to antagonize invasive melanoma cells.


Subject(s)
Acute-Phase Proteins/metabolism , Epithelial-Mesenchymal Transition/drug effects , Lipocalins/metabolism , Melanoma/pathology , Proto-Oncogene Proteins/metabolism , Triterpenes/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Lipocalin-2 , Neoplasm Invasiveness , Pentacyclic Triterpenes , Phenotype , Triterpenes/chemistry , Betulinic Acid
6.
Mol Cell Biol ; 32(19): 3938-48, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22851691

ABSTRACT

Tumor cell-derived factors, such as interleukin 10 (IL-10), polarize macrophages toward a regulatory M2 phenotype, characterized by the expression of anti-inflammatory cytokines and protumorigenic mediators. Here we explored molecular mechanisms allowing IL-10 to upregulate the protumorigenic protein NGAL in primary human macrophages. Reporter assays of full-length or deletion constructs of the NGAL promoter provided evidence that NGAL production is STAT3 dependent, activated downstream of the IL-10-Janus kinase (Jak) axis, as well as being C/EBPß dependent. The involvement of STAT3 and C/EBPß was shown by chromatin immunoprecipitation (ChIP) and ChIP-Western analysis, as well as decoy oligonucleotides scavenging both STAT3 and C/EBPß in human macrophages. Furthermore, the production of NGAL in macrophages in response to IL-10 induces cellular growth and proliferation of MCF-7 breast cancer cells. We conclude that both STAT3 and C/EBPß are needed to elicit IL-10-mediated NGAL expression in primary human macrophages. Macrophage-secreted NGAL shapes the protumorigenic macrophage phenotype to promote growth of MCF-7 breast cancer cells. Our data point to a macrophage-dependent IL-10-STAT3-NGAL axis that might contribute to tumor progression.


Subject(s)
Acute-Phase Proteins/immunology , Breast Neoplasms/immunology , Interleukin-10/immunology , Lipocalins/immunology , Macrophages/immunology , Proto-Oncogene Proteins/immunology , Acute-Phase Proteins/genetics , Breast/cytology , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Female , Humans , Janus Kinases/immunology , Lipocalin-2 , Lipocalins/genetics , Macrophages/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...