Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Dis ; 10(10): 760, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31591378

ABSTRACT

Melanoma immunotherapy is still not satisfactory due to immunosuppressive cell populations within the tumor stroma. Targeting tumor-associated macrophages (TAM) can help to restore an anti-tumor immunity. Previously, we could show that classical TAM markers expressed in vivo need a 7 day M-CSF/dexamethasone/IL-4 (MDI) stimulation for their induction in peripheral blood monocytes (pBM) in vitro. To identify possible novel therapeutic targets on TAM, gene expression analysis of MDI-treated pBM was performed. This identified up-regulation of the purinergic G-protein coupled receptor P2Y12, the therapeutic target of the clinically approved anti-thrombotic drugs cangrelor, clopidogrel, ticagrelor, and prasugrel. We generated a peptide antibody and validated its specificity using transgenic P2Y12+ U937 cells. With the help of this antibody, P2Y12 expression was confirmed on CD68+ CD163+ TAM of melanoma in situ. Functional analysis revealed that treatment of transgenic P2Y12+ U937 cells with the receptor agonist 2-MeSADP induced ERK1/2 and Akt phosphorylation and increased the secretion of the chemokines CXCL2, CXCL7, and CXCL8. These effects could be abolished with the P2Y12 antagonist PSB0739 or with Akt and ERK inhibitors. In addition, P2Y12+ macrophages migrated towards the ADP-rich culture medium of puromycin-treated dying B16F1 melanoma cells. Cangrelor treatment blocked migration. Taken together, our results indicate that P2Y12 is an important chemotaxis receptor, which triggers migration of macrophages towards nucleotide-rich, necrotic tumor areas, and modulates the inflammatory environment upon ADP binding.


Subject(s)
Chemokines/genetics , Chemotaxis/drug effects , Melanoma/drug therapy , Receptors, Purinergic P2Y12/genetics , Adenosine Diphosphate/biosynthesis , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Cell Line, Tumor , Chemokine CXCL2/genetics , Chemotaxis/genetics , Clopidogrel/pharmacology , Dexamethasone/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-4/pharmacology , Interleukin-8/genetics , MAP Kinase Signaling System/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Melanoma/genetics , Melanoma/pathology , Monocytes/drug effects , Phosphorylation/drug effects , Prasugrel Hydrochloride/pharmacology , Ticagrelor/pharmacology , beta-Thromboglobulin/genetics
2.
Cell Death Dis ; 9(10): 939, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30232321

ABSTRACT

Melanoma is a highly immunogenic tumor with a good response to treatment with immune checkpoint inhibitors. Tumor-associated macrophages (TAMs) play an important immunosuppressive role in such tumors and have therefore been identified as possible future therapeutic targets in oncology. The aim of this study was to identify novel immunoregulatory receptors specifically expressed on TAM. Expression of Slamf9, a member of the signaling lymphocytic-activating molecule (Slam) immunoreceptor family, was found to be upregulated in a gene expression analysis of murine bone marrow-derived macrophages (BMDM) stimulated with tumor-conditioned medium of B16F1 melanoma cells. SLAMF9+ macrophages were identified in human and murine melanomas by using self-generated antibodies against human and murine SLAMF9. A comprehensive immunohistochemical analysis of tissue microarrays detected SLAMF9+ TAM in 73.3% of human melanomas, but also in 95.5% of naevi of melanoma patients and in 50% of naevi from healthy controls. In addition, 20% of melanomas and 2.3% of naevi from melanoma patients displayed a positive SLAMF9 expression also in melanocytic cells. No SLAMF9 expression was detected in naevus cells of healthy donors. Although SLAMF9 has no intracellular signaling motif, a comprehensive functional analysis revealed that the molecule was able to significantly enhance TNF-α secretion after LPS-stimulation. In addition, SLAMF9 delayed the wound closure of RAW 264.7 cells in a scratch assay, while proliferation and cell death were not affected. Taken together, SLAMF9 is a novel type-I-transmembrane receptor with immunomodulatory properties in macrophages. Further studies are required to evaluate whether SLAMF9 classifies as a promising future therapeutic target in melanoma.


Subject(s)
Melanoma/metabolism , Signaling Lymphocytic Activation Molecule Family/metabolism , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Humans , Interferon-gamma/pharmacology , Lipopolysaccharides , Macrophages/drug effects , Macrophages/metabolism , Melanocytes , Melanoma/genetics , Mice , Signal Transduction/drug effects , Signal Transduction/physiology , Signaling Lymphocytic Activation Molecule Family/genetics , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
Oncotarget ; 8(61): 103682-103692, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29262593

ABSTRACT

Targeting immune cells that support tumor growth is an effective therapeutic strategy in tumor entities such as melanoma. M2-like tumor-associated macrophages (TAM) sustain tumor growth by secreting anti-inflammatory cytokines, proteases and growth factors. In this study, we show that a protein derived from M2-like macrophages namely the shedded ectodomain of Lyve-1 (sLyve-1) decreases human HT144 and murine B16F1 melanoma cell proliferation significantly by acting as a decoy receptor for low-molecular weight hyaluronic acid (LMW-HA) although the LMW-HA/Lyve-1 interaction on lymphatic endothelial cells has been described to induce lymphangiogenesis. This is in line with our finding that the number of LYVE-1+ TAM decreases in higher human melanoma stages and that the early growth of B16 transplant tumors is enhanced in Lyve-1 knockout mice when compared to wild-type mice due to an increased melanoma cell proliferation. LYVE-1 expressing TAM are however true M2 macrophages as they co-express typical M2-markers such as CD163 and CD206. The results of the present study highlight the necessity to carefully determine the net effect particular TAM subpopulations have on tumors before establishing a treatment to target these immune cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...