Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters











Database
Language
Publication year range
1.
Front Immunol ; 11: 1885, 2020.
Article in English | MEDLINE | ID: mdl-33042110

ABSTRACT

Cytokines are known to shape the tumor microenvironment and although progress has been made in understanding their role in carcinogenesis, much remains to learn regarding their role in tumor growth and progression. We have identified granulocyte colony-stimulating factor (G-CSF) as one such cytokine, showing that G-CSF is linked with metastasis in human gastrointestinal tumors and neutralizing G-CSF in a mouse model of colitis-associated cancer is protective. Here, we set out to identify the role of G-CSF and its receptor, G-CSFR, in CD4+ and CD8+ T cell responses in the tumor microenvironment. MC38 colon cancer cells were injected into WT, G-CSFR-/- mice, or Rag2-/- mice. Flow cytometry, Real Time PCR and Multiplex cytokine array analysis were used for in vitro T cell phenotype analysis. Adoptive transfer of WT or G-CSFR-/- CD4+ of CD8+ T cells were performed. Mouse tumor size, cytokine expression, T cell phenotype, and cytotoxic activity were analyzed. We established that in G-CSFR-/- mice, tumor growth of MC38 colon cancer cells is significantly decreased. T cell phenotype and cytokine production were also altered, as both in vitro and in vivo approaches revealed that the G-CSF/G-CSFR stimulate IL-10-producing, FoxP3-expressing CD4+ and CD8+ T cells, whereas G-CSFR-/- T cells exhibit increased IFNγ and IL-17A production, leading to increased cytotoxic activity in the tumor microenvironment. Furthermore, peritumoral injection of recombinant IFNγ or IL-17A inhibited colon and pancreas tumor growth compared to controls. Taken together, our data reveal an unknown mechanism by which G-CSF, through its receptor G-CSFR, promotes an inhibitory Treg phenotype that limits tumor immune responses and furthermore suggest that targeting this cytokine/receptor axis could represent a novel therapeutic approach for gastrointestinal, and likely other tumors with high expression of these factors.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Colonic Neoplasms/metabolism , Cytotoxicity, Immunologic , Granulocyte Colony-Stimulating Factor/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Receptors, Colony-Stimulating Factor/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Coculture Techniques , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-17/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Colony-Stimulating Factor/deficiency , Receptors, Colony-Stimulating Factor/genetics , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Tumor Burden , Tumor Microenvironment
2.
Front Immunol ; 9: 1857, 2018.
Article in English | MEDLINE | ID: mdl-30298062

ABSTRACT

A major risk factor for colon cancer growth and progression is chronic inflammation. We have shown that the MAPK-activated protein kinase 2 (MK2) pathway is critical for colon tumor growth in colitis-associated and spontaneous colon cancer models. This pathway is known to regulate expression of the tumor-promoting cytokines, IL-1, IL-6, and TNF-α. However, little is known about the ability of MK2 to regulate chemokine production. This is the first study to demonstrate this pathway also regulates the chemokines, MCP-1, Mip-1α, and Mip-2α (MMM). We show that these chemokines induce tumor cell growth and invasion in vitro and that MK2 inhibition suppresses tumor cell production of chemokines and reverses the resulting pro-tumorigenic effects. Addition of MMM to colon tumors in vivo significantly enhances tumor growth in control tumors and restores tumor growth in the presence of MK2 inhibition. We also demonstrate that MK2 signaling is critical for chemokine expression and macrophage influx to the colon tumor microenvironment. MK2 signaling in macrophages was essential for inflammatory cytokine/chemokine production, whereas MK2-/- macrophages or MK2 inhibition suppressed cytokine expression. We show that addition of bone marrow-derived macrophages to the tumor microenvironment enhances tumor growth in control tumors and restores tumor growth in tumors treated with MK2 inhibitors, while addition of MK2-/- macrophages had no effect. This is the first study to demonstrate the critical role of the MK2 pathway in chemokine production, macrophage influx, macrophage function, and tumor growth.


Subject(s)
Chemotaxis/immunology , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Cytokines/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/immunology , Macrophages/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Line, Tumor , Cell Movement/immunology , Cell Proliferation , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Cytokines/genetics , Disease Models, Animal , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Macrophage Activation/immunology , Macrophages/drug effects , Mice , Mice, Knockout , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Tumor Microenvironment , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL