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.
Exp Dermatol ; 29(1): 71-78, 2020 01.
Article in English | MEDLINE | ID: mdl-31721311

ABSTRACT

Several cytokines signalling via Janus Kinase (JAK) proteins have been implicated in the pathogenesis of immune-mediated inflammatory diseases, including psoriasis and rheumatoid arthritis (RA). Tofacitinib, a small JAK inhibitor, is approved for the treatment of RA and has demonstrated good efficacy in psoriasis phase III clinical trials. In this work, we analysed the in vitro effects of tofacitinib on the functions of human dendritic cells (DCs) and macrophages. When assessing the effects of tofacitinib on monocyte-derived DCs, we observed reduced differentiation of monocytes into immature DCs, as evidenced by a decreased transcription of CD209 and CD80. Phenotype assessment in the presence of tofacitinib suggested a switch towards a M1-like macrophage phenotype, as evidenced by the expression of M1 markers such as iNOS, as well as cytokines typically expressed by M1 cells, including IL-12 and IL-23. Of note, Arginase1 and CD200R, typically expressed by M2 cells, were absent on tofacitinib-treated DCs. Furthermore, tofacitinib affected the response of differentiated DCs to maturation stimuli such as LPS and IFNγ, resulting in a partial up-regulation of IL-23 and down-regulation of IL-12, as assessed by qPCR. When investigating macrophage development, we found that tofacitinib inhibited the ability of monocytes to differentiate and polarize into regulatory M2 macrophages, while rather enhancing the ability to develop into inflammatory M1-like macrophages, as evidenced by decreased expression of the M2 marker CD200R and enhanced production of IL-12 and IL-23. In conclusion, tofacitinib impacts the differentiation of human DCs and macrophages, it particularly favours generation of M1-like pro-inflammatory macrophages.


Subject(s)
Cell Differentiation/drug effects , Janus Kinase Inhibitors/pharmacology , Langerhans Cells/physiology , Macrophages/physiology , Monocytes/physiology , Piperidines/pharmacology , Pyrimidines/pharmacology , Cells, Cultured , Dendritic Cells , Down-Regulation/drug effects , Gene Expression/drug effects , Humans , Interferon-gamma/pharmacology , Interleukin-12/genetics , Interleukin-23/genetics , Lipopolysaccharides/pharmacology , Orexin Receptors/metabolism , Phenotype , RNA, Messenger/metabolism , Up-Regulation/drug effects
2.
Oncotarget ; 7(47): 77825-77837, 2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27788481

ABSTRACT

In a previous study on a xenograft model of melanoma, we showed that the beta-adrenergic receptor antagonist propranolol inhibits melanoma development by modulating angiogenesis, proliferation and cell survival. Stress hormones can influence tumor development in different ways and norepinephrine was shown to downregulate antitumor immune responses by favoring the accumulation of immunosuppressive cells, impairing the function of lymphocytes. We assessed the effect of propranolol on antitumor immune response in the MT/Ret mouse model of melanoma. Propranolol treatment delayed primary tumor growth and metastases development in MT/Ret mice. Consistent with our previous observations in human melanoma xenografts, propranolol induces a decrease in cell proliferation and vessel density in the primary tumors and in metastases. In this immunocompetent model, propranolol significantly reduced the infiltration of myeloid cells, particularly neutrophils, in the primary tumor. Inversely, cytotoxic tumor infiltrating lymphocytes were more frequent in the tumor stroma of treated mice. In a consistent manner, we observed the same shift in the proportions of infiltrating leukocytes in the metastases of treated mice. Our results suggest that propranolol, by decreasing the infiltration of immunosuppressive myeloid cells in the tumor microenvironment, restores a better control of the tumor by cytotoxic cells.


Subject(s)
Antineoplastic Agents/administration & dosage , Melanoma, Experimental/drug therapy , Propranolol/administration & dosage , Skin Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Propranolol/pharmacology , Proto-Oncogene Proteins c-ret/genetics , Skin Neoplasms/immunology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...