Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 10: 2604, 2019.
Article in English | MEDLINE | ID: mdl-31787980

ABSTRACT

High aspect ratio zinc oxide nanowires (ZnONWs) have become one of the most important products in nanotechnology. The wide range applications of ZnONWs have heightened the need for evaluating the risks and biological consequences to these particles. In this study, we investigated inflammatory pathways activated by ZnONWs in cultured cells as well as the consequences of systemic exposure in mouse models. Confocal microscopy showed rapid phagocytic uptake of FITC-ZnONWs by macrophages. Exposure of macrophages or lung epithelial cells to ZnONWs induced the production of CCL2 and CCL11. Moreover, ZnONWs exposure induced both IL-6 and TNF-α production only in macrophages but not in LKR13 cells. Intratracheal instillation of ZnONWs in C57BL/6 mice induced a significant increase in the total numbers of immune cells in the broncho alveolar lavage fluid (BALFs) 2 days after instillation. Macrophages and eosinophils were the predominant cellular infiltrates of ZnONWs exposed mouse lungs. Similar cellular infiltrates were also observed in a mouse air-pouch model. Pro-inflammatory cytokines IL-6 and TNF-α as well as chemokines CCL11, and CCL2 were increased both in BALFs and air-pouch lavage fluids. These results suggest that exposure to ZnONWs may induce distinct inflammatory responses through phagocytic uptake and formation of soluble Zn2+ ions.


Subject(s)
Chemokine CCL11/immunology , Eosinophils/drug effects , Eosinophils/immunology , Inflammation/etiology , Nanowires/adverse effects , Zinc Oxide/adverse effects , Animals , Chemokine CCL11/genetics , Chemokine CCL2/genetics , Disease Models, Animal , In Vitro Techniques , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/metabolism , Interleukin-6/genetics , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Nanowires/chemistry , Neutrophils/drug effects , Neutrophils/immunology , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/genetics , Up-Regulation/drug effects , Zinc Oxide/chemistry
2.
J Immunol ; 200(10): 3556-3567, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29610142

ABSTRACT

Silicosis is a lung inflammatory disease caused by chronic exposure to crystalline silica (CS). Leukotriene B4 (LTB4) plays an important role in neutrophilic inflammation, which drives silicosis and promotes lung cancer. In this study, we examined the mechanisms involved in CS-induced inflammatory pathways. Phagocytosis of CS particles is essential for the production of LTB4 and IL-1ß in mouse macrophages, mast cells, and neutrophils. Phagosomes enclosing CS particles trigger the assembly of lipidosome in the cytoplasm, which is likely the primary source of CS-induced LTB4 production. Activation of the JNK pathway is essential for both CS-induced LTB4 and IL-1ß production. Studies with bafilomycin-A1- and NLRP3-deficient mice revealed that LTB4 synthesis in the lipidosome is independent of inflammasome activation. Small interfering RNA knockdown and confocal microscopy studies showed that GTPases Rab5c, Rab40c along with JNK1 are essential for lipidosome formation and LTB4 production. BI-78D3, a JNK inhibitor, abrogated CS-induced neutrophilic inflammation in vivo in an air pouch model. These results highlight an inflammasome-independent and JNK activation-dependent lipidosome pathway as a regulator of LTB4 synthesis and CS-induced sterile inflammation.


Subject(s)
Inflammasomes/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Leukotriene B4/metabolism , Silicon Dioxide/pharmacology , Animals , Cell Line , Humans , Interleukin-1beta/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Phagosomes/drug effects , Phagosomes/metabolism , RAW 264.7 Cells , Silicosis/metabolism , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism
3.
Cancer Immunol Res ; 6(3): 332-347, 2018 03.
Article in English | MEDLINE | ID: mdl-29382671

ABSTRACT

The presence of mast cells in some human colorectal cancers is a positive prognostic factor, but the basis for this association is incompletely understood. Here, we found that mice with a heterozygous mutation in the adenomatous polyposis coli gene (ApcMin/+) displayed reduced intestinal tumor burdens and increased survival in a chemokine decoy receptor, ACKR2-null background, which led to discovery of a critical role for mast cells in tumor defense. ACKR2-/-ApcMin/+ tumors showed increased infiltration of mast cells, their survival advantage was lost in mast cell-deficient ACKR2-/-SA-/-ApcMin/+ mice as the tumors grew rapidly, and adoptive transfer of mast cells restored control of tumor growth. Mast cells from ACKR2-/- mice showed elevated CCR2 and CCR5 expression and were also efficient in antigen presentation and activation of CD8+ T cells. Mast cell-derived leukotriene B4 (LTB4) was found to be required for CD8+ T lymphocyte recruitment, as mice lacking the LTB4 receptor (ACKR2-/-BLT1-/-ApcMin/+) were highly susceptible to intestinal tumor-induced mortality. Taken together, these data demonstrate that chemokine-mediated recruitment of mast cells is essential for initiating LTB4/BLT1-regulated CD8+ T-cell homing and generation of effective antitumor immunity against intestinal tumors. We speculate that the pathway reported here underlies the positive prognostic significance of mast cells in selected human tumors. Cancer Immunol Res; 6(3); 332-47. ©2018 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Intestinal Neoplasms/immunology , Mast Cells/immunology , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/immunology , Animals , Female , Immunologic Surveillance , Leukotriene B4/immunology , Male , Mice, Transgenic , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Receptors, Leukotriene B4/genetics , Receptors, Leukotriene B4/immunology
4.
Semin Immunol ; 33: 58-64, 2017 10.
Article in English | MEDLINE | ID: mdl-28982616

ABSTRACT

The high affinity leukotriene B4 receptor, BLT1 mediates chemotaxis of diverse leukocyte subsets to the sites of infection or inflammation. Whereas the pathological functions of LTB4/BLT1 axis in allergy, autoimmunity and cardiovascular disorders are well established; its role in cancer is only beginning to emerge. In this review, we summarize recent findings on LTB4/BLT1 axis enabling distinct outcomes toward tumor progression. In a mouse lung tumor model promoted by silicosis-induced inflammation, genetic deletion of BLT1 attenuated neutrophilic inflammation and tumor promotion. In contrast, in a spontaneous model of intestinal tumorigenesis, absence of BLT1 led to defective mucosal host response, altered microbiota and bacteria dependent colon tumor progression. Furthermore, BLT1 mediated CD8+ T cell recruitment was shown to be essential for initiating anti-tumor immunity in number of xenograft models and is critical for effective PD1 based immunotherapy. BLT2 mediated chemotherapy resistance, tumor promotion and metastasis are also discussed. This new information points to a paradigm shift in our understanding of the LTB4 pathways in cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Inflammation/immunology , Leukocytes/immunology , Leukotriene B4/metabolism , Neoplasms/immunology , Receptors, Leukotriene B4/metabolism , Animals , Carcinogenesis , Cell Movement , Chemotaxis , Humans , Mice , Mice, Knockout , Signal Transduction , Xenograft Model Antitumor Assays
5.
Nat Commun ; 6: 7064, 2015 Apr 29.
Article in English | MEDLINE | ID: mdl-25923988

ABSTRACT

Chronic exposure to crystalline silica (CS) causes silicosis, an irreversible lung inflammatory disease that may eventually lead to lung cancer. In this study, we demonstrate that in K-ras(LA1) mice, CS exposure markedly enhances the lung tumour burden and genetic deletion of leukotriene B4 receptor-1 (BLT1(-/-)) attenuates this increase. Pulmonary neutrophilic inflammation induced by CS is significantly reduced in BLT1(-/-)K-ras(LA1) mice. CS exposure induces LTB4 production by mast cells and macrophages independent of inflammasome activation. In an air-pouch model, CS-induced neutrophil recruitment is dependent on LTB4 production by mast cells and BLT1 expression on neutrophils. In an implantable lung tumour model, CS exposure results in rapid tumour growth and decreased survival that is attenuated in the absence of BLT1. These results suggest that the LTB4/BLT1 axis sets the pace of CS-induced sterile inflammation that promotes lung cancer progression. This knowledge may facilitate development of immunotherapeutic strategies to fight silicosis and lung cancer.


Subject(s)
Inflammation/pathology , Leukotriene B4/metabolism , Lung Neoplasms/pathology , Silicon Dioxide/adverse effects , Animals , Cell Proliferation , Chemokines/biosynthesis , Chemotactic Factors/metabolism , Crystallization , Disease Progression , Inflammation Mediators/metabolism , Interleukin-1beta/biosynthesis , Lung Neoplasms/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Neutrophil Infiltration , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Leukotriene B4/deficiency , Receptors, Leukotriene B4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...