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.
J Pathol ; 243(3): 320-330, 2017 11.
Article in English | MEDLINE | ID: mdl-28799208

ABSTRACT

Monocytes infiltrating scar tissue are predominantly viewed as progenitor cells. Here, we show that tissue CCR2+ monocytes have specific immunosuppressive and profibrotic functions. CCR2+ monocytic cells are acutely recruited to the lung before the onset of silica-induced fibrosis in mice. These tissue monocytes are defined as monocytic myeloid-derived suppressor cells (M-MDSCs) because they significantly suppress T-lymphocyte proliferation in vitro. M-MDSCs collected from silica-treated mice also express transforming growth factor (TGF)-ß1, which stimulates lung fibroblasts to release tissue inhibitor of metalloproteinase (TIMP)-1, an inhibitor of metalloproteinase collagenolytic activity. By using LysMCreCCR2loxP/loxP mice, we show that limiting CCR2+ M-MDSC accumulation reduces the pulmonary contents of TGF-ß1, TIMP-1 and collagen after silica treatment. M-MDSCs do not differentiate into lung macrophages, granulocytes or fibrocytes during pulmonary fibrogenesis. Collectively, our data indicate that M-MDSCs contribute to lung fibrosis by specifically promoting a non-degrading collagen microenvironment. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Monocytes/metabolism , Myeloid-Derived Suppressor Cells/cytology , Pulmonary Fibrosis/metabolism , Receptors, CCR2/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cell Proliferation/physiology , Collagen/metabolism , Lung/pathology , Lymphocyte Activation/physiology , Mice, Inbred C57BL , Pulmonary Fibrosis/pathology
2.
Part Fibre Toxicol ; 13(1): 46, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27549627

ABSTRACT

BACKGROUND: The asbestos-like toxicity of some engineered carbon nanotubes (CNT), notably their capacity to induce mesothelioma, is a serious cause of concern for public health. Here we show that carcinogenic CNT induce an early and sustained immunosuppressive response characterized by the accumulation of monocytic Myeloid Derived Suppressor Cells (M-MDSC) that counteract effective immune surveillance of tumor cells. METHODS: Wistar rats and C57BL/6 mice were intraperitoneally injected with carcinogenic multi-walled Mitsui-7 CNT (CNT-7) or crocidolite asbestos. Peritoneal mesothelioma development and immune cell accumulation were assessed until 12 months. Leukocyte sub-populations were identified by recording expression of CD11b/c and His48 by flow cytometry. The immunosuppressive activity on T lymphocytes of purified peritoneal leukocytes was assessed in a co-culture assay with activated spleen cells. RESULTS: We demonstrate that long and short mesotheliomagenic CNT-7 injected in the peritoneal cavity of rats induced, like asbestos, an early and selective accumulation of monocytic cells (CD11b/c(int) and His48(hi)) which possess the ability to suppress polyclonal activation of T lymphocytes and correspond to M-MDSC. Peritoneal M-MDSC persisted during the development of peritoneal mesothelioma in CNT-7-treated rats but were only transiently recruited after non-carcinogenic CNT (CNT-M, CNT-T) injection. Peritoneal M-MDSC did not accumulate in mice which are resistant to mesothelioma development. CONCLUSIONS: Our data provide new insights into the initial pathogenic events induced by CNT, adding a new component to the adverse outcome pathway leading to mesothelioma development. The specificity of the M-MDSC response after carcinogenic CNT exposure highlights the interest of this response for detecting the ability of new nanomaterials to cause cancer.


Subject(s)
Carcinogens/toxicity , Mesothelioma/chemically induced , Monocytes/immunology , Nanotubes, Carbon/toxicity , Animals , Heterografts , Humans , Male , Mesothelioma/immunology , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar
3.
J Pathol ; 235(5): 698-709, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25421226

ABSTRACT

Macrophages play a central role in immune and tissue responses of granulomatous lung diseases induced by pathogens and foreign bodies. Circulating monocytes are generally viewed as central precursors of these tissue effector macrophages. Here, we provide evidence that granulomas derive from alveolar macrophages serving as a local reservoir for the expansion of activated phagocytic macrophages. By exploring lung granulomatous responses to silica particles in IL-1-deficient mice, we found that the absence of IL-1α, but not IL-1ß, was associated with reduced CD11b(high) phagocytic macrophage accumulation and fewer granulomas. This defect was associated with impaired alveolar clearance and resulted in the development of pulmonary alveolar proteinosis (PAP). Reconstitution of IL-1α(-/-) mice with recombinant IL-1α restored lung clearance functions and the pulmonary accumulation of CD11b(high) phagocytic macrophages. Mechanistically, IL-1α induced the proliferation of CD11b(low) alveolar macrophages and differentiated these cells into CD11b(high) macrophages which perform critical phagocytic functions and organize granuloma. We newly discovered here that IL-1α triggers lung responses requiring macrophage proliferation and maturation from tissue-resident macrophages.


Subject(s)
CD11b Antigen/metabolism , Cell Proliferation , Granuloma/metabolism , Interleukin-1alpha/metabolism , Lung Diseases/metabolism , Macrophage Activation , Macrophages, Alveolar/metabolism , Pulmonary Alveolar Proteinosis/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Granuloma/chemically induced , Granuloma/genetics , Granuloma/pathology , Interleukin-1alpha/deficiency , Interleukin-1alpha/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lung Diseases/chemically induced , Lung Diseases/genetics , Lung Diseases/pathology , Macrophages, Alveolar/pathology , Mice, Knockout , Phagocytosis , Phenotype , Pulmonary Alveolar Proteinosis/chemically induced , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Alveolar Proteinosis/pathology , Silicon Dioxide , Time Factors
4.
Part Fibre Toxicol ; 11: 69, 2014 Dec 13.
Article in English | MEDLINE | ID: mdl-25497724

ABSTRACT

BACKGROUND: Inflammasome-activated IL-1ß plays a major role in lung neutrophilic inflammation induced by inhaled silica. However, the exact mechanisms that contribute to the initial production of precursor IL-1ß (pro-IL-1ß) are still unclear. Here, we assessed the implication of alarmins (IL-1α, IL-33 and HMGB1) in the lung response to silica particles and found that IL-1α is a master cytokine that regulates IL-1ß expression. METHODS: Pro- and mature IL-1ß as well as alarmins were assessed by ELISA, Western Blot or qRT-PCR in macrophage cultures and in mouse lung following nano- and micrometric silica exposure. Implication of these immune mediators in the establishment of lung inflammatory responses to silica was investigated in knock-out mice or after antibody blockade by evaluating pulmonary neutrophil counts, CXCR2 expression and degree of histological injury. RESULTS: We found that the early release of IL-1α and IL-33, but not HMGB1 in alveolar space preceded the lung expression of pro-IL-1ß and neutrophilic inflammation in silica-treated mice. In vitro, the production of pro-IL-1ß by alveolar macrophages was significantly induced by recombinant IL-1α but not by IL-33. Neutralization or deletion of IL-1α reduced IL-1ß production and neutrophil accumulation after silica in mice. Finally, IL-1α released by J774 macrophages after in vitro exposure to a range of micro- and nanoparticles of silica was correlated with the degree of lung inflammation induced in vivo by these particles. CONCLUSIONS: We demonstrated that in response to silica exposure, IL-1α is rapidly released from pre-existing stocks in alveolar macrophages and promotes subsequent lung inflammation through the stimulation of IL-1ß production. Moreover, we demonstrated that in vitro IL-1α release from macrophages can be used to predict the acute inflammogenic activity of silica micro- and nanoparticles.


Subject(s)
Air Pollutants/toxicity , Inhalation Exposure/adverse effects , Interleukin-1alpha/metabolism , Lung/drug effects , Nanoparticles/toxicity , Pneumonia/chemically induced , Silicon Dioxide/toxicity , Air Pollutants/chemistry , Animals , Antibodies, Neutralizing/metabolism , Cell Line , Cells, Cultured , Female , Interleukin-1alpha/antagonists & inhibitors , Interleukin-1alpha/genetics , Interleukin-1beta/metabolism , Lung/immunology , Lung/metabolism , Lung/pathology , Macrophages, Alveolar/cytology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microspheres , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neutrophil Infiltration/drug effects , Particle Size , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry , Toxicity Tests, Acute
5.
PLoS One ; 9(7): e99383, 2014.
Article in English | MEDLINE | ID: mdl-25050810

ABSTRACT

The exact implication of innate immunity in granuloma formation and irreversible lung fibrosis remains to be determined. In this study, we examined the lung inflammatory and fibrotic responses to silica in MyD88-knockout (KO) mice. In comparison to wild-type (WT) mice, we found that MyD88-KO animals developed attenuated lung inflammation, neutrophil accumulation and IL-1ß release in response to silica. Granuloma formation was also less pronounced in MyD88-KO mice after silica. This limited inflammatory response was not accompanied by a concomitant attenuation of lung collagen accumulation after silica. Histological analyses revealed that while pulmonary fibrosis was localized in granulomas in WT animals, it was diffusely distributed throughout the parenchyma in MyD88-KO mice. Robust collagen accumulation was also observed in mice KO for several other components of innate immunity (IL-1R, IL-1, ASC, NALP3, IL-18R, IL-33R, TRIF, and TLR2-3-4,). We additionally show that pulmonary fibrosis in MyD88-KO mice was associated with the accumulation of pro-fibrotic regulatory T lymphocytes (T regs) and pro-fibrotic cytokine expression (TGF-ß, IL-10 and PDGF-B), not with T helper (Th) 17 cell influx. Our findings indicate that the activation of MyD88-related innate immunity is central in the establishment of particle-induced lung inflammatory and granuloma responses. The development of lung fibrosis appears uncoupled from inflammation and may be orchestrated by a T reg-associated pathway.


Subject(s)
Inflammation/immunology , Myeloid Differentiation Factor 88/immunology , Pulmonary Fibrosis/immunology , Silicon Dioxide/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Flow Cytometry , Granuloma/genetics , Granuloma/immunology , Granuloma/metabolism , Immunity, Innate/genetics , Immunity, Innate/immunology , Immunohistochemistry , Inflammation/chemically induced , Inflammation/genetics , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Interleukin-1alpha/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Silicon Dioxide/toxicity , Silicosis/etiology , Silicosis/genetics , Silicosis/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...