Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 10353, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31316109

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is associated with neutrophilic lung inflammation and CD8 T cell exhaustion and is an important risk factor for the development of non-small cell lung cancer (NSCLC). The clinical response to programmed cell death-1 (PD-1) blockade in NSCLC patients is variable and likely affected by a coexisting COPD. The pro-inflammatory cytokine interleukin-17C (IL-17C) promotes lung inflammation and is present in human lung tumors. Here, we used a Kras-driven lung cancer model to examine the function of IL-17C in inflammation-promoted tumor growth. Genetic ablation of Il-17c resulted in a decreased recruitment of inflammatory cells into the tumor microenvironment, a decreased expression of tumor-promoting cytokines (e.g. interleukin-6 (IL-6)), and a reduced tumor proliferation in the presence of Haemophilus influenzae- (NTHi) induced COPD-like lung inflammation. Chronic COPD-like inflammation was associated with the expression of PD-1 in CD8 lymphocytes and the membrane expression of the programmed death ligand (PD-L1) independent of IL-17C. Tumor growth was decreased in Il-17c deficient mice but not in wildtype mice after anti-PD-1 treatment. Our results suggest that strategies targeting innate immune mechanisms, such as blocking of IL-17C, may improve the response to anti-PD-1 treatment in lung cancer patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Immunity, Innate , Interleukin-17/physiology , Lung Neoplasms/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/physiology , Animals , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/etiology , Carcinoma, Non-Small-Cell Lung/immunology , Cell Line, Tumor , Cytokines/physiology , Female , Genes, ras , Humans , Interleukin-17/deficiency , Interleukin-17/genetics , Interleukin-17/pharmacology , Lung Neoplasms/etiology , Lung Neoplasms/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neutrophils/physiology , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/genetics , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/immunology , Recombinant Proteins/pharmacology , Tumor Microenvironment
2.
Oncogene ; 36(29): 4182-4190, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28346430

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is associated with an increased risk for lung cancer and an aberrant microbiota of the lung. Microbial colonization contributes to chronic neutrophilic inflammation in COPD. Nontypeable Haemophilus influenzae (NTHi) is frequently found in lungs of stable COPD patients and is the major pathogen triggering exacerbations. The epithelial cytokine interleukin-17C (IL-17C) promotes the recruitment of neutrophils into inflamed tissues. The purpose of this study was to investigate the function of IL-17C in the pulmonary tumor microenvironment. We subjected mice deficient for IL-17C (IL-17C-/-) and mice double deficient for Toll-like receptor 2 and 4 (TLR-2/4-/-) to a metastatic lung cancer model. Tumor proliferation and growth as well as the number of tumor-associated neutrophils was significantly decreased in IL-17C-/- and TLR-2/4-/- mice exposed to NTHi. The NTHi-induced pulmonary expression of IL-17C was dependent on TLR-2/4. In vitro, IL-17C increased the NTHi- and tumor necrosis factor-α-induced expression of the neutrophil chemokines keratinocyte-derived chemokine and macrophage inflammatory protein 2 in lung cancer cells but did not affect proliferation. Human lung cancer samples stained positive for IL-17C, and in non-small cell lung cancer patients with lymph node metastasis, IL-17C was identified as a negative prognostic factor. Our data indicate that epithelial IL-17C promotes neutrophilic inflammation in the tumor microenvironment and suggest that IL-17C links a pathologic microbiota, as present in COPD patients, with enhanced tumor growth.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Interleukin-17/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Neutrophils/immunology , Animals , Female , Humans , Interleukin-17/genetics , Lung Neoplasms/genetics , Lung Neoplasms/microbiology , Mice , Mice, Inbred C57BL , Microbiota , Neutrophils/pathology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/microbiology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/immunology , Tumor Microenvironment
3.
Infect Immun ; 84(7): 2031-2041, 2016 07.
Article in English | MEDLINE | ID: mdl-27113358

ABSTRACT

In Staphylococcus aureus, metabolism is intimately linked with virulence determinant biosynthesis, and several metabolite-responsive regulators have been reported to mediate this linkage. S. aureus possesses at least three members of the RpiR family of transcriptional regulators. Of the three RpiR homologs, RpiRc is a potential regulator of the pentose phosphate pathway, which also regulates RNAIII levels. RNAIII is the regulatory RNA of the agr quorum-sensing system that controls virulence determinant synthesis. The effect of RpiRc on RNAIII likely involves other regulators, as the regulators that bind the RNAIII promoter have been intensely studied. To determine which regulators might bridge the gap between RpiRc and RNAIII, sarA, sigB, mgrA, and acnA mutations were introduced into an rpiRc mutant background, and the effects on RNAIII were determined. Additionally, phenotypic and genotypic differences were examined in the single and double mutant strains, and the virulence of select strains was examined using two different murine infection models. The data suggest that RpiRc affects RNAIII transcription and the synthesis of virulence determinants in concert with σ(B), SarA, and the bacterial metabolic status to negatively affect virulence.


Subject(s)
Bacterial Proteins/metabolism , Repressor Proteins/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Disease Models, Animal , Female , Genetic Loci , Mice , Mutation , Operon , Repressor Proteins/genetics , Staphylococcal Infections/mortality , Transcription, Genetic , Virulence/genetics , Virulence Factors/genetics
4.
Cell Tissue Res ; 364(2): 225-9, 2016 May.
Article in English | MEDLINE | ID: mdl-26984603

ABSTRACT

This mini-review examines the role of the pro-inflammatory cytokine interleukin (IL)-1ß in the interaction of Pseudomonas aeruginosa and the host immune system during lung infection. Different studies show that the reduction of the inflammatory response, especially a decrease in IL-1ß, leads to a better outcome in acute lung infection with this bacterium. This includes a higher survival rate, reduced damage to the lung tissue and, in particular, a better clearance of the airways and the tissue of the lungs from P. aeruginosa.


Subject(s)
Interleukin-1beta/immunology , Lung/pathology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Animals , Humans , Inflammasomes/immunology , Inflammation/immunology , Lung/microbiology , Mice , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology
5.
Am J Physiol Lung Cell Mol Physiol ; 309(6): L605-13, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26209273

ABSTRACT

Microorganisms have an important role in tumorgenesis by the induction of inflammation and by a direct impact on tumor cells. Chronic obstructive pulmonary disease (COPD) is associated with an increased risk for lung cancer and microbial colonization. We asked whether bacterial pathogens act as tumor promoters during CS-induced pulmonary inflammation. In a metastatic lung cancer (LC) model, Lewis lung carcinoma (LLC) cells were injected in mice to initiate the growth of tumors in the lung. Exposure to the combination of cigarette smoke (CS) and nontypeable Haemophilus influenzae (NTHi) synergistically increased metastatic growth. Lung levels of albumin and LDH, translocation of bacterial factors into tumor tissue, tumor inflammation, and tumor proliferation were significantly increased in mice exposed to CS in combination with NTHi. Bacterial pathogens increased the proliferation of cultured LLC cells and human cancer cell lines. Metastatic growth induced by the exposure to CS in combination with NTHi was reduced in mice deficient for IL-17. Our data provide evidence that CS-induced loss of pulmonary barrier integrity allows bacterial factors to translocate into tumor tissue and to regulate tumor-associated inflammation and tumor proliferation. Translocation of bacterial factors in tumor tissue links CS-induced inflammation with tumor proliferation.


Subject(s)
Carcinoma, Lewis Lung/immunology , Lung Neoplasms/immunology , Smoking/adverse effects , Animals , Bacterial Translocation , Carcinoma, Lewis Lung/microbiology , Carcinoma, Lewis Lung/secondary , Cell Line, Tumor , Cell Proliferation , Female , Humans , Lung Neoplasms/microbiology , Lung Neoplasms/pathology , Mice, Inbred C57BL , Neoplasm Transplantation , Tumor Burden
6.
Oncogene ; 33(10): 1239-48, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-23563178

ABSTRACT

Smoking is the most important risk factor for both lung cancer (LC) and chronic obstructive pulmonary disease. The aim of this study was to investigate the role of myeloid cell nuclear factor-κB in the regulation of tumor cell growth signaling. We subjected mice lacking myeloid RelA/p65 (rela(Δ-/-)) to a metastatic LC model. Cigarette smoke (CS) exposure significantly increased the proliferation of Lewis lung carcinoma cell tumors in wild-type mice. In CS-exposed rela(Δ-/-) mice, the tumor growth was largely inhibited. Transcriptome and pathway analysis of cancer tissue revealed a fundamental impact of myeloid cells on various growth signaling pathways, including the Wnt/ß-catenin pathway. In conclusion, myeloid RelA/p65 is necessary to link smoke-induced inflammation with LC growth and has a role in the activation of Wnt/ß-catenin signaling in tumor cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation , Lung Neoplasms/metabolism , Myeloid Cells/metabolism , Transcription Factor RelA/physiology , Wnt Signaling Pathway , Animals , Carcinoma, Lewis Lung/etiology , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Carcinoma, Non-Small-Cell Lung/etiology , Carcinoma, Non-Small-Cell Lung/secondary , Cell Line, Tumor , Coculture Techniques , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Macrophages, Alveolar/metabolism , Mice , Mice, Knockout , Neoplasm Transplantation , Pneumonia/etiology , Pneumonia/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Smoking/adverse effects , Transcriptome , Tumor Burden , Tumor Necrosis Factor-alpha/metabolism , beta Catenin/metabolism
7.
Oncogene ; 33(21): 2709-16, 2014 May 22.
Article in English | MEDLINE | ID: mdl-23812430

ABSTRACT

Antimicrobial peptides, such as the cathelicidin LL-37/hCAP-18 and its mouse homolog cathelicidin-related antimicrobial peptide (CRAMP), are important effectors of the innate immune system with direct anti-bacterial activity. Cathelicidin is possibly involved in the regulation of tumor cell growth. The aim of this study was to characterize the role of cathelicidin expressed in non-tumorous cells in a preclinical mouse model of tumor growth. Wild-type and CRAMP-deficient animals were exposed to cigarette smoke (CS) and Lewis lung carcinoma cells were injected to initiate the growth of tumors in the lung. CS exposure significantly increased the proliferation of lung tumors in wild-type mice, but not in CRAMP-deficient mice. CS exposure induced the recruitment of myeloid cell into tumor tissue in a CRAMP-dependent manner. Mice lacking RelA/p65 specifically in myeloid cells showed impaired recruitment of CRAMP-positive cells into the lung. In vitro studies with human cells showed that LL-37/hCAP-18 in macrophages is induced by soluble factors derived from cancer cells. Taken together, these data indicate that cathelicidin expressed from myeloid cells promotes CS-induced lung tumor growth by further recruitment of inflammatory cells. The regulation of cathelicidin expression involves myeloid p65/RelA and soluble factor from tumor cells.


Subject(s)
Carcinoma, Lewis Lung/metabolism , Cathelicidins/biosynthesis , Lung Neoplasms/metabolism , Myeloid Cells/metabolism , Animals , Antimicrobial Cationic Peptides , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/secondary , Cell Line, Tumor , Coculture Techniques , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Neoplasm Transplantation , Smoking/adverse effects , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Burden
8.
Allergy ; 68(12): 1487-98, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24215555

ABSTRACT

Deeper insight into pathogenetic pathways and into the biological effects of immunomodulatory agents will help to optimize or adopt therapeutic strategies for atopic disorders. In this article, we highlight selected findings of potential therapeutic relevance that emerged from recent mechanistic studies with focus on molecular and cellular aspects of allergic inflammation. Furthermore, the often complex mechanisms of action of pleiotropic immunomodulatory agents, such as glucocorticoids, vitamin D, or intravenous immunoglobulin (IVIG), are discussed, as their dissection might reveal targets for novel therapeutics or lead to a more rational use of these compounds. Besides reporting novel evidence, this article points to areas of current debate or uncertainty and aims at stimulating scientific discussion and experimental work.


Subject(s)
Hypersensitivity/complications , Inflammation/etiology , Inflammation/therapy , Animals , Basophils/immunology , Basophils/metabolism , Extracellular Space/immunology , Extracellular Space/metabolism , Glucocorticoids/biosynthesis , Humans , Immunoglobulins, Intravenous/therapeutic use , Nucleotides/immunology , Nucleotides/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , Vitamin D/metabolism
9.
Pneumologie ; 67(4): 223-7, 2013 Apr.
Article in German | MEDLINE | ID: mdl-23479400

ABSTRACT

BACKGROUND: The effect of cigarette smoke (CS) on the phagocytosis of alveolar macrophages is discussed controversially on the basis of in vitro experiments. In this short report we describe the in vivo observations that we have performed. METHODS: For this purpose mice were exposed to CS for three consecutive days. One day later the fluorescent microspheres were administered intratracheally and the lung surface was investigated using long-distance fluorescence microscopy. RESULTS: We found that the numbers of neutrophils which engulfed particles was increased in the CS group as compared to controls. The overall phagocytic activity was not significantly different after CS exposure. CONCLUSIONS: In conclusion the phagocytosis of alveolar macrophages and neutrophils after short time CS exposure was not affected. Further investigations will need to look for the effects of long-term CS exposure and the phagocytosis of living bacteria.


Subject(s)
Disease Models, Animal , Macrophages/drug effects , Phagocytosis/drug effects , Pulmonary Alveoli/pathology , Pulmonary Alveoli/physiopathology , Smoking/physiopathology , Tars/pharmacology , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Pulmonary Alveoli/drug effects
10.
Eur Respir J ; 39(2): 411-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21778165

ABSTRACT

Aspergillus fumigatus is a fungal pathogen of major clinical importance. However, little is known about the role of human bronchial epithelial cells (HBECs) during A. fumigatus conidia induced inflammation. Here, we show that differentiated respiratory epithelial cells recognise inactivated resting conidia but not swollen conidia or hyphae, resulting in the induction of the interferon (IFN)-ß signalling pathway and the expression of IFN-ß-inducible genes, such as IFN-γ-inducible protein (IP)-10. This induction was internalisation dependent. We identified double-stranded conidial RNA recognised by Toll-like receptor-3 as a factor responsible for the expression of IFN-ß and IP-10. Inhibition of receptor-interacting protein-1/TANK-binding kinase-1, known to mediate IFN-ß signalling, was sufficient to inhibit the induction of IFN-ß and IP-10 expression by conidia. Even though conidia induced the activation of nuclear factor (NF)-κB in HBECs, IP-10 expression was only partially dependent on NF-κB signalling. These results provide evidence that respiratory cells are activated by the double-stranded RNA of resting conidia and initiate a first immune response to inhaled conidia in an IFN-ß-dependent manner.


Subject(s)
Aspergillosis/metabolism , Aspergillus fumigatus/metabolism , Interferon-beta/metabolism , Respiratory Mucosa/microbiology , Signal Transduction/immunology , Aspergillosis/immunology , Aspergillosis/microbiology , Aspergillus fumigatus/genetics , Aspergillus fumigatus/immunology , Cells, Cultured , Chemokine CXCL10/metabolism , DNA, Fungal/metabolism , Endosomes/immunology , Endosomes/metabolism , Endosomes/microbiology , Humans , Interferon-beta/genetics , NF-kappa B/metabolism , RNA, Messenger/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Spores, Fungal/immunology , Spores, Fungal/metabolism , Tankyrases/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism
11.
Eur Respir J ; 35(2): 343-52, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19679599

ABSTRACT

The pulmonary innate immune system in the respiratory tract eliminates inhaled pathogens. Several cell types contribute to host defense within a complex network. The aim of this study was to evaluate the role of macrophages during pneumonia and in the regulation of the epithelial response to microorganisms. We performed lung infection models in mice lacking myeloid RelA/p65. To study the mechanistic relationships between individual cell types, we applied co-culture models composed of airway epithelial cells (AECs) and macrophages. Mice lacking myeloid RelA/p65 showed significantly decreased bacterial clearance, cytokine expression and neutrophil influx. In addition, the induction of epithelial keratinocyte chemoattractant expression was blunted in these animals. In vitro, AECs were largely insensitive to ligands of Toll-like receptor (TLR)2 or TLR5. Exposure to secretory products of macrophages results in an increased release of pro-inflammatory cytokines and augmented antimicrobial activity. This was associated with increased expression of TLR genes and surface expression of the proteins. Experiments with blocking antibodies showed that the effect of macrophages depends on secreted mediators, including tumour necrosis factor-alpha. In conclusion, the present data show that myeloid RelA is critical for pulmonary host defense. One important mechanism is that macrophages induce the sensitivity of AEC's to microbial patterns.


Subject(s)
Lung/immunology , Myeloid Cells/metabolism , Transcription Factor RelA/metabolism , Animals , Antimicrobial Cationic Peptides/chemistry , Asthma/immunology , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/immunology , Flow Cytometry , Lung/metabolism , Macrophages/metabolism , Mice , Neutrophils/metabolism , Pulmonary Disease, Chronic Obstructive/immunology , Toll-Like Receptors/metabolism
12.
Thorax ; 64(2): 144-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18852155

ABSTRACT

BACKGROUND: Smoking increases the susceptibility to pulmonary infection and is a risk factor for the development of chronic obstructive pulmonary disease (COPD). It is postulated that cigarette smoke suppresses the activation of the innate immune system in response to bacterial infection. METHODS: Using sensitive ex vivo analysis, the level of the endogenous antibiotic peptide human beta-defensin-2 (hBD-2) was measured in pharyngeal washing fluid and sputum from patients with community acquired pneumonia. The regulation of antibacterial host defence molecules was studied in vitro. The effect of cigarette smoke on the antibacterial activity of differentiated airway epithelium and the expression of host defence molecules was studied in an in vitro infection model. RESULTS: Current or former smoking was associated with significantly reduced hBD-2 levels in pharyngeal washing fluid and sputum from patients with acute pneumonia. Exposure of airway epithelium to smoke in vitro inhibited the induction of hBD-2 by bacteria. This correlated with decreased antimicrobial activity. This effect was mimicked by hydrogen peroxide, and catalase blunted the smoke-induced inhibition of epithelial host defence. CONCLUSIONS: Smoke exposure suppresses the induction of epithelial antibacterial host defences. These findings link smoking with increased susceptibility to infection. This mechanism may be important in the pathogenesis of pneumonia and COPD.


Subject(s)
Community-Acquired Infections/immunology , Immunity, Innate/immunology , Pneumonia, Bacterial/immunology , Smoking/immunology , beta-Defensins/metabolism , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/immunology , Epithelial Cells/microbiology , Female , Humans , Hydrogen Peroxide/pharmacology , Immune Tolerance/immunology , Immunity, Mucosal/immunology , Male , Middle Aged , Oxidants/pharmacology , Pharynx/chemistry , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/isolation & purification , Sputum/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...