Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 14(2): e0213092, 2019.
Article in English | MEDLINE | ID: mdl-30807600

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0096681.].

2.
J Cyst Fibros ; 17(4): 454-461, 2018 07.
Article in English | MEDLINE | ID: mdl-29241629

ABSTRACT

INTRODUCTION: Cystic fibrosis (CF) is a multi-organ disorder characterized by chronic sino-pulmonary infections and inflammation. Many patients with CF suffer from repeated pulmonary exacerbations that are predictors of worsened long-term morbidity and mortality. There are no reliable markers that associate with the onset or progression of an exacerbation or pulmonary deterioration. Previously, we found that the Mirc1/Mir17-92a cluster which is comprised of 6 microRNAs (Mirs) is highly expressed in CF mice and negatively regulates autophagy which in turn improves CF transmembrane conductance regulator (CFTR) function. Therefore, here we sought to examine the expression of individual Mirs within the Mirc1/Mir17-92 cluster in human cells and biological fluids and determine their role as biomarkers of pulmonary exacerbations and response to treatment. METHODS: Mirc1/Mir17-92 cluster expression was measured in human CF and non-CF plasma, blood-derived neutrophils, and sputum samples. Values were correlated with pulmonary function, exacerbations and use of CFTR modulators. RESULTS: Mirc1/Mir17-92 cluster expression was not significantly elevated in CF neutrophils nor plasma when compared to the non-CF cohort. Cluster expression in CF sputum was significantly higher than its expression in plasma. Elevated CF sputum Mirc1/Mir17-92 cluster expression positively correlated with pulmonary exacerbations and negatively correlated with lung function. Patients with CF undergoing treatment with the CFTR modulator Ivacaftor/Lumacaftor did not demonstrate significant change in the expression Mirc1/Mir17-92 cluster after six months of treatment. CONCLUSIONS: Mirc1/Mir17-92 cluster expression is a promising biomarker of respiratory status in patients with CF including pulmonary exacerbation.


Subject(s)
Aminophenols/administration & dosage , Aminopyridines/administration & dosage , Benzodioxoles/administration & dosage , Cystic Fibrosis , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , Quinolones/administration & dosage , Respiratory System , Adolescent , Adult , Biomarkers/metabolism , Chloride Channel Agonists/administration & dosage , Correlation of Data , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Progression , Drug Combinations , Drug Monitoring/methods , Female , Gene Expression Profiling , Humans , Male , RNA, Long Noncoding , Respiratory Function Tests/methods , Respiratory System/drug effects , Respiratory System/metabolism , Respiratory System/physiopathology , Sputum/metabolism
3.
Autophagy ; 12(11): 2026-2037, 2016 11.
Article in English | MEDLINE | ID: mdl-27541364

ABSTRACT

Cystic fibrosis (CF) is a fatal, genetic disorder that critically affects the lungs and is directly caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, resulting in defective CFTR function. Macroautophagy/autophagy is a highly regulated biological process that provides energy during periods of stress and starvation. Autophagy clears pathogens and dysfunctional protein aggregates within macrophages. However, this process is impaired in CF patients and CF mice, as their macrophages exhibit limited autophagy activity. The study of microRNAs (Mirs), and other noncoding RNAs, continues to offer new therapeutic targets. The objective of this study was to elucidate the role of Mirs in dysregulated autophagy-related genes in CF macrophages, and then target them to restore this host-defense function and improve CFTR channel function. We identified the Mirc1/Mir17-92 cluster as a potential negative regulator of autophagy as CF macrophages exhibit decreased autophagy protein expression and increased cluster expression when compared to wild-type (WT) counterparts. The absence or reduced expression of the cluster increases autophagy protein expression, suggesting the canonical inverse relationship between Mirc1/Mir17-92 and autophagy gene expression. An in silico study for targets of Mirs that comprise the cluster suggested that the majority of the Mirs target autophagy mRNAs. Those targets were validated by luciferase assays. Notably, the ability of macrophages expressing mutant F508del CFTR to transport halide through their membranes is compromised and can be restored by downregulation of these inherently elevated Mirs, via restoration of autophagy. In vivo, downregulation of Mir17 and Mir20a partially restored autophagy expression and hence improved the clearance of Burkholderia cenocepacia. Thus, these data advance our understanding of mechanisms underlying the pathobiology of CF and provide a new therapeutic platform for restoring CFTR function and autophagy in patients with CF.


Subject(s)
Autophagy/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Gene Expression Regulation , Macrophages/metabolism , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Antagomirs/pharmacology , Autophagy/drug effects , Autophagy-Related Proteins/metabolism , Burkholderia cenocepacia/physiology , Cells, Cultured , Cystic Fibrosis/microbiology , Gene Expression Regulation/drug effects , Homozygote , Lung/metabolism , Lung/microbiology , Lung/pathology , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , NIH 3T3 Cells
4.
Am J Physiol Lung Cell Mol Physiol ; 308(11): L1136-44, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25840995

ABSTRACT

As the eighth leading cause of annual mortality in the USA, influenza A viruses are a major public health concern. In 20% of patients, severe influenza progresses to acute lung injury (ALI). However, pathophysiological mechanisms underlying ALI development are poorly defined. We reported that, unlike wild-type (WT) C57BL/6 controls, influenza A virus-infected mice that are heterozygous for the F508del mutation in the cystic fibrosis transmembrane conductance regulator (HETs) did not develop ALI. This effect was associated with higher IL-6 and alveolar macrophages (AMs) at 6 days postinfection (d.p.i.) in HET bronchoalveolar lavage fluid (BALF). In the present study, we found that HET AMs were an important source of IL-6 at 6 d.p.i. Infection also induced TGF-ß production by HET but not WT mice at 2 d.p.i. TGF-ß neutralization at 2 d.p.i. (TGF-N) significantly reduced BALF IL-6 in HETs at 6 d.p.i. Neither TGF-N nor IL-6 neutralization at 4 d.p.i. (IL-6-N) altered postinfection weight loss or viral replication in either mouse strain. However, both treatments increased influenza A virus-induced hypoxemia, pulmonary edema, and lung dysfunction in HETs to WT levels at 6 d.p.i. TGF-N and IL-6-N did not affect BALF AM and neutrophil numbers but attenuated the CXCL-1/keratinocyte chemokine response in both strains and reduced IFN-γ production in WT mice. Finally, bone marrow transfer experiments showed that HET stromal and myeloid cells are both required for protection from ALI in HETs. These findings indicate that TGF-ß-dependent production of IL-6 by AMs later in infection prevents ALI development in influenza A virus-infected HET mice.


Subject(s)
Acute Lung Injury/virology , Influenza A virus/immunology , Interleukin-6/physiology , Orthomyxoviridae Infections/immunology , Transforming Growth Factor beta/physiology , Acute Lung Injury/immunology , Animals , Bronchoalveolar Lavage Fluid , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Immunity, Innate , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/virology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred CFTR , Sequence Deletion
5.
PLoS One ; 9(5): e96681, 2014.
Article in English | MEDLINE | ID: mdl-24798083

ABSTRACT

Burkholderia cenocepacia is a virulent pathogen that causes significant morbidity and mortality in patients with cystic fibrosis (CF), survives intracellularly in macrophages, and uniquely causes systemic infections in CF. Autophagy is a physiologic process that involves engulfing non-functional organelles and proteins and delivering them for lysosomal degradation, but also plays a role in eliminating intracellular pathogens, including B. cenocepacia. Autophagy is defective in CF but can be stimulated in murine CF models leading to increased clearance of B. cenocepacia, but little is known about autophagy stimulation in human CF macrophages. IFN-γ activates macrophages and increases antigen presentation while also inducing autophagy in macrophages. We therefore, hypothesized that treatment with IFN-γ would increase autophagy and macrophage activation in patients with CF. Peripheral blood monocyte derived macrophages (MDMs) were obtained from CF and non-CF donors and subsequently infected with B. cenocepacia. Basal serum levels of IFN-γ were similar between CF and non-CF patients, however after B. cenocepacia infection there is deficient IFN-γ production in CF MDMs. IFN-γ treated CF MDMs demonstrate increased co-localization with the autophagy molecule p62, increased autophagosome formation, and increased trafficking to lysosomes compared to untreated CF MDMs. Electron microscopy confirmed IFN-γ promotes double membrane vacuole formation around bacteria in CF MDMs, while only single membrane vacuoles form in untreated CF cells. Bacterial burden is significantly reduced in autophagy stimulated CF MDMs, comparable to non-CF levels. IL-1ß production is decreased in CF MDMs after IFN-γ treatment. Together, these results demonstrate that IFN-γ promotes autophagy-mediated clearance of B. cenocepacia in human CF macrophages.


Subject(s)
Autophagy , Burkholderia Infections/immunology , Burkholderia cenocepacia , Cystic Fibrosis/immunology , Interferon-gamma/immunology , Macrophages/immunology , Adolescent , Adult , Azithromycin/chemistry , Child , Cystic Fibrosis/microbiology , Female , Humans , Interleukin-1beta/immunology , Lysosomes/metabolism , Macrophages/microbiology , Male , Microscopy, Confocal , Monocytes/immunology , Monocytes/microbiology , Phagosomes , Young Adult
6.
Biochem Biophys Res Commun ; 424(2): 221-7, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22728038

ABSTRACT

Cystic fibrosis (CF) is accompanied with heightened inflammation worsened by drug resistant Burkholderia cenocepacia. Human CF macrophage responses to B. cenocepacia are poorly characterized and variable in the literature. Therefore, we examined human macrophage responses to the epidemic B. cenocepacia J2315 strain in order to identify novel anti-inflammatory targets. Peripheral blood monocyte derived macrophages were obtained from 23 CF and 27 non-CF donors. Macrophages were infected with B. cenocepacia J2315 and analyzed for cytokines, cytotoxicity, and microscopy. CF macrophages demonstrated significant increases in IL-1ß, IL-10, MCP-1, and IFN-γ production in comparison to non-CF controls. CF patients on prednisone exhibited globally diminished cytokines compared to controls and other CF patients. CF macrophages also displayed increased bacterial burden and cell death. In conclusion, CF macrophages demonstrate exaggerated IL-1ß, IL-10, MCP-1, and IFN-γ production and cell death during B. cenocepacia infection. Treatment with corticosteroids acutely suppressed cytokine responses.


Subject(s)
Burkholderia Infections/microbiology , Burkholderia cenocepacia , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Macrophages/immunology , Adolescent , Adrenal Cortex Hormones/pharmacology , Adult , Apoptosis , Chemokine CCL2/metabolism , Child , Child, Preschool , Cystic Fibrosis/immunology , Female , Humans , Infant , Inflammation/microbiology , Inflammation/pathology , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Lysosomes/microbiology , Macrophages/drug effects , Macrophages/ultrastructure , Male , Microscopy, Electron , Middle Aged , Young Adult
7.
Infect Immun ; 80(4): 1390-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22311922

ABSTRACT

The mechanisms by which Streptococcus pneumoniae obtains carbohydrates for growth during airway colonization remain to be elucidated. The low concentration of free carbohydrates in the normal human airway suggests that pneumococci must utilize complex glycan structures for growth. The glycosaminoglycan hyaluronic acid is present on the apical surface of airway epithelial cells. As pneumococci express a hyaluronate lyase (Hyl) that cleaves hyaluronic acid into disaccharides, we hypothesized that during colonization pneumococci utilize the released carbohydrates for growth. Hyaluronic acid supported significant pneumococcal growth in an hyl-dependent manner. A phosphoenolpyruvate-dependent phosphotransferase system (PTS) and an unsaturated glucuronyl hydrolase (Ugl) encoded downstream of hyl are also essential for growth on hyaluronic acid. This genomic arrangement is present in several other organisms, suggesting conservation of the utilization mechanism between species. In vivo experiments support the hypothesis that S. pneumoniae utilizes hyaluronic acid as a carbon source during colonization. We also demonstrate that pneumococci can utilize the hyaluronic acid capsule of other bacterial species for growth, suggesting an alternative carbohydrate source for pneumococcal growth. Together, these data support a novel function for pneumococcal degradation of hyaluronic acid in vivo and provide mechanistic details of growth on this glycosaminoglycan.


Subject(s)
Hyaluronic Acid/metabolism , Streptococcus pneumoniae/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Glycoside Hydrolases/metabolism , Humans , Mice , Phosphotransferases/metabolism , Polysaccharide-Lyases/metabolism , Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/growth & development
8.
Int J Cancer ; 125(9): 2198-204, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19623660

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is activated in a variety of human cancers, including ovarian cancer. The molecular mechanism by which the STAT3 is activated in cancer cells is poorly understood. We observed that human ovarian xenograft tumors (A2780) in mice were severely hypoxic (pO(2) approximately 2 mmHg). We further observed that hypoxic exposure significantly increased the phosphorylation of STAT3 (pSTAT3) at the Tyr705 residue in A2780 cell line. The pSTAT3 (Tyr705) level was highly dependent on cellular oxygenation levels, with a significant increase at <2% O(2), and without any change in the pSTAT3 (Ser727) or total STAT3 levels. The pSTAT3 (Tyr705) elevation following hypoxic exposure could be reversed within 12 hr after returning the cells to normoxia. The increased level of pSTAT3 was partly mediated by increased levels of reactive oxygen species generation in the hypoxic cancer cells. Conventional chemotherapeutic drugs cisplatin and taxol were far less effective in eliminating the hypoxic ovarian cancer cells suggesting a role for pSTAT3 in cellular resistance to chemotherapy. Inhibition of STAT3 by AG490 followed by treatment with cisplatin or taxol resulted in a significant increase in apoptosis suggesting that hypoxia-induced STAT3 activation is responsible for chemoresistance. The results have important clinical implications for the treatment of hypoxic ovarian tumors using STAT3-specific inhibitors.


Subject(s)
Cell Hypoxia , Ovarian Neoplasms/drug therapy , STAT3 Transcription Factor/physiology , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Female , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Oxygen/pharmacology , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...