Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 13(4): e0192709, 2018.
Article in English | MEDLINE | ID: mdl-29677209

ABSTRACT

The molecular mechanisms underlying susceptibility to severe respiratory syncytial virus (RSV) infection remain poorly understood. Herein, we report on the role of osteopontin (OPN) in regulation of RSV infection in human epithelial cells and how interleukin-1 beta (IL-1ß), a cytokine secreted soon after RSV infection, when persistently expressed can induce OPN expression leading to increased viral infection. We first compared OPN expression in two human epithelial cell lines: HEK-293 and HEp-2. In contrast to HEp-2, HEK-293 expresses low levels of pro-caspase-1 resulting in decreased IL-1ß expression in response to RSV infection. We found a correlation between low IL-1ß levels and a delay in induction of OPN expression in RSV-infected HEK-293 cells compared to HEp-2. This phenomenon could partially explain the high susceptibility of HEp-2 cells to RSV infection versus the moderate susceptibility of HEK-293 cells. Also, HEK-293 cells expressing low levels of pro-caspase-1 exhibit decreased IL-1ß expression and delayed OPN expression in response to RSV infection. HEK-293 cells incubated with human rIL-1ß showed a dose-dependent increase in OPN expression upon RSV infection. Also, incubation with rOPN increased RSV viral load. Moreover, HEp-2 cells or mice infected with a mucogenic RSV strain RSV-L19F showed elevated levels of OPN in contrast to mice infected with the laboratory RSV strain rA2. This correlated with elevated levels of OPN following infection with RSV-L19F compared to rA2. Together, these results demonstrate that increased OPN expression is regulated in part by IL-1ß, and the interplay between IL-1ß and OPN signaling may play a pivotal role in the spread of RSV infection.


Subject(s)
Host-Pathogen Interactions/physiology , Osteopontin/metabolism , Respiratory Syncytial Virus Infections/etiology , Animals , Female , Gene Expression Regulation , HEK293 Cells , Humans , Hyaluronan Receptors/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Mice, Knockout , Osteopontin/genetics , Osteopontin/pharmacology , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/physiology , Viral Load , Virus Replication
2.
PLoS One ; 11(10): e0163709, 2016.
Article in English | MEDLINE | ID: mdl-27695127

ABSTRACT

Respiratory syncytial virus (RSV) has been reported to infect human mesenchymal stem cells (MSCs) but the consequences are poorly understood. MSCs are present in nearly every organ including the nasal mucosa and the lung and play a role in regulating immune responses and mediating tissue repair. We sought to determine whether RSV infection of MSCs enhances their immune regulatory functions and contributes to RSV-associated lung disease. RSV was shown to replicate in human MSCs by fluorescence microscopy, plaque assay, and expression of RSV transcripts. RSV-infected MSCs showed differentially altered expression of cytokines and chemokines such as IL-1ß, IL6, IL-8 and SDF-1 compared to epithelial cells. Notably, RSV-infected MSCs exhibited significantly increased expression of IFN-ß (~100-fold) and indoleamine-2,3-dioxygenase (IDO) (~70-fold) than in mock-infected MSCs. IDO was identified in cytosolic protein of infected cells by Western blots and enzymatic activity was detected by tryptophan catabolism assay. Treatment of PBMCs with culture supernatants from RSV-infected MSCs reduced their proliferation in a dose dependent manner. This effect on PBMC activation was reversed by treatment of MSCs with the IDO inhibitors 1-methyltryptophan and vitamin K3 during RSV infection, a result we confirmed by CRISPR/Cas9-mediated knockout of IDO in MSCs. Neutralizing IFN-ß prevented IDO expression and activity. Treatment of MSCs with an endosomal TLR inhibitor, as well as a specific inhibitor of the TLR3/dsRNA complex, prevented IFN-ß and IDO expression. Together, these results suggest that RSV infection of MSCs alters their immune regulatory function by upregulating IFN-ß and IDO, affecting immune cell proliferation, which may account for the lack of protective RSV immunity and for chronicity of RSV-associated lung diseases such as asthma and COPD.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Interferon-beta/biosynthesis , Mesenchymal Stem Cells/immunology , Respiratory Syncytial Virus Infections/immunology , Cell Proliferation/genetics , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/immunology , Epithelial Cells/immunology , Epithelial Cells/virology , Fetal Blood/immunology , Fetal Blood/virology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-beta/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-1beta/biosynthesis , Interleukin-1beta/immunology , Interleukin-8/biosynthesis , Interleukin-8/immunology , Leukocytes, Mononuclear , Lung/immunology , Lung/pathology , Lung/virology , Mesenchymal Stem Cells/virology , Microscopy, Fluorescence , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/pathogenicity
3.
J Virol ; 86(3): 1832-43, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22090136

ABSTRACT

Respiratory syncytial virus (RSV) is one of the major causes of respiratory infections in children, and it is the main pathogen causing bronchiolitis in infants. The binding and entry mechanism by which RSV infects respiratory epithelial cells has not yet been determined. In this study, the earliest stages of RSV infection in normal human bronchial epithelial cells were probed by tracking virions with fluorescent lipophilic dyes in their membranes. Virions colocalized with cholesterol-containing plasma membrane microdomains, identified by their ability to bind cholera toxin subunit B. Consistent with an important role for cholesterol in RSV infection, cholesterol depletion profoundly inhibited RSV infection, while cholesterol repletion reversed this inhibition. Merger of the outer leaflets of the viral envelope and the cell membrane appeared to be triggered at these sites. Using small-molecule inhibitors, RSV infection was found to be sensitive to Pak1 inhibition, suggesting the requirement of a subsequent step of cytoskeletal reorganization that could involve plasma membrane rearrangements or endocytosis. It appears that RSV entry depends on its ability to dock to cholesterol-rich microdomains (lipid rafts) in the plasma membrane where hemifusion events begin, assisted by a Pak1-dependent process.


Subject(s)
Bronchi/metabolism , Cholesterol/metabolism , Membrane Fusion , Respiratory Syncytial Viruses/physiology , Bronchi/cytology , Cell Line , Epithelial Cells/metabolism , Flow Cytometry , Fluorescent Dyes , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...