Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Cyst Fibros ; 22(4): 756-762, 2023 07.
Article in English | MEDLINE | ID: mdl-37211502

ABSTRACT

Short chain fatty acids (SCFA) are produced by anaerobic bacteria. The most common SCFAs are acetate, propionate and butyrate. SCFAs have been implicated in several inflammatory diseases including cystic fibrosis (CF) where they are present in the airways at millimolar concentrations. Staphylococcus aureus is one of the main respiratory pathogens in CF. Polymorphonuclear neutrophil granulocytes (PMN) represent the most important immune defense the host uses against S. aureus. However, the reason why PMNs are unable to clear S. aureus in CF remains largely unclear. We hypothesized that SCFAs impair effector functions of PMNs in response to S. aureus. To test this, human PMNs were exposed to CF clinical isolates of S. aureus in vitro in the presence or absence of SCFAs and effector functions of PMNs were assessed. Our data show that SCFAs do not affect the viability of PMNs and do not stimulate the release of neutrophil extracellular traps (NET) from human PMNs. Production of reactive oxygen species (ROS), another important antimicrobial function of PMNs, on the other hand, was significantly inhibited by SCFAs in response to the bacterium. SCFAs did not compromise the ability of PMNs to kill CF isolates of S. aureus in vitro. Overall, our results provide new knowledge into the interactions between SCFAs and the immune system, and indicate that SCFAs produced by anaerobic bacteria in the CF lung could interfere with reactive oxidant production of PMNs in response to S. aureus, one of the prominent respiratory pathogens in this disease.


Subject(s)
Cystic Fibrosis , Staphylococcal Infections , Humans , Neutrophils , Cystic Fibrosis/microbiology , Staphylococcus aureus , Respiratory Burst , Fatty Acids, Volatile , Staphylococcal Infections/microbiology
2.
Front Immunol ; 12: 772288, 2021.
Article in English | MEDLINE | ID: mdl-34912341

ABSTRACT

Seasonal influenza epidemics represent a significant global health threat. The exacerbated immune response triggered by respiratory influenza virus infection causes severe pulmonary damage and contributes to substantial morbidity and mortality. Regulator of G-protein signaling 10 (RGS10) belongs to the RGS protein family that act as GTPase activating proteins for heterotrimeric G proteins to terminate signaling pathways downstream of G protein-coupled receptors. While RGS10 is highly expressed in immune cells, in particular monocytes and macrophages, where it has strong anti-inflammatory effects, its physiological role in the respiratory immune system has not been explored yet. Here, we show that Rgs10 negatively modulates lung immune and inflammatory responses associated with severe influenza H1N1 virus respiratory infection in a mouse model. In response to influenza A virus challenge, mice lacking RGS10 experience enhanced weight loss and lung viral titers, higher mortality and significantly faster disease onset. Deficiency of Rgs10 upregulates the levels of several proinflammatory cytokines and chemokines and increases myeloid leukocyte accumulation in the infected lung, markedly neutrophils, monocytes, and inflammatory monocytes, which is associated with more pronounced lung damage. Consistent with this, influenza-infected Rgs10-deficent lungs contain more neutrophil extracellular traps and exhibit higher neutrophil elastase activities than wild-type lungs. Overall, these findings propose a novel, in vivo role for RGS10 in the respiratory immune system controlling myeloid leukocyte infiltration, viral clearance and associated clinical symptoms following lethal influenza challenge. RGS10 also holds promise as a new, potential therapeutic target for respiratory infections.


Subject(s)
Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections/immunology , Pneumonia/immunology , RGS Proteins/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Cytokines/immunology , Female , Lung/immunology , Lung/pathology , Lung/virology , Male , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Pneumonia/pathology , Pneumonia/virology , RGS Proteins/genetics
3.
Front Immunol ; 12: 745326, 2021.
Article in English | MEDLINE | ID: mdl-34621276

ABSTRACT

Cystic Fibrosis (CF) is a genetic disease that causes chronic and severe lung inflammation and infection associated with high rates of mortality. In CF, disrupted ion exchange in the epithelium results in excessive mucus production and reduced mucociliary clearance, leading to immune system exacerbation and chronic infections with pathogens such as P. aeruginosa and S. aureus. Constant immune stimulation leads to altered immune responses including T cell impairment and neutrophil dysfunction. Specifically, CF is considered a Th17-mediated disease, and it has been proposed that both P. aeruginosa and a subset of neutrophils known as granulocytic myeloid suppressor cells (gMDSCs) play a role in T cell suppression. The exact mechanisms behind these interactions are yet to be determined, but recent works demonstrate a role for arginase-1. It is also believed that P. aeruginosa drives gMDSC function as a means of immune evasion, leading to chronic infection. Herein, we review the current literature regarding immune suppression in CF by gMDSCs with an emphasis on T cell impairment and the role of P. aeruginosa in this dynamic interaction.


Subject(s)
Cystic Fibrosis/immunology , Granulocytes/immunology , Immune Evasion , Myeloid-Derived Suppressor Cells/immunology , Pseudomonas aeruginosa/immunology , Th17 Cells/immunology , Arginase/physiology , Cystic Fibrosis/complications , Cytotoxicity, Immunologic , Humans , Neutrophils/immunology , Neutrophils/pathology , Persistent Infection , Pseudomonas Infections/complications , Pseudomonas Infections/immunology , T-Lymphocytes, Regulatory/immunology
4.
Cell Signal ; 86: 110099, 2021 10.
Article in English | MEDLINE | ID: mdl-34339853

ABSTRACT

Regulator of G-protein signaling 10 (RGS10) is a member of the superfamily of RGS proteins that canonically act as GTPase activating proteins (GAPs). RGS proteins accelerate GTP hydrolysis on the G-protein α subunits and result in termination of signaling pathways downstream of G protein-coupled receptors. Beyond its GAP function, RGS10 has emerged as an anti-inflammatory protein by inhibiting LPS-mediated NF-κB activation and expression of inflammatory cytokines, in particular TNF-α. Although RGS10 is abundantly expressed in resting macrophages, previous studies have shown that RGS10 expression is suppressed in macrophages following Toll-like receptor 4 (TLR4) activation by LPS. However, the molecular mechanism by which LPS induces Rgs10 silencing has not been clearly defined. The goal of the current study was to determine whether LPS silences Rgs10 expression through an NF-κB-mediated proinflammatory mechanism in pulmonary macrophages, a unique type of innate immune cells. We demonstrate that Rgs10 transcript and RGS10 protein levels are suppressed upon LPS treatment in the murine MH-S alveolar macrophage cell line. We show that pharmacological inhibition of PI3K/ NF-κB/p300 (NF-κB co-activator)/TNF-α signaling cascade and the activities of HDAC (1-3) enzymes block LPS-induced silencing of Rgs10 in MH-S cells as well as microglial BV2 cells and BMDMs. Further, loss of RGS10 generated by using CRISPR/Cas9 amplifies NF-κB phosphorylation and inflammatory gene expression following LPS treatment in MH-S cells. Together, our findings strongly provide critical insight into the molecular mechanism underlying RGS10 suppression by LPS in pulmonary macrophages.


Subject(s)
NF-kappa B , RGS Proteins , Animals , Cytokines/metabolism , GTP-Binding Proteins/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/metabolism , Mice , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , RGS Proteins/genetics , RGS Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Pathogens ; 10(6)2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34200034

ABSTRACT

Cystic fibrosis (CF) airway disease is characterized by chronic microbial infections and infiltration of inflammatory polymorphonuclear (PMN) granulocytes. Staphylococcus aureus (S. aureus) is a major lung pathogen in CF that persists despite the presence of PMNs and has been associated with CF lung function decline. While PMNs represent the main mechanism of the immune system to kill S. aureus, it remains largely unknown why PMNs fail to eliminate S. aureus in CF. The goal of this study was to observe how the CF airway environment affects S. aureus killing by PMNs. PMNs were isolated from the blood of healthy volunteers and CF patients. Clinical isolates of S. aureus were obtained from the airways of CF patients. The results show that PMNs from healthy volunteers were able to kill all CF isolates and laboratory strains of S. aureus tested in vitro. The extent of killing varied among strains. When PMNs were pretreated with supernatants of CF sputum, S. aureus killing was significantly inhibited suggesting that the CF airway environment compromises PMN antibacterial functions. CF blood PMNs were capable of killing S. aureus. Although bacterial killing was inhibited with CF sputum, PMN binding and phagocytosis of S. aureus was not diminished. The S. aureus-induced respiratory burst and neutrophil extracellular trap release from PMNs also remained uninhibited by CF sputum. In summary, our data demonstrate that the CF airway environment limits killing of S. aureus by PMNs and provides a new in vitro experimental model to study this phenomenon and its mechanism.

6.
Int J Mol Sci ; 22(4)2021 Feb 20.
Article in English | MEDLINE | ID: mdl-33672733

ABSTRACT

Sigma factor C (SigC) contributes to Mycobacterium tuberculosis virulence in various animal models, but the stress response coordinated by this transcription factor was undefined. The results presented here indicate that SigC prevents copper starvation. Whole genome expression studies demonstrate short-term (4-h) induction of sigC, controlled from a tetracycline-inducible promoter, upregulates ctpB and genes in the nonribosomal peptide synthase (nrp) operon. These genes are expressed at higher levels after 48-h sigC induction, but also elevated are genes encoding copper-responsive regulator RicR and RicR-regulated copper toxicity response operon genes rv0846-rv0850, suggesting prolonged sigC induction results in excessive copper uptake. No growth and global transcriptional differences are observed between a sigC null mutant relative to its parent strain in 7H9 medium. In a copper-deficient medium, however, growth of the sigC deletion strain lags the parent, and 40 genes (including those in the nrp operon) are differentially expressed. Copper supplementation reverses the growth defect and silences most transcriptional differences. Together, these data support SigC as a transcriptional regulator of copper acquisition when the metal is scarce. Attenuation of sigC mutants in severe combined immunodeficient mice is consistent with an inability to overcome innate host defenses that sequester copper ions to deprive invading microbes of this essential micronutrient.


Subject(s)
Copper/pharmacology , Immunity/drug effects , Mycobacterium tuberculosis/metabolism , Sigma Factor/metabolism , Animals , Bacterial Proteins/metabolism , Biological Transport/drug effects , Copper Sulfate/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation, Bacterial/drug effects , Mice, SCID , Microbial Viability/drug effects , Mutation/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Phenotype , Transcription, Genetic/drug effects , Virulence/drug effects , Virulence/genetics
7.
BMC Immunol ; 22(1): 7, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33478382

ABSTRACT

BACKGROUND: Neutrophils are key components of the exacerbated inflammation and tissue damage in cystic fibrosis (CF) airways. Neutrophil extracellular traps (NETs) trap and kill extracellular pathogens. While NETs are abundant in the airways of CF patients and have been hypothesized to contribute to lung damage in CF, the in vivo role of NETs remains controversial, partially due to lack of appropriate animal models. The goal of this study was to detect NETs and to further characterize neutrophil-mediated inflammation in the airways of mice overexpressing the epithelial sodium channel (ßENaC-Tg mice on C57BL/6 background) in their lung with CF-like airway disease, in the absence of any apparent bacterial infections. METHODS: Histology scoring of lung tissues, flow cytometry, multiplex ELISA, immunohistochemistry and immunofluorescence were used to characterize NETs and the airway environment in uninfected, ßENaC-Tg mice at 6 and 8 weeks of age, the most chronic time points so far studied in this model. RESULTS: Excessive neutrophilic infiltration characterized the lungs of uninfected, ßENaC-Tg mice at 6 and 8 weeks of age. The bronchoalveolar lavage fluid (BALF) of ßENaC-Tg mice contains increased levels of CF-associated cytokines and chemokines: KC, MIP-1α/ß, MCP-1, G-CSF, IL-5, and IL-6. The BALF of ßENaC-Tg mice contain MPO-DNA complexes, indicative of the presence of NETs. Immunofluorescence and flow cytometry of BALF neutrophils and lung tissues demonstrated increased histone citrullination, a NET-specific marker, in ßENaC-Tg mice. CONCLUSIONS: NETs are detected in the airways of ßENaC-Tg mice, in the absence of bacterial infections. These data demonstrate the usefulness of the ßENaC-Tg mouse to serve as a model for studying the role of NETs in chronic CF airway inflammation.


Subject(s)
Cystic Fibrosis/immunology , Epithelial Sodium Channels/immunology , Extracellular Traps/immunology , Lung/immunology , Neutrophils/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Citrullination , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cytokines/metabolism , Epithelial Sodium Channels/genetics , Histones/metabolism , Inflammation , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neutrophil Infiltration
SELECTION OF CITATIONS
SEARCH DETAIL
...