Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Front Pharmacol ; 12: 682614, 2021.
Article in English | MEDLINE | ID: mdl-34867313

ABSTRACT

Claudins are transmembrane proteins constituting one of three tight junction protein families. In patients with inflammatory bowel disease (IBD), disease activity-dependent changes in expression of certain claudins have been noted, thus making certain claudin family members potential therapy targets. A study was undertaken with the aim of exploring expression of claudins in human disease and two different animal models of IBD: dextrane sulfate sodium-induced colitis and adoptive transfer model of colitis. The expression of sealing claudin-1, claudin-3, claudin-4, and claudin-8, and pore-forming claudin-2 in humans and rodents has been evaluated by immunohistochemistry and quantitative polymerase chain reaction. Claudins were expressed by epithelial and cells of mesodermal origin and were found to be situated at the membrane, within the cytoplasm, or within the nuclei. Claudin expression by human mononuclear cells isolated from lamina propria has been confirmed by Western blot and flow cytometry. The claudin expression pattern in uninflamed and inflamed colon varied between species and murine strains. In IBD and both animal models, diverse alterations in claudin expression by epithelial and inflammatory cells were recorded. Tissue mRNA levels for each studied claudin reflected changes within cell lineage and, at the same time, mirrored the ratio between various cell types. Based on the results of the study, it can be concluded that 1) claudins are not expressed exclusively by epithelial cells, but by certain types of cells of mesodermal origin as well; 2) changes in the claudin mRNA level should be interpreted in the context of overall tissue alterations; and 3) both IBD animal models that were analyzed can be used for investigating claudins as a therapy target, respecting their similarities and differences highlighted in this study.

2.
Antimicrob Agents Chemother ; 60(9): 5337-48, 2016 09.
Article in English | MEDLINE | ID: mdl-27353268

ABSTRACT

As we face an alarming increase in bacterial resistance to current antibacterial chemotherapeutics, expanding the available therapeutic arsenal in the fight against resistant bacterial pathogens causing respiratory tract infections is of high importance. The antibacterial potency of macrolones, a novel class of macrolide antibiotics, against key respiratory pathogens was evaluated in vitro and in vivo MIC values against Streptococcus pneumoniae, Streptococcus pyogenes, Staphylococcus aureus, and Haemophilus influenzae strains sensitive to macrolide antibiotics and with defined macrolide resistance mechanisms were determined. The propensity of macrolones to induce the expression of inducible erm genes was tested by the triple-disk method and incubation in the presence of subinhibitory concentrations of compounds. In vivo efficacy was assessed in a murine model of S. pneumoniae-induced pneumonia, and pharmacokinetic (PK) profiles in mice were determined. The in vitro antibacterial profiles of macrolones were superior to those of marketed macrolide antibiotics, including the ketolide telithromycin, and the compounds did not induce the expression of inducible erm genes. They acted as typical protein synthesis inhibitors in an Escherichia coli transcription/translation assay. Macrolones were characterized by low to moderate systemic clearance, a large volume of distribution, a long half-life, and low oral bioavailability. They were highly efficacious in a murine model of pneumonia after intraperitoneal application even against an S. pneumoniae strain with constitutive resistance to macrolide-lincosamide-streptogramin B antibiotics. Macrolones are the class of macrolide antibiotics with an outstanding antibacterial profile and reasonable PK parameters resulting in good in vivo efficacy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Macrolides/pharmacology , Pneumonia, Pneumococcal/drug therapy , Protein Synthesis Inhibitors/pharmacology , Streptococcus pneumoniae/drug effects , Animals , Anti-Bacterial Agents/pharmacokinetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Disease Models, Animal , Drug Resistance, Bacterial/genetics , Escherichia coli/chemistry , Haemophilus influenzae/drug effects , Haemophilus influenzae/growth & development , Ketolides/pharmacology , Lincosamides/pharmacology , Macrolides/pharmacokinetics , Male , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Pneumonia, Pneumococcal/microbiology , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacokinetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/growth & development , Streptococcus pyogenes/drug effects , Streptococcus pyogenes/growth & development , Streptogramin B/pharmacology , Structure-Activity Relationship
3.
Int Immunopharmacol ; 15(3): 498-504, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23462295

ABSTRACT

Recent reports suggest that azithromycin can shift macrophage polarization towards the alternatively activated M2 phenotype. In order to investigate its immunomodulatory activity in vivo, the influence of azithromycin on survival and cytokine production was assessed in the LPS tolerance model which is characterized by an M2 skewed response. For induction of tolerance, mice received an intraplantar injection of 30 µg LPS, 24 h prior to intravenous challenge with 350 µg LPS. Azithromycin (100 mg/kg) was administered orally, 2 h before LPS application. Influence of treatment on survival and cytokine concentration in serum was monitored. Azithromycin alone, instead of LPS, could not induce an LPS tolerant state. However, when administered before LPS priming it significantly increased survival, which was enhanced by concomitant azithromycin before LPS challenge. Azithromycin had no effect on survival when administered only prior to the LPS challenge. Tolerance induction by LPS priming was associated, upon LPS challenge, with decreased serum concentrations of pro-inflammatory cytokines, TNFα, IL-12p40 and CCL5, and increased serum concentrations of the anti-inflammatory cytokines, IL-10 and IL-1ra. Azithromycin treatment, prior to LPS priming, further reduced serum TNFα and CCL5, yielding the greatest inhibition when the macrolide was also given prior to LPS challenge. Serum concentrations of the anti-inflammatory cytokines, IL-10 and IL-1ra, were unchanged following azithromycin treatment. In summary, we have confirmed the immunomodulatory activity of azithromycin, as reflected in its ability to augment tolerance induction to LPS, promoting increased survival and reduced pro-inflammatory cytokine production, without affecting overt inflammation to LPS or anti-inflammatory cytokine production.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Azithromycin/administration & dosage , Immunomodulation , Animals , Cells, Cultured , Chemokine CCL5/blood , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Immunomodulation/drug effects , Inflammation Mediators/blood , Interleukin 1 Receptor Antagonist Protein/blood , Interleukin-10/blood , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/blood
4.
Inflammation ; 34(5): 471-86, 2011 Oct.
Article in English | MEDLINE | ID: mdl-20872058

ABSTRACT

Inflammatory and antioxidant responses, in male C57Bl6J mice, to single intranasal inoculations with live or heat-killed Streptococcus pneumoniae were studied in order to tease out differences in responses. Heat-killed bacteria elicited weak lung neutrophil infiltration and raised concentrations (peak 6-8 h), in serum or lung tissue, of CXCL1 and 2, tumor necrosis factor alpha (TNFα), interleukin-6 (IL-6), and granulocyte-macrophage-colony stimulating factor, with later increases in CCL2 and IL-1ß. Live bacteria induced profound pulmonary neutrophil infiltration and acute chemokine/cytokine elevations. After 72-96 h, live S. pneumoniae induced a delayed rise in chemokines CXCL2 and CCL2, preceded by increases in TNFα, IL-1ß, and IL-6 and mononuclear infiltration of lungs. With both live and heat-killed bacteria, alveolar epithelial type II cells and alveolar macrophages were the main sources of TNFα and IL-1ß. Only live bacteria caused an acute decrease in lung glutathione peroxidase, an increase in superoxide dismutase, and a sustained increase in serum amyloid protein A. Acute innate immune responses to live and heat-killed S. pneumoniae are similar. In response to live bacteria, inflammation is greater, accompanied by changes in antioxidant enzymes and has an additional, later mononuclear component.


Subject(s)
Antioxidants/metabolism , Inflammation Mediators/metabolism , Pneumonia, Pneumococcal/metabolism , Streptococcus pneumoniae/pathogenicity , Animals , Bacterial Load , Chemokine CXCL1/metabolism , Chemokine CXCL2/metabolism , Cytokines/metabolism , Glutathione Peroxidase/metabolism , Hot Temperature , Immunity, Innate , Interleukin-6/metabolism , Lung/immunology , Lung/metabolism , Lung/microbiology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/pathology , Streptococcus pneumoniae/immunology , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...