Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Front Microbiol ; 12: 697232, 2021.
Article in English | MEDLINE | ID: mdl-34276631

ABSTRACT

Neisseria meningitidis is a gram-negative bacterium that often asymptomatically colonizes the human nasopharyngeal tract. These bacteria cross the epithelial barrier can cause life-threatening sepsis and/or meningitis. Antimicrobial peptides are one of the first lines of defense against invading bacterial pathogens. Human beta-defensin 2 (hBD2) is an antimicrobial peptide with broad antibacterial activity, although its mechanism of action is poorly understood. Here, we investigated the effect of hBD2 on N. meningitidis. We showed that hBD2 binds to and kills actively growing meningococcal cells. The lethal effect was evident after 2 h incubation with the peptide, which suggests a slow killing mechanism. Further, the membrane integrity was not changed during hBD2 treatment. Incubation with lethal doses of hBD2 decreased the presence of diplococci; the number and size of bacterial microcolonies/aggregates remained constant, indicating that planktonic bacteria may be more susceptible to the peptide. Meningococcal DNA bound hBD2 in mobility shift assays and inhibited the lethal effect of hBD2 in a dose-dependent manner both in suspension and biofilms, supporting the interaction between hBD2 and DNA. Taken together, the ability of meningococcal DNA to bind hBD2 opens the possibility that extracellular DNA due to bacterial lysis may be a means of N. meningitidis to evade immune defenses.

2.
Article in English | MEDLINE | ID: mdl-33468461

ABSTRACT

Antimicrobial peptides (AMPs) play an important role in the defense against pathogens by targeting and killing invading microbes. Some pathogenic bacteria have been shown to negatively regulate AMP expression, while several commensals may induce AMP expression. The expression of certain AMPs, such as human beta-defensin 2 (hBD2), can be induced via nuclear factor NF-κB, which, in turn, is negatively controlled by tumor necrosis factor alpha-induced protein 3 (TNFAIP3, or A20). In this work, we examined the expression of hBD1 and hBD2 during coincubation of pharyngeal epithelial cells with pathogenic Neisseria meningitidis and commensal lactobacilli. The Lactobacillus strains induced hBD2 expression in human pharyngeal cells, while the pathogen N. meningitidis did not. In coincubation experiments, meningococci were able to dampen the AMP expression induced by lactobacilli. We found that N. meningitidis induced the NF-κB inhibitor A20. Further, RNA silencing of A20 resulted in increased hBD2 expression after meningococcal infection. Since it is known that induction of A20 reduces NF-κB activity and thus hBD2 levels, meningococcal-mediated A20 induction could be a way for the pathogen to dampen AMP expression. Finally, treatment of N. meningitidis and lactobacilli with synthetic hBD2 reduced N. meningitidis viability more efficiently than Lactobacillus reuteri, explaining why maintaining low AMP levels is important for the survival of the pathogen.


Subject(s)
Neisseria meningitidis , beta-Defensins , Epithelial Cells , Humans , Lactobacillus , NF-kappa B/genetics , Neisseria meningitidis/genetics , beta-Defensins/genetics
3.
Front Microbiol ; 10: 422, 2019.
Article in English | MEDLINE | ID: mdl-30891026

ABSTRACT

Neisseria meningitidis is a Gram-negative bacterium that asymptomatically colonizes the human nasopharyngeal mucosa. Pilus-mediated initial adherence of N. meningitidis to the epithelial mucosa is followed by the formation of three-dimensional aggregates, called microcolonies. Dispersal from microcolonies contributes to the transmission of N. meningitidis across the epithelial mucosa. We have recently discovered that environmental concentrations of host cell-derived lactate influences N. meningitidis microcolony dispersal. Here, we examined the ability of N. meningitidis mutants deficient in lactate metabolism to form biofilms. A lactate dehydrogenease A (ldhA) mutant had an increased level of biofilm formation. Deletion of ldhA increased the N. meningitidis cell surface hydrophobicity and aggregation. In this study, we used FAM20, which belongs to clonal complex ST-11 that forms biofilms independently of extracellular DNA (eDNA). However, treatment with DNase I abolished the increased biofilm formation and aggregation of the ldhA-deficient mutant, suggesting a critical role for eDNA. Compared to wild-type, the ldhA-deficient mutant exhibited an increased autolytic rate, with significant increases in the eDNA concentrations in the culture supernatants and in biofilms. Within the ldhA mutant biofilm, the transcription levels of the capsule, pilus, and bacterial lysis genes were downregulated, while norB, which is associated with anaerobic respiration, was upregulated. These findings suggest that the absence of ldhA in N. meningitidis promotes biofilm formation and aggregation through autolysis-mediated DNA release.

4.
Article in English | MEDLINE | ID: mdl-28180113

ABSTRACT

The essential first step in bacterial colonization is adhesion to the host epithelial cells. The early host-responses post-bacterial adhesions are still poorly understood. Early growth response 1 (EGR1) is an early response transcriptional regulator that can be rapidly induced by various environmental stimuli. Several bacteria can induce EGR1 expression in host cells, but the involved bacterial characteristics and the underlying molecular mechanisms of this response are largely unknown. Here, we show that EGR1 can be induced in host epithelial cells by different species of bacteria independent of the adherence level, Gram-staining type and pathogenicity. However, bacterial viability and contact with host cells is necessary, indicating that an active interaction between bacteria and the host is important. Furthermore, the strongest response is observed in cells originating from the natural site of the infection, suggesting that the EGR1 induction is cell type specific. Finally, we show that EGFR-ERK1/2 and ß1-integrin signaling are the main pathways used for bacteria-mediated EGR1 upregulation. In conclusion, the increase of EGR1 expression in epithelial cells is a common stress induced, cell type specific response upon host-bacteria interaction that is mediated by EGFR-ERK1/2 and ß1-integrin signaling.


Subject(s)
Bacteria/immunology , Early Growth Response Protein 1/biosynthesis , Early Growth Response Protein 2/metabolism , Epithelial Cells/immunology , Epithelial Cells/microbiology , Host-Pathogen Interactions , MAP Kinase Signaling System , Cell Line , Humans , Integrin beta1/metabolism , Stress, Physiological , Transcription, Genetic
5.
Int J Antimicrob Agents ; 45(5): 447-54, 2015 May.
Article in English | MEDLINE | ID: mdl-25532742

ABSTRACT

Antimicrobial peptides (AMPs) are a key component of the immune system and are expressed by a large variety of organisms. AMPs are capable of eliminating a broad range of micro-organisms, illustrated by murine models where lack of AMP expression resulted in enhanced susceptibility to infection. Despite the importance of AMPs in immune defences, it is not clear whether a change in AMP expression is pathogen-specific or reflects a general response to groups of pathogens. Furthermore, it is unclear how the evoked change in AMP expression affects the host. To fully exploit the therapeutic potential of AMPs - by direct application of peptides or by using AMP-inducers - it is crucial to gain an insight into the complexity involved in pathogen-mediated regulation of AMP expression. This review summarises current knowledge on how AMP expression is affected by pathogens. In addition, the relevance and specificity of these changes in AMPs during infection will be discussed.


Subject(s)
Antimicrobial Cationic Peptides/biosynthesis , Communicable Diseases/immunology , Gene Expression Regulation , Host-Pathogen Interactions , Immunity, Innate , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...