Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Infect Immun ; 82(3): 1256-67, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24379284

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen that is a major cause of respiratory tract and other nosocomial infections. The sensor kinase CbrA is a central regulator of carbon and nitrogen metabolism and in vitro also regulates virulence-related processes in P. aeruginosa. Here, we investigated the role of CbrA in two murine models of infection. In both peritoneal infections in leukopenic mice and lung infection models, the cbrA mutant was less virulent since substantially larger numbers of cbrA mutant bacteria were required to cause the same level of infection as wild-type or complemented bacteria. In contrast, in the chronic rat lung model the cbrA mutant grew and persisted as well as the wild type, indicating that the decrease of in vivo virulence of the cbrA mutant did not result from growth deficiencies on particular carbon substrates observed in vitro. In addition, a mutant in the cognate response regulator CbrB showed no defect in virulence in the peritoneal infection model, ruling out the involvement of certain alterations of virulence properties in the cbrA mutant including defective swarming motility, increased biofilm formation, and cytotoxicity, since these alterations are controlled through CbrB. Further investigations indicated that the mutant was more susceptible to uptake by phagocytes in vitro, resulting in greater overall bacterial killing. Consistent with the virulence defect, it took a smaller number of Dictyostelium discoideum amoebae to kill the cbrA mutant than to kill the wild type. Transcriptional analysis of the cbrA mutant during D. discoideum infection led to the conclusion that CbrA played an important role in the iron metabolism, protection of P. aeruginosa against oxidative stress, and the regulation of certain virulence factors.


Subject(s)
Bacterial Proteins/genetics , Pseudomonas Infections/genetics , Pseudomonas aeruginosa/genetics , Respiratory Tract Infections/genetics , Respiratory Tract Infections/microbiology , Transcription Factors/genetics , Virulence/genetics , Animals , Bacterial Proteins/metabolism , Dictyostelium/genetics , Dictyostelium/metabolism , Dictyostelium/microbiology , Female , Humans , Lung/metabolism , Lung/microbiology , Macrophages/metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Neutrophils/metabolism , Neutrophils/microbiology , Phagocytes/metabolism , Phagocytes/microbiology , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/metabolism , Rats , Rats, Sprague-Dawley , Respiratory Tract Infections/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
2.
Innate Immun ; 19(3): 315-27, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23109507

ABSTRACT

Innate immunity is triggered by a variety of bacterial molecules, resulting in both protective and potentially harmful pro-inflammatory responses. Further, innate immunity also provides a mechanism for the maintenance of homeostasis between the host immune system and symbiotic or non-pathogenic microorganisms. However, the bacterial factors that mediate these protective effects have been incompletely defined. Here, it was demonstrated that the lantiobiotic nisin Z is able to modulate host immune responses and mediate protective host immunity. Nisin Z induced the secretion of the chemokines MCP-1, IL-8 and Gro-α, and significantly reduced TNF-α induction in response to bacterial LPS in human PBMC. The results correlated with the ability of nisin Z to confer protection against both the Gram-positive organism Staphylococcus aureus, and the Gram-negatives Salmonella enterica sv. Typhimurium and Escherichia coli in murine challenge models. Mechanistic studies revealed that nisin Z modulates host immunity through similar mechanisms as natural host defense peptides, engaging multiple signal transduction pathways and growth factor receptors. The results presented herein demonstrate that, in addition to nisin Z, other bacterial cationic peptides and, in particular, the lantibiotics, could represent a new class of secreted bacterial molecule with immunomodulatory activities.


Subject(s)
Antimicrobial Cationic Peptides/administration & dosage , Escherichia coli/immunology , Nisin/analogs & derivatives , Salmonella enterica/immunology , Staphylococcus aureus/immunology , Animals , Bacterial Load/radiation effects , Cell Line , Chemokines/metabolism , Female , Humans , Immunity, Innate/drug effects , Immunomodulation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nisin/administration & dosage , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
3.
PLoS One ; 7(11): e49123, 2012.
Article in English | MEDLINE | ID: mdl-23145092

ABSTRACT

Pseudomonas aeruginosa PAO1 lon mutants are supersusceptible to ciprofloxacin, and exhibit a defect in cell division and in virulence-related properties, such as swarming, twitching and biofilm formation, despite the fact that the Lon protease is not a traditional regulator. Here we set out to investigate the influence of a lon mutation in a series of infection models. It was demonstrated that the lon mutant had a defect in cytotoxicity towards epithelial cells, was less virulent in an amoeba model as well as a mouse acute lung infection model, and impacted on in vivo survival in a rat model of chronic infection. Using qRT-PCR it was demonstrated that the lon mutation led to a down-regulation of Type III secretion genes. The Lon protease also influenced motility and biofilm formation in a mucin-rich environment. Thus alterations in several virulence-related processes in vitro in a lon mutant were reflected by defective virulence in vivo.


Subject(s)
Ciprofloxacin/pharmacology , Protease La , Pseudomonas Infections , Pseudomonas aeruginosa/pathogenicity , Animals , Cell Division , Disease Models, Animal , Gene Expression Regulation, Bacterial/drug effects , Mice , Mutation , Protease La/genetics , Protease La/metabolism , Pseudomonas Infections/genetics , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Rats , Virulence/genetics
4.
Sci Transl Med ; 4(135): 135ra64, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22623740

ABSTRACT

Case fatality rates for severe malaria remain high even in the best clinical settings because antimalarial drugs act against the parasite without alleviating life-threatening inflammation. We assessed the potential for host-directed therapy of severe malaria of a new class of anti-inflammatory drugs, the innate defense regulator (IDR) peptides, based on host defense peptides. The Plasmodium berghei ANKA model of experimental cerebral malaria was adapted to use as a preclinical screen by combining late-stage intervention in established infections with advanced bioinformatic analysis of early transcriptional changes in co-regulated gene sets. Coadministration of IDR-1018 with standard first-line antimalarials increased survival of infected mice while down-regulating key inflammatory networks associated with fatality. Thus, IDR peptides provided host-directed adjunctive therapy for severe disease in combination with antimalarial treatment.


Subject(s)
Antimalarials/therapeutic use , Antimicrobial Cationic Peptides/therapeutic use , Malaria/drug therapy , Animals , Cells, Cultured , Female , Mice , Mice, Inbred C57BL , Plasmodium berghei/drug effects , Plasmodium berghei/pathogenicity
5.
J Immunol ; 185(2): 1177-85, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20566828

ABSTRACT

In adaptive immunity, Th17 lymphocytes produce the IL-17 and IL-22 cytokines that stimulate mucosal antimicrobial defenses and tissue repair. In this study, we observed that the TLR5 agonist flagellin induced swift and transient transcription of genes encoding IL-17 and IL-22 in lymphoid, gut, and lung tissues. This innate response also temporarily enhanced the expression of genes associated with the antimicrobial Th17 signature. The source of the Th17-related cytokines was identified as novel populations of CD3(neg)CD127(+) immune cells among which CD4-expressing cells resembling lymphoid tissue inducer cells. We also demonstrated that dendritic cells are essential for expression of Th17-related cytokines and so for stimulation of innate cells. These data define that TLR-induced activation of CD3(neg)CD127(+) cells and production of Th17-related cytokines may be crucial for the early defenses against pathogen invasion of host tissues.


Subject(s)
Interleukin-17/immunology , Interleukins/immunology , Mucous Membrane/immunology , Signal Transduction/immunology , Spleen/immunology , Toll-Like Receptor 5/immunology , Animals , CD3 Complex/genetics , CD3 Complex/immunology , CD3 Complex/metabolism , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Flagellin/pharmacology , Flow Cytometry , Gene Expression/drug effects , Gene Expression/immunology , Ileum/drug effects , Ileum/immunology , Ileum/metabolism , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-7 Receptor alpha Subunit/genetics , Interleukin-7 Receptor alpha Subunit/immunology , Interleukin-7 Receptor alpha Subunit/metabolism , Interleukins/genetics , Interleukins/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Mice, Transgenic , Mucous Membrane/cytology , Mucous Membrane/metabolism , Oligonucleotide Array Sequence Analysis , Spleen/cytology , Spleen/metabolism , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/metabolism , Interleukin-22
6.
Eur J Immunol ; 39(6): 1587-96, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19424969

ABSTRACT

Bacterial products (such as endotoxins and flagellin) trigger innate immune responses through TLRs. Flagellin-induced signalling involves TLR5 and MyD88 and, according to some reports, TLR4. Whereas epithelial and dendritic cells are stimulated by flagellin in vitro, the cell contribution to the in vivo response is still unclear. Here, we studied the respective roles of radioresistant and radiosensitive cells in flagellin-induced airway inflammation in mice. We found that i.n. delivery of flagellin elicits a transient change in respiratory function and an acute, pro-inflammatory response in the lungs, characterized by TLR5- and MyD88-dependent chemokine secretion and neutrophil recruitment. In contrast, TLR4, CD14 and TRIF were not essential for flagellin-mediated responses, indicating that TLR4 does not cooperate with TLR5 in the lungs. Respiratory function, chemokine secretion and airway infiltration by neutrophils were dependent on radioresistant, TLR5-expressing cells. Furthermore, lung haematopoietic cells also responded to flagellin by activating TNF-alpha production. We suggest that the radioresistant lung epithelial cells are essential for initiating early, TLR5-dependent signalling in response to flagellin and thus triggering the lung's innate immune responses.


Subject(s)
Epithelial Cells/immunology , Flagellin/immunology , Immunity, Innate/immunology , Radiation Tolerance , Respiratory Mucosa/immunology , Toll-Like Receptor 5/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Administration, Intranasal , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoconstriction/immunology , Cell Movement/genetics , Cell Movement/immunology , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/metabolism , Gene Expression/immunology , Inflammation/immunology , Inflammation/metabolism , Lung/immunology , Lung/metabolism , Lung/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Neutrophils/cytology , Peroxidase/metabolism , Plethysmography, Whole Body , Radiation Chimera/immunology , Radiation Chimera/metabolism , Toll-Like Receptor 5/deficiency , Toll-Like Receptor 5/genetics
7.
J Infect Dis ; 198(1): 115-24, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18462086

ABSTRACT

Toll-like receptor 2 (TLR2) signaling has been shown to contribute to resistance to Listeria monocytogenes infection, as TLR2-deficient mice have a heightened susceptibility to infection with this organism. Because CD14 may associate with TLR2, we investigated the role of CD14 in Listeria responses. In both CD14-deficient and TLR2-deficient macrophages, nuclear factor kappaB translocation; CD40 and CD86; and the production of interleukin (IL)-12, IL-6, tumor necrosis factor, and nitric oxide are reduced. The absence of CD14 augmented susceptibility to Listeria infection, reduced survival, and diminished bacterial clearance, as observed in TLR2-deficient mice. Compared with C57BL/6 control mice, CD14-deficient mice were observed to have a greater number of hepatic microabscesses containing abundant neutrophils, these abscesses were larger in size, and there was reduced inducible nitric oxide synthase expression. Further, mice that are both CD14 deficient and TLR2 deficient display susceptibility to infection that is comparable to that of mice deficient in either CD14 or TLR2 alone. Therefore, the present data demonstrate the role of CD14 and TLR2 in the recognition and control of Listeria infection and host resistance.


Subject(s)
Lipopolysaccharide Receptors/immunology , Listeriosis/immunology , Toll-Like Receptor 2/metabolism , Animals , Immunity, Innate , Interleukin-12 Subunit p40/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharide Receptors/genetics , Listeria monocytogenes , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Specific Pathogen-Free Organisms , Toll-Like Receptor 2/genetics , Tumor Necrosis Factor-alpha/biosynthesis
8.
Int J Exp Pathol ; 88(6): 387-91, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18039275

ABSTRACT

Recent studies on endotoxin/lipopolysaccharide (LPS)-induced acute inflammatory response in the lung are reviewed. The acute airway inflammatory response to inhaled endotoxin is mediated through Toll-like receptor 4 (TLR4) and CD14 signalling as mice deficient for TLR4 or CD14 are unresponsive to endotoxin. Acute bronchoconstriction, tumour necrosis factor (TNF), interleukin (IL)-12 and keratinocyte-derived chemokine (KC) production, protein leak and neutrophil recruitment in the lung are abrogated in mice deficient for the adaptor molecules myeloid differentiation factor 88 (MyD88) and Toll/Interleukin-1 receptor (TIR)-domain-containing adaptor protein (TIRAP), but independent of TIR-domain-containing adaptor-inducing interferon-beta (TRIF). In particular, LPS-induced TNF is required for bronchoconstriction, but dispensable for inflammatory cell recruitment. Lipopolysaccharide induces activation of the p38 mitogen-activated protein kinase (MAPK). Inhibition of pulmonary MAPK activity abrogates LPS-induced TNF production, bronchoconstriction, neutrophil recruitment into the lungs and broncho-alveolar space. In conclusion, TLR4-mediated, bronchoconstriction and acute inflammatory lung pathology to inhaled endotoxin are dependent on TLR4/CD14/MD2 expression using the adapter proteins TIRAP and MyD88, while TRIF, IL-1R1 or IL-18R signalling pathways are dispensable. Further downstream in this axis of signalling, TNF blockade reduces only acute bronchoconstriction, while MAPK inhibition abrogates completely endotoxin-induced inflammation.


Subject(s)
Lung/immunology , MAP Kinase Signaling System , Pneumonia/immunology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Bronchoconstriction , Cytokines/immunology , Enzyme Activation , Humans , Lipopolysaccharides , Mice , Mice, Transgenic , Pneumonia/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
9.
J Immunol ; 179(3): 1872-83, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17641054

ABSTRACT

TNF is a pleiotropic cytokine required for normal development and function of the immune system; however, TNF overexpression also induces inflammation and is associated with autoimmune diseases. TNF exists as both a soluble and a transmembrane protein. Genetic studies in mice have suggested that inflammation in disease models involves soluble TNF (solTNF) and that maintenance of innate immune function involves transmembrane TNF (tmTNF). These findings imply that selective pharmacologic inhibition of solTNF may be anti-inflammatory and yet preserve innate immunity to infection. To address this hypothesis, we now describe dominant-negative inhibitors of TNF (DN-TNFs) as a new class of biologics that selectively inhibits solTNF. DN-TNFs blocked solTNF activity in human and mouse cells, a human blood cytokine release assay, and two mouse arthritis models. In contrast, DN-TNFs neither inhibited the activity of human or mouse tmTNF nor suppressed innate immunity to Listeria infection in mice. These results establish DN-TNFs as the first selective inhibitors of solTNF, demonstrate that inflammation in mouse arthritis models is primarily driven by solTNF, and suggest that the maintenance of tmTNF activity may improve the therapeutic index of future anti-inflammatory agents.


Subject(s)
Arthritis, Experimental/immunology , Immunity, Innate , Inflammation Mediators/physiology , Listeriosis/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/prevention & control , Caspase Inhibitors , Caspases/metabolism , Cell Line, Tumor , Female , Humans , Immunity, Innate/genetics , Inflammation Mediators/metabolism , Interleukin-8/antagonists & inhibitors , Interleukin-8/blood , Interleukin-8/metabolism , Listeriosis/genetics , Listeriosis/pathology , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Paracrine Communication/immunology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Solubility , Tumor Necrosis Factor-alpha/genetics , U937 Cells
10.
Am J Pathol ; 167(6): 1677-87, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16314479

ABSTRACT

Tumor necrosis factor (TNF) plays a critical role in the host response to the intracellular pathogen Listeria monocytogenes (LM). TNF exists in soluble and membrane-bound forms and exhibits both unique and overlapping activities. We examined the role of membrane TNF in the absence of secreted TNF for host resistance in knockin mice in which the endogenous TNF was replaced by a regulated, noncleavable allele (mem-TNF). Macrophages expressing mem-TNF produced nitric oxide and displayed normal bactericidal activity. Although mice completely deficient in TNF (TNF(-/-)) succumbed to LM infection within 4 days, mem-TNF mice controlled LM infection at a low dose (10(4) CFU) but succumbed at a higher dose of infection (10(5) CFU). In contrast to complete TNF deficiency, mem-TNF mice developed confined microabscesses that expressed inducible nitric oxide synthase. The transfer of lymphocytes from immunized mem-TNF, but not TNF(-/-), mice protected TNF(-/-) mice from fatal infection. Taken together the data suggest that in the absence of soluble TNF, the presence of membrane-expressed TNF on phagocytes and lymphocytes partially restores host defense to LM infection.


Subject(s)
Cell Membrane/physiology , Listeria monocytogenes/pathogenicity , Listeriosis/prevention & control , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Membrane/microbiology , Lymphocytes/microbiology , Lymphocytes/physiology , Macrophages/microbiology , Macrophages/physiology , Mice , Mice, Knockout , Mice, Transgenic , Nitric Oxide/physiology , Phagocytes/microbiology , Phagocytes/physiology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...