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










Publication year range
1.
Cell Rep ; 43(5): 114153, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38687643

ABSTRACT

Gut-draining mesenteric and celiac lymph nodes (mLNs and celLNs) critically contribute to peripheral tolerance toward food and microbial antigens by supporting the de novo induction of regulatory T cells (Tregs). These tolerogenic properties of mLNs and celLNs are stably imprinted within stromal cells (SCs) by microbial signals and vitamin A (VA), respectively. Here, we report that a single, transient gastrointestinal infection in the neonatal, but not adult, period durably abrogates the efficient Treg-inducing capacity of celLNs by altering the subset composition and gene expression profile of celLNSCs. These cells carry information about the early-life pathogen encounter until adulthood and durably instruct migratory dendritic cells entering the celLN with reduced tolerogenic properties. Mechanistically, transiently reduced VA levels cause long-lasting celLN functional impairment, which can be rescued by early-life treatment with VA. Together, our data highlight the therapeutic potential of VA to prevent sequelae post gastrointestinal infections in infants.


Subject(s)
Lymph Nodes , T-Lymphocytes, Regulatory , Vitamin A , Animals , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymph Nodes/drug effects , Vitamin A/pharmacology , Vitamin A/therapeutic use , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Mice , Animals, Newborn , Immune Tolerance/drug effects , Dendritic Cells/immunology , Mice, Inbred C57BL , Female
2.
J Exp Med ; 221(3)2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38305765

ABSTRACT

The intestinal epithelium is the first line of defense against enteric pathogens. Removal of infected cells by exfoliation prevents mucosal translocation and systemic infection in the adult host, but is less commonly observed in the neonatal intestine. Instead, here, we describe non-professional efferocytosis of Salmonella-infected enterocytes by neighboring epithelial cells in the neonatal intestine. Intestinal epithelial stem cell organoid cocultures of neonatal and adult cell monolayers with damaged enterocytes replicated this observation, confirmed the age-dependent ability of intestinal epithelial cells for efferocytosis, and identified the involvement of the "eat-me" signals and adaptors phosphatidylserine and C1q as well as the "eat-me" receptors integrin-αv (CD51) and CD36 in cellular uptake. Consistent with this, massive epithelial cell membrane protrusions and CD36 accumulation at the contact site with apoptotic cells were observed in the infected neonatal host in vivo. Efferocytosis of infected small intestinal enterocytes by neighboring epithelial cells may represent a previously unrecognized mechanism of neonatal antimicrobial host defense to maintain barrier integrity.


Subject(s)
Efferocytosis , Intestines , Epithelial Cells , Intestinal Mucosa/metabolism , Salmonella
3.
Infect Immun ; 90(2): e0022221, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34978927

ABSTRACT

Hypoxia-inducible transcription factor 1 (HIF-1) has been shown to enhance microbial killing and ameliorate the course of bacterial infections. While the impact of HIF-1 on inflammatory diseases of the gut has been studied intensively, its function in bacterial infections of the gastrointestinal tract remains largely elusive. With the help of a publicly available gene expression data set, we inferred significant activation of HIF-1 after oral infection of mice with Salmonella enterica serovar Typhimurium. Immunohistochemistry and Western blot analyses confirmed marked HIF-1α protein stabilization, especially in the intestinal epithelium. This prompted us to analyze conditional Hif1a-deficient mice to examine cell type-specific functions of HIF-1 in this model. Our results demonstrate enhanced noncanonical induction of HIF-1 activity upon Salmonella infection in the intestinal epithelium as well as in macrophages. Surprisingly, Hif1a deletion in intestinal epithelial cells did not impact inflammatory gene expression, bacterial spread, or disease outcomes. In contrast, Hif1a deletion in myeloid cells enhanced intestinal Cxcl2 expression and reduced the cecal Salmonella load. In vitro, HIF-1α-deficient macrophages showed overall impaired transcription of mRNA encoding proinflammatory factors; however, the intracellular survival of Salmonella was not impacted by HIF-1α deficiency.


Subject(s)
Salmonella Infections , Salmonella typhimurium , Animals , Epithelial Cells/microbiology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intestinal Mucosa/microbiology , Macrophages , Mice , Salmonella Infections/genetics , Salmonella typhimurium/genetics
4.
Adv Exp Med Biol ; 1117: 111-129, 2019.
Article in English | MEDLINE | ID: mdl-30980356

ABSTRACT

Antimicrobial peptides (AMPs) are in the focus of scientific research since the 1990s. In most cases, the main aim was laid on the design of AMP to kill bacteria effectively, with particular emphasis on broadband action and independency on antibiotic resistance. However, so far no approved drug on the basis of AMP has entered the market.Our approach of constructing AMP, called synthetic anti-lipopolysaccharide peptides (SALPs), on the basis of inhibiting the inflammatory action of lipopolysaccharide (LPS, endotoxin) from Gram-negative bacteria was focused on the neutralization of the decisive toxins. These are, beside LPS from Gram-negative bacteria, the lipoproteins (LP) from Gram-positive origin. Although some of the SALPs have an antibacterial action, the most important property is the high-affinity binding to LPS and LP, whether as constituent of the bacteria or in free form which prevents the damaging inflammation, that could otherwise lead to life-threatening septic shock. Most importantly, the SALP may inhibit inflammation independently of the resistance status of the bacteria, and so far the repeated use of the peptides apparently does not cause resistance of the attacking pathogens.In this chapter, an overview is given over the variety of possible applications in the field of fighting against severe bacterial infections, from the use in systemic infection/inflammation up to various topical applications such as anti-biofilm action and severe skin and soft tissue infections.


Subject(s)
Anti-Bacterial Agents/chemistry , Pathogen-Associated Molecular Pattern Molecules/antagonists & inhibitors , Peptides/chemistry , Bacterial Infections/drug therapy , Endotoxins , Gram-Negative Bacteria , Humans , Lipopolysaccharides
5.
Nature ; 563(7731): E25, 2018 11.
Article in English | MEDLINE | ID: mdl-30158704

ABSTRACT

In Fig. 1d of this Letter, the third group along should have been labelled 'WT' rather than 'Tlr5'. This has been corrected online.

6.
Nature ; 560(7719): 489-493, 2018 08.
Article in English | MEDLINE | ID: mdl-30089902

ABSTRACT

Alterations in enteric microbiota are associated with several highly prevalent immune-mediated and metabolic diseases1-3, and experiments involving faecal transplants have indicated that such alterations have a causal role in at least some such conditions4-6. The postnatal period is particularly critical for the development of microbiota composition, host-microbe interactions and immune homeostasis7-9. However, the underlying molecular mechanisms of this neonatal priming period have not been defined. Here we report the identification of a host-mediated regulatory circuit of bacterial colonization that acts solely during the early neonatal period but influences life-long microbiota composition. We demonstrate age-dependent expression of the flagellin receptor Toll-like receptor 5 (TLR5) in the gut epithelium of neonate mice. Using competitive colonization experiments, we demonstrate that epithelial TLR5-mediated REG3γ production is critical for the counter-selection of colonizing flagellated bacteria. Comparative microbiota transfer experiments in neonate and adult wild-type and Tlr5-deficient germ-free mice reveal that neonatal TLR5 expression strongly influences the composition of the microbiota throughout life. Thus, the beneficial microbiota in the adult host is shaped during early infancy. This might explain why environmental factors that disturb the establishment of the microbiota during early life can affect immune homeostasis and health in adulthood.


Subject(s)
Aging/immunology , Animals, Newborn/immunology , Gastrointestinal Microbiome/immunology , Toll-Like Receptor 5/immunology , Aging/genetics , Animals , Animals, Newborn/genetics , Crosses, Genetic , Environment , Female , Flagellin/immunology , Flagellin/metabolism , Gastrointestinal Microbiome/genetics , Homeostasis , Host Microbial Interactions , Housing, Animal , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Male , Mice , Mice, Inbred C57BL , Pregnancy , Toll-Like Receptor 5/genetics
7.
PLoS Pathog ; 14(3): e1006925, 2018 03.
Article in English | MEDLINE | ID: mdl-29522566

ABSTRACT

Effector molecules translocated by the Salmonella pathogenicity island (SPI)1-encoded type 3 secretion system (T3SS) critically contribute to the pathogenesis of human Salmonella infection. They facilitate internalization by non-phagocytic enterocytes rendering the intestinal epithelium an entry site for infection. Their function in vivo has remained ill-defined due to the lack of a suitable animal model that allows visualization of intraepithelial Salmonella. Here, we took advantage of our novel neonatal mouse model and analyzed various bacterial mutants and reporter strains as well as gene deficient mice. Our results demonstrate the critical but redundant role of SopE2 and SipA for enterocyte invasion, prerequisite for transcriptional stimulation and mucosal translocation in vivo. In contrast, the generation of a replicative intraepithelial endosomal compartment required the cooperative action of SipA and SopE2 or SipA and SopB but was independent of SopA or host MyD88 signaling. Intraepithelial growth had no critical influence on systemic spread. Our results define the role of SPI1-T3SS effector molecules during enterocyte invasion and intraepithelial proliferation in vivo providing novel insight in the early course of Salmonella infection.


Subject(s)
Bacterial Proteins/metabolism , Enterocytes/microbiology , Intestinal Mucosa/microbiology , Myeloid Differentiation Factor 88/physiology , Salmonella Infections/microbiology , Salmonella typhimurium/pathogenicity , Type III Secretion Systems/metabolism , Animals , Bacterial Proteins/genetics , Cell Proliferation , Enterocytes/metabolism , Enterocytes/pathology , Genetic Complementation Test , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Salmonella Infections/metabolism , Signal Transduction , Type III Secretion Systems/genetics
8.
PLoS Pathog ; 12(5): e1005616, 2016 05.
Article in English | MEDLINE | ID: mdl-27159323

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) represents a major causative agent of infant diarrhea associated with significant morbidity and mortality in developing countries. Although studied extensively in vitro, the investigation of the host-pathogen interaction in vivo has been hampered by the lack of a suitable small animal model. Using RT-PCR and global transcriptome analysis, high throughput 16S rDNA sequencing as well as immunofluorescence and electron microscopy, we characterize the EPEC-host interaction following oral challenge of newborn mice. Spontaneous colonization of the small intestine and colon of neonate mice that lasted until weaning was observed. Intimate attachment to the epithelial plasma membrane and microcolony formation were visualized only in the presence of a functional bundle forming pili (BFP) and type III secretion system (T3SS). Similarly, a T3SS-dependent EPEC-induced innate immune response, mediated via MyD88, TLR5 and TLR9 led to the induction of a distinct set of genes in infected intestinal epithelial cells. Infection-induced alterations of the microbiota composition remained restricted to the postnatal period. Although EPEC colonized the adult intestine in the absence of a competing microbiota, no microcolonies were observed at the small intestinal epithelium. Here, we introduce the first suitable mouse infection model and describe an age-dependent, virulence factor-dependent attachment of EPEC to enterocytes in vivo.


Subject(s)
Disease Models, Animal , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Host-Pathogen Interactions/physiology , Animals , Animals, Newborn , Disease Susceptibility/microbiology , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Fimbriae, Bacterial/ultrastructure , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Oligonucleotide Array Sequence Analysis , Type III Secretion Systems/metabolism , Virulence Factors/metabolism
9.
Gut ; 65(2): 225-37, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25887379

ABSTRACT

OBJECTIVES: Dysbiosis of the intestinal microbiota is associated with Crohn's disease (CD). Functional evidence for a causal role of bacteria in the development of chronic small intestinal inflammation is lacking. Similar to human pathology, TNF(deltaARE) mice develop a tumour necrosis factor (TNF)-driven CD-like transmural inflammation with predominant ileal involvement. DESIGN: Heterozygous TNF(deltaARE) mice and wildtype (WT) littermates were housed under conventional (CONV), specific pathogen-free (SPF) and germ-free (GF) conditions. Microbial communities were analysed by high-throughput 16S ribosomal RNA gene sequencing. Metaproteomes were measured using LC-MS. Temporal and spatial resolution of disease development was followed after antibiotic treatment and transfer of microbial communities into GF mice. Granulocyte infiltration and Paneth cell function was assessed by immunofluorescence and gene expression analysis. RESULTS: GF-TNF(deltaARE) mice were free of inflammation in the gut and antibiotic treatment of CONV-TNF(deltaARE) mice attenuated ileitis but not colitis, demonstrating that disease severity and location are microbiota-dependent. SPF-TNF(deltaARE) mice developed distinct ileitis-phenotypes associated with gradual loss of antimicrobial defence. 16S analysis and metaproteomics revealed specific compositional and functional alterations of bacterial communities in inflamed mice. Transplantation of disease-associated but not healthy microbiota transmitted CD-like ileitis to GF-TNF(deltaARE) recipients and triggered loss of lysozyme and cryptdin-2 expression. Monoassociation of GF-TNF(deltaARE) mice with the human CD-related Escherichia coli LF82 did not induce ileitis. CONCLUSIONS: We provide clear experimental evidence for the causal role of gut bacterial dysbiosis in the development of chronic ileal inflammation with subsequent failure of Paneth cell function.


Subject(s)
Crohn Disease/etiology , Dysbiosis/complications , Ileitis/etiology , Intestines/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Colitis , Fluoroimmunoassay , Germ-Free Life , Ileitis/microbiology , Inflammation/physiopathology , Mice , Microbiota/physiology , Tumor Necrosis Factor-alpha
10.
Cell Microbiol ; 17(11): 1561-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26294173

ABSTRACT

A single layer of epithelial cells separates the intestinal lumen from the underlying sterile tissue. It is exposed to a multitude of nutrients and a large number of commensal bacteria. Although the presence of commensal bacteria significantly contributes to nutrient digestion, vitamin synthesis and tissue maturation, their high number represents a permanent challenge to the integrity of the epithelial surface keeping the local immune system constantly on alert. In addition, the intestinal mucosa is challenged by a variety of enteropathogenic microorganisms. In both circumstances, the epithelium actively contributes to maintaining host-microbial homeostasis and antimicrobial host defence. It deploys a variety of mechanisms to restrict the presence of commensal bacteria to the intestinal lumen and to prevent translocation of commensal and pathogenic microorganisms to the underlying tissue. Enteropathogenic microorganisms in turn have learnt to evade the host's immune system and circumvent the antimicrobial host response. In the present article, we review recent advances that illustrate the intense and intimate host-microbial interaction at the epithelial level and improve our understanding of the mechanisms that maintain the integrity of the intestinal epithelial barrier.


Subject(s)
Gastrointestinal Microbiome , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Intestinal Mucosa/physiology , Animals , Humans
11.
Front Immunol ; 6: 45, 2015.
Article in English | MEDLINE | ID: mdl-25713575

ABSTRACT

According to the NHS, it is estimated that over 50% of the adult population are, to some extent, affected by gum disease and approximately 15% of UK population have been diagnosed with severe periodontitis. Periodontitis, a chronic polymicrobial disease of the gums, causes inflammation in its milder form, whereas in its severe form affects the surrounding tissues and can result in tooth loss. During periodontitis, plaque accumulates and sits between the junctional epithelium and the tooth itself, resulting in inflammation and the formation of a periodontal pocket. An interface is formed directly between the subgingival bacteria and the junctional epithelial cells. Bacterial pathogens commonly associated with periodontal disease are, among others, Porphyromonas gingivalis, Tannerella forsythia, and Treponema denticola, together known as the "red complex." This review will mostly concentrate on the role of P. gingivalis, a Gram-negative anaerobic bacterium and one of the major and most studied contributors of this disease. Because periodontal disease is associated with the development of atherosclerosis, it is important to understand the local immune response to P. gingivalis. Innate immune players, in particular, complement and antimicrobial peptides and their effects with regard to P. gingivalis during periodontitis and in the development of atherosclerosis will be presented.

12.
Gut ; 64(2): 222-32, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24811998

ABSTRACT

OBJECTIVE: Antimicrobial peptides (AMP) provide protection from infection by pathogenic microorganisms and restrict bacterial growth at epithelial surfaces to maintain mucosal homeostasis. In addition, they exert a significant anti-inflammatory activity. Here we analysed the anatomical distribution and biological activity of an orally administered AMP in the context of bacterial infection and host-microbial homeostasis. DESIGN: The anatomical distribution as well as antibacterial and anti-inflammatory activity of the endogenous AMP cryptdin 2 and the synthetic peptide Pep19-2.5 at the enteric mucosal surface were analysed by immunostaining, functional viability and stimulation assays, an oral Salmonella enterica subsp. enterica sv. Typhimurium (S. Typhimurium) model and comparative microbiota analysis. RESULTS: Endogenous cryptdin 2 was found attached to bacteria of the enteric microbiota within the intestinal mucus layer. Similarly, the synthetic peptide Pep19-2.5 attached rapidly to bacterial cells, exhibited a marked affinity for the intestinal mucus layer in vivo, altered the structural organisation of endotoxin in a mucus matrix and demonstrated potent anti-inflammatory and antibacterial activity. Oral Pep19-2.5 administration induced significant changes in the composition of the enteric microbiota as determined by high-throughput 16S rDNA sequencing. This may have contributed to the only transient improvement of the clinical symptoms after oral infection with S. Typhimurium. CONCLUSIONS: Our findings demonstrate the anti-inflammatory activity and mucus affinity of the synthetic AMP Pep19-2.5 and characterise the influence on microbiota composition and enteropathogen infection after oral administration.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacokinetics , Intestinal Mucosa/metabolism , Peptide Fragments/pharmacokinetics , Administration, Oral , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Anti-Inflammatory Agents/administration & dosage , Cells, Cultured , Defensins , Drug Evaluation, Preclinical/methods , Female , Host-Pathogen Interactions/physiology , Humans , Intestinal Mucosa/microbiology , Mice, Inbred C57BL , Microbiota/drug effects , Mucus/metabolism , Mucus/microbiology , Peptide Fragments/administration & dosage , Peptide Fragments/therapeutic use , Proteins/metabolism , Salmonella Infections/drug therapy , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella typhimurium/drug effects , Salmonella typhimurium/metabolism , Salmonella typhimurium/physiology
13.
Gut Microbes ; 5(6): 761-5, 2014.
Article in English | MEDLINE | ID: mdl-25483327

ABSTRACT

The intestinal mucosa squares the circle by allowing efficient nutrient absorption while generating a firm barrier toward the enteric microbiota, enteropathogenic microorganisms and high luminal concentrations of potent immunostimulatory molecules. The mucus layer together with local antimicrobial and anti-inflammatory peptides significantly contribute to this ability. Here we summarize the recent progress made to better understand the critical importance of this dynamic, complex and highly structured anti-inflammatory and antimicrobial barrier.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Intestinal Mucosa/immunology , Microbiota , Animals , Bacterial Physiological Phenomena , Humans , Intestinal Mucosa/microbiology
14.
PLoS Pathog ; 10(9): e1004385, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25210785

ABSTRACT

The coordinated action of a variety of virulence factors allows Salmonella enterica to invade epithelial cells and penetrate the mucosal barrier. The influence of the age-dependent maturation of the mucosal barrier for microbial pathogenesis has not been investigated. Here, we analyzed Salmonella infection of neonate mice after oral administration. In contrast to the situation in adult animals, we observed spontaneous colonization, massive invasion of enteroabsorptive cells, intraepithelial proliferation and the formation of large intraepithelial microcolonies. Mucosal translocation was dependent on enterocyte invasion in neonates in the absence of microfold (M) cells. It further resulted in potent innate immune stimulation in the absence of pronounced neutrophil-dominated pathology. Our results identify factors of age-dependent host susceptibility and provide important insight in the early steps of Salmonella infection in vivo. We also present a new small animal model amenable to genetic manipulation of the host for the analysis of the Salmonella enterocyte interaction in vivo.


Subject(s)
Enterocytes/microbiology , Epithelial Cells/microbiology , Macrophages/microbiology , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/pathogenicity , Virulence/immunology , Age Factors , Animals , Animals, Newborn , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Enterocytes/immunology , Enterocytes/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Gene Expression Regulation, Bacterial , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/pathology
15.
Med Microbiol Immunol ; 203(4): 257-71, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24728387

ABSTRACT

Streptococcus pneumoniae and Listeria monocytogenes, pathogens which can cause severe infectious disease in human, were used to infect properdin-deficient and wildtype mice. The aim was to deduce a role for properdin, positive regulator of the alternative pathway of complement activation, by comparing and contrasting the immune response of the two genotypes in vivo. We show that properdin-deficient and wildtype mice mounted antipneumococcal serotype-specific IgM antibodies, which were protective. Properdin-deficient mice, however, had increased survival in the model of streptococcal pneumonia and sepsis. Low activity of the classical pathway of complement and modulation of FcγR2b expression appear to be pathogenically involved. In listeriosis, however, properdin-deficient mice had reduced survival and a dendritic cell population that was impaired in maturation and activity. In vitro analyses of splenocytes and bone marrow-derived myeloid cells support the view that the opposing outcomes of properdin-deficient and wildtype mice in these two infection models is likely to be due to a skewing of macrophage activity to an M2 phenotype in the properdin-deficient mice. The phenotypes observed thus appear to reflect the extent to which M2- or M1-polarised macrophages are involved in the immune responses to S. pneumoniae and L. monocytogenes. We conclude that properdin controls the strength of immune responses by affecting humoral as well as cellular phenotypes during acute bacterial infection and ensuing inflammation.


Subject(s)
Listeria monocytogenes/immunology , Properdin/immunology , Sepsis/immunology , Sepsis/pathology , Streptococcus pneumoniae/immunology , Animals , Disease Models, Animal , Mice, Inbred C57BL , Properdin/deficiency , Sepsis/microbiology , Survival Analysis
16.
Antimicrob Agents Chemother ; 57(3): 1480-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23318793

ABSTRACT

Bacterial infections are known to cause severe health-threatening conditions, including sepsis. All attempts to get this disease under control failed in the past, and especially in times of increasing antibiotic resistance, this leads to one of the most urgent medical challenges of our times. We designed a peptide to bind with high affinity to endotoxins, one of the most potent pathogenicity factors involved in triggering sepsis. The peptide Pep19-2.5 reveals high endotoxin neutralization efficiency in vitro, and here, we demonstrate its antiseptic/anti-inflammatory effects in vivo in the mouse models of endotoxemia, bacteremia, and cecal ligation and puncture, as well as in an ex vivo model of human tissue. Furthermore, we show that Pep19-2.5 can bind and neutralize not only endotoxins but also other bacterial pathogenicity factors, such as those from the Gram-positive bacterium Staphylococcus aureus. This broad neutralization efficiency and the additive action of the peptide with common antibiotics makes it an exceptionally appropriate drug candidate against bacterial sepsis and also offers multiple other medication opportunities.


Subject(s)
Lipopolysaccharides/antagonists & inhibitors , Peptides/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Virulence Factors/antagonists & inhibitors , Amino Acid Sequence , Animals , Anti-Bacterial Agents/pharmacology , Bacteremia/drug therapy , Bacteremia/metabolism , Bacteremia/microbiology , Bacteremia/mortality , Disease Models, Animal , Drug Synergism , Endotoxemia/drug therapy , Endotoxemia/metabolism , Endotoxemia/microbiology , Endotoxemia/mortality , Female , Humans , Lipopolysaccharides/biosynthesis , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptides/chemical synthesis , Sepsis/drug therapy , Sepsis/metabolism , Sepsis/microbiology , Sepsis/mortality , Staphylococcal Infections/drug therapy , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/mortality , Staphylococcus aureus/growth & development , Survival Analysis , Virulence Factors/biosynthesis
17.
EMBO Mol Med ; 4(12): 1308-19, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23143987

ABSTRACT

Intestinal ischemia/reperfusion (I/R) injury causes inflammation and tissue damage and is associated with high morbidity and mortality. Uncontrolled activation of the innate immune system through toll-like receptors (Tlr) plays a key role in I/R-mediated tissue damage but the underlying mechanisms have not been fully resolved. Here, we identify post-transcriptional upregulation of the essential Tlr signalling molecule interleukin 1 receptor-associated kinase (Irak) 1 as the causative mechanism for post-ischemic immune hyper-responsiveness of intestinal epithelial cells. Increased Irak1 protein levels enhanced epithelial ligand responsiveness, chemokine secretion, apoptosis and mucosal barrier disruption in an experimental intestinal I/R model using wild-type, Irak1(-/-) and Tlr4(-/-) mice and ischemic human intestinal tissue. Irak1 accumulation under hypoxic conditions was associated with reduced K48 ubiquitination and enhanced Senp1-mediated deSUMOylation of Irak1. Importantly, administration of microRNA (miR)-146a or induction of miR-146a by the phytochemical diindolylmethane controlled Irak1 upregulation and prevented immune hyper-responsiveness in mouse and human tissue. These findings indicate that Irak1 accumulation triggers I/R-induced epithelial immune hyper-responsiveness and suggest that the induction of miR-146a offers a promising strategy to prevent I/R tissue injury.


Subject(s)
Down-Regulation , Interleukin-1 Receptor-Associated Kinases/genetics , Intestine, Small/metabolism , Ischemia/genetics , MicroRNAs/metabolism , Reperfusion Injury/genetics , Animals , Humans , Interleukin-1 Receptor-Associated Kinases/metabolism , Intestine, Small/blood supply , Intestine, Small/immunology , Ischemia/metabolism , Mice , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Signal Transduction
18.
Curr Drug Targets ; 13(9): 1131-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22664073

ABSTRACT

The bacterial cell wall represents the primary target for antimicrobial agents. Microbial destruction is accompanied by the release of potent immunostimulatory membrane constituents. Both Gram-positive and Gram-negative bacteria release a variety of lipoproteins and peptidoglycan fragments. Gram-positive bacteria additionally provide lipoteichoic acids, whereas Gram-negative bacteria also release lipopolysaccharide (LPS, endotoxin), essential component of the outer leaflet of the bacterial cell wall and one of the most potent immunostimulatory molecules known. Immune activation therefore can be considered as an adverse effect of antimicrobial destruction and killing during anti-infective treatment. In contrast to antibiotics, the use of cationic amphiphilic antimicrobial peptides allows both effective bacterial killing and inhibition of the immunostimulatory effect of the released bacterial membrane constituents. The administration of antimicrobial peptides alone or in combination with antibiotic agents thus represents a novel strategy in the antiinfective treatment with potentially important beneficial aspects. Here, data are presented which describe immunological and clinical aspects of the use of antimicrobial peptides (AMPs) as therapeutic agents to treat bacterial infection and neutralize the immunostimulatory activity of released cell wall constituents.


Subject(s)
Adjuvants, Immunologic/pharmacology , Anti-Infective Agents/pharmacology , Bacteria/drug effects , Cell Wall/drug effects , Bacteria/immunology , Bacteria/metabolism , Cell Wall/immunology , Cell Wall/metabolism , Humans
19.
Curr Drug Targets ; 13(9): 1121-30, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22664072

ABSTRACT

The first barrier that an antimicrobial agent must overcome when interacting with its target is the microbial cell wall. In the case of Gram-negative bacteria, additional to the cytoplasmic membrane and the peptidoglycan layer, an outer membrane (OM) is the outermost barrier. The OM has an asymmetric distribution of the lipids with phospholipids and lipopolysaccharide (LPS) located in the inner and outer leaflets, respectively. In contrast, Gram-positive bacteria lack OM and possess a much thicker peptidoglycan layer compared to their Gram-negative counterparts. An additional class of amphiphiles exists in Gram-positives, the lipoteichoic acids (LTA), which may represent important structural components. These long molecules cross-bridge the entire cell envelope with their lipid component inserting into the outer leaflet of the cytoplasmic membrane and the teichoic acid portion penetrating into the peptidoglycan layer. Furthermore, both classes of bacteria have other important amphiphiles, such as lipoproteins, whose importance has become evident only recently. It is not known yet whether any of these amphiphilic components are able to stimulate the immune system under physiological conditions as constituents of intact bacteria. However, all of them have a very high pro-inflammatory activity when released from the cell. Such a release may take place through the interaction with the immune system, or with antibiotics (particularly with those targeting cell wall components), or simply by the bacterial division. Therefore, a given antimicrobial agent must ideally have a double character, namely, it must overcome the bacterial cell wall barrier, without inducing the liberation of the pro-inflammatory amphiphiles. Here, new data are presented which describe the development and use of membrane-active antimicrobial agents, in particular antimicrobial peptides (AMPs) and lipopolyamines. In this way, essential progress was achieved, in particular with respect to the inhibition of deleterious consequences of bacterial infections such as severe sepsis and septic shock.


Subject(s)
Anti-Infective Agents/pharmacology , Bacteria/chemistry , Cell Wall/chemistry , Polyamines/pharmacology , Amino Acid Sequence , Bacterial Proteins/chemistry , Molecular Sequence Data
20.
J Immunol ; 180(5): 3313-8, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18292556

ABSTRACT

Properdin is a positive regulator of complement activation so far known to be instrumental in the survival of infections with certain serotypes of Neisseria meningitidis. We have generated a fully backcrossed properdin-deficient mouse line by conventional gene-specific targeting. In vitro, properdin-deficient serum is impaired in alternative pathway-dependent generation of complement fragment C3b when activated by Escherichia coli DH5alpha. Properdin-deficient mice and wild-type littermates compare in their levels of C3 and IgM. In an in vivo model of polymicrobial septic peritonitis induced by sublethal cecal ligation and puncture, properdin-deficient mice appear immunocompromised, because they are significantly impaired in their survival compared with wild-type littermates. We further show that properdin localizes to mast cells and that properdin has the ability to directly associate with E. coli DH5alpha. We conclude that properdin plays a significant role in the outcome of polymicrobial sepsis.


Subject(s)
Peritonitis/immunology , Peritonitis/prevention & control , Properdin/physiology , Sepsis/immunology , Sepsis/prevention & control , Animals , Cecum , Complement C3/metabolism , Complement Pathway, Alternative/immunology , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/prevention & control , Escherichia coli K12/immunology , Female , Ligation , Male , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Peritonitis/metabolism , Properdin/deficiency , Properdin/genetics , Punctures , Sepsis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...