Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 217
Filter
2.
J Intern Med ; 285(6): 635-652, 2019 06.
Article in English | MEDLINE | ID: mdl-30584680

ABSTRACT

It is now well recognized that cardiovascular events (CVE) occur quite commonly, both in the acute phase and in the long-term, in patients with community-acquired pneumonia (CAP). CVE have been noted in up to 30% of patients hospitalized with all-cause CAP. One systematic review and meta-analysis of hospitalized patients with all-cause CAP noted that the incidence rates for overall cardiac events were 17.7%, for incident heart failure were 14.1%, for acute coronary syndromes were 5.3% and for incident cardiac arrhythmias were 4.7%. In the case of pneumococcal CAP, almost 20% of patients studied had one or more of these cardiac events. Recent research has provided insights into the pathogenesis of the acute cardiac events occurring in pneumococcal infections. With respect to the former, key involvements of the major pneumococcal protein virulence factor, pneumolysin, are now well documented, whilst systemic platelet-driven neutrophil activation may also contribute. However, events involved in the pathogenesis of the long-term cardiovascular sequelae remain largely unexplored. Emerging evidence suggests that persistent antigenaemia may predispose to the development of a systemic pro-inflammatory/prothrombotic phenotype underpinning the risk of future cardiovascular events. The current manuscript briefly reviews the occurrence of cardiovascular events in patients with all-cause CAP, as well as in pneumococcal and influenza infections. It highlights the close interaction between influenza and pneumococcal pneumonia. It also includes a brief discussion of mechanisms of the acute cardiac events in CAP. However, the primary focus is on the prevalence, pathogenesis and prevention of the longer-term cardiac sequelae of severe pneumococcal disease, particularly in the context of persistent antigenaemia and associated inflammation.


Subject(s)
Cardiovascular Diseases/microbiology , Cardiovascular Diseases/prevention & control , Pneumonia, Pneumococcal/complications , Cardiovascular Diseases/epidemiology , Community-Acquired Infections/complications , Humans , Pneumococcal Vaccines , Risk Assessment
4.
Mucosal Immunol ; 4(3): 325-34, 2011 May.
Article in English | MEDLINE | ID: mdl-20980996

ABSTRACT

Innate immune recognition of the bacterial cell wall constituent peptidoglycan by the cytosolic nucleotide-binding oligomerization domain 2 (Nod2) receptor has a pivotal role in the maintenance of intestinal mucosal homeostasis. Whereas peptidoglycan cleavage by gut-derived lysozyme preserves the recognition motif, the N-acetylmuramoyl-L-alanine amidase activity of the peptidoglycan recognition protein 2 (PGLYRP-2) destroys the Nod2-detected muramyl dipeptide structure. PGLYRP-2 green fluorescent protein (GFP) reporter and wild-type mice were studied by flow cytometry and quantitative RT-PCR to identify Pglyrp-2 expression in cells of the intestinal mucosa and reveal a potential regulatory function on epithelial peptidoglycan recognition. CD3(+)/CD11c(+) T lymphocytes revealed significant Pglyrp-2 expression, whereas epithelial cells and intestinal myeloid cells were negative. The mucosal Pglyrp-2-expressing lymphocyte population demonstrated a mixed T-cell receptor (TCR) αß or γδ phenotype with predominant CD8α and less so CD8ß expression, as well as significant staining for the activation markers B220 and CD69, presenting a typical intraepithelial lymphocyte phenotype. Importantly, exposure of peptidoglycan to PGLYRP-2 significantly reduced Nod2/Rip2-mediated epithelial activation. Also, moderate but significant alterations of the intestinal microbiota composition were noted in Pglyrp-2-deficient animals. PGLYRP-2 might thus have a significant role in regulation of the enteric host-microbe homeostasis.


Subject(s)
Intestinal Mucosa/metabolism , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Proteins/metabolism , T-Lymphocytes/metabolism , Animals , Antigens, CD/metabolism , Cells, Cultured , Genetic Engineering , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Host-Pathogen Interactions , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Lymphocyte Activation , Metagenome , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/immunology , Peptidoglycan/immunology , Proteins/genetics , Proteins/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology
5.
J Infect Dis ; 199(7): 1032-42, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19203261

ABSTRACT

BACKGROUND: The invasive disease potential (IDP) of Streptococcus pneumoniae differs between serotypes, but the reason for this is unknown. METHODS: A total of 47 pneumococcal isolates from 13 serotypes with different IDPs in humans that belonged to 37 multilocus sequence types were compared by whole genome microarrays and mutant analyses. RESULTS: Approximately 34% of the genes were variable, including 95 genes previously shown by signature-tagged mutagenesis (STM) to be required for invasive disease in mice. Many variable genes were localized to 41 accessory regions (ARs), of which 24 contained genes previously identified by STM as required for invasive disease. Only AR6 and AR34 were preferentially found in isolates of serotypes with high IDPs. Neither AR6, which carries a gene previously identified bySTMas required for invasive disease and encodes a 6-phospho-betaglucosidase, nor the putative adhesin expressed by AR34 was required for mouse virulence in TIGR4. CONCLUSIONS: Pneumococci possess a repertoire of ARs that differ between clones and even between isolates of the same clone. The ARs required for invasive disease in humans may be redundant, as no unique pattern distinguished the most invasive clones from others. The ARs that contained genes previously identified by STM as required for virulence in mice were frequently absent from invasive human isolates. Only 1 AR (AR6) was present in almost all isolates from the serotypes with the highest IDP (1, 4, and 7F), whereas it was missing from many others.


Subject(s)
Pneumococcal Infections/microbiology , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/pathogenicity , Animals , Bacterial Adhesion/genetics , Bacterial Proteins/genetics , Genome, Bacterial , Humans , Mice , Mice, Inbred C57BL , Protein Array Analysis , Streptococcus pneumoniae/genetics , Virulence/genetics
6.
Article in English | MEDLINE | ID: mdl-19153457

ABSTRACT

The pilus-associated sortase C from Streptococcus pneumoniae (SrtC or Srt-2) acts as a polymerase for the pilus subunit proteins RrgA and RrgB. Here, the crystallization and preliminary X-ray diffraction analysis of three crystal forms of SrtC are reported. One crystal form belongs to space group P2(1)2(1)2(1), with unit-cell parameters a = 48.9, b = 96.9, c = 98.9 A, alpha = beta = gamma = 90 degrees . The other two crystal forms belong to space group P222, with unit-cell parameters a = 48.8, b = 97.2, c = 99.2 A, alpha = beta = gamma = 90 degrees and a = 48.6, b = 96.5, c = 98.8 A, alpha = beta = gamma = 90 degrees , respectively. Preliminary analysis indicates the presence of two molecules in the asymmetric unit of the crystal for all three forms.


Subject(s)
Aminoacyltransferases/chemistry , Bacterial Proteins/chemistry , Cysteine Endopeptidases/chemistry , Fimbriae, Bacterial/metabolism , Streptococcus pneumoniae/enzymology , Cloning, Molecular , Crystallization , Crystallography, X-Ray/methods , Diffusion , Electrophoresis, Polyacrylamide Gel , Models, Statistical , X-Ray Diffraction
7.
Mol Microbiol ; 66(2): 329-40, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17850254

ABSTRACT

Adherence to host cells is important in microbial colonization of a mucosal surface, and Streptococcus pneumoniae adherence was significantly enhanced by expression of an extracellular pilus composed of three subunits, RrgA, RrgB and RrgC. We sought to determine which subunit(s) confers adherence. Bacteria deficient in RrgA are significantly less adherent than wild-type organisms, while overexpression of RrgA enhances adherence. Recombinant monomeric RrgA binds to respiratory cells, as does RrgC with less affinity, and pre-incubation of epithelial cells with RrgA reduces adherence of wild-type piliated pneumococci. Non-adherent RrgA-negative, RrgB- and RrgC-positive organisms produce pili, suggesting that pilus-mediated adherence is due to expression of RrgA, rather than the pilus backbone itself. In contrast, RrgA-positive strains with disrupted rrgB and rrgC genes exhibit wild-type adherence despite failure to produce pili by Western blot or immunoelectron microscopy. The density of bacteria colonizing the upper respiratory tract of mice inoculated with piliated RrgA-negative pneumococci was significantly less compared with wild-type; in contrast, non-piliated pneumococci expressing non-polymeric RrgA had similar numbers of bacteria in the nasopharynx as piliated wild-type bacteria. These data suggest that RrgA is central in pilus-mediated adherence and disease, even in the absence of polymeric pilus production.


Subject(s)
Adhesins, Bacterial/physiology , Bacterial Adhesion/physiology , Fimbriae Proteins/physiology , Streptococcus pneumoniae/metabolism , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Animals , Bacterial Adhesion/genetics , Blotting, Western , Cell Line, Tumor , Epithelial Cells/microbiology , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Gene Expression Regulation, Bacterial , Humans , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/ultrastructure
8.
Proc Natl Acad Sci U S A ; 104(31): 12907-12, 2007 Jul 31.
Article in English | MEDLINE | ID: mdl-17644611

ABSTRACT

Antibiotic resistance in pneumococci is due to the spread of strains belonging to a limited number of clones. The Spain(9V)-3 clone of sequence type (ST)156 is one of the most successful clones with reduced susceptibility to penicillin [pneumococci nonsusceptible to penicillin (PNSP)]. In Sweden during 2000-2003, a dramatic increase in the number of PNSP isolates was observed. Molecular characterization of these isolates showed that a single clone of sequence type ST156 increased from 40% to 80% of all serotype 14, thus causing the serotype expansion. Additionally, during the same time period, we examined the clonal composition of two serotypes 9V and 19F: all 9V and 20% of 19F isolates belonged to the clonal cluster of ST156, and overall approximately 50% of all PNSP belonged to the ST156 clonal cluster. Moreover, microarray and PCR analysis showed that all ST156 isolates, irrespective of capsular type, carried the rlrA pilus islet. This islet was also found to be present in the penicillin-sensitive ST162 clone, which is believed to be the drug-susceptible ancestor of ST156. Competitive experiments between related ST156 serotype 19F strains confirmed that those containing the rlrA pilus islet were more successful in an animal model of carriage. We conclude that the pilus island is an important biological factor common to ST156 isolates and other successful PNSP clones. In Sweden, a country where the low antibiotic usage does not explain the spread of resistant strains, at least 70% of all PNSP isolates collected during year 2003 carried the pilus islet.


Subject(s)
Penicillins/biosynthesis , Streptococcus pneumoniae/isolation & purification , Streptococcus pneumoniae/metabolism , Bacterial Adhesion , Genetic Variation/genetics , Genome, Bacterial/genetics , Humans , Nasopharynx/microbiology , Streptococcus pneumoniae/genetics , Sweden , Trans-Activators/genetics , Trans-Activators/metabolism
9.
J Intern Med ; 261(6): 511-28, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17547708

ABSTRACT

The innate immunity plays a critical role in host protection against pathogens and it relies amongst others on pattern recognition receptors such as the Toll-like receptors (TLRs) and the nucleotide-binding oligomerization domains proteins (NOD-like receptors, NLRs) to alert the immune system of the presence of invading bacteria. Since their recent discovery less than a decade ago, both TLRs and NLRs have been shown to be crucial in host protection against microbial infections but also in homeostasis of the colonizing microflora. They recognize specific microbial ligands and with the use of distinct adaptor molecules, they activate different signalling pathways that in turns trigger subsequent inflammatory and immune responses that allows a immediate response towards bacterial infections and the initiation of the long-lasting adaptive immunity. In this review, we will focus on the role of the TLRs against bacterial infections in humans in contrast to mice that have been used extensively in experimental models of infections and discuss their role in controlling normal flora or nonpathogenic bacteria. We also highlight how bacteria can evade recognition by TLRs.


Subject(s)
Bacterial Infections/immunology , Immunity, Innate , Signal Transduction/physiology , Toll-Like Receptors/metabolism , Animals , Humans , Intestines/immunology , Intestines/microbiology , Mice , Species Specificity
10.
Proc Natl Acad Sci U S A ; 103(8): 2857-62, 2006 Feb 21.
Article in English | MEDLINE | ID: mdl-16481624

ABSTRACT

Streptococcus pneumoniae (pneumococcus) is a major cause of morbidity and mortality world-wide. The initial event in invasive pneumococcal disease is the attachment of encapsulated pneumococci to epithelial cells in the upper respiratory tract. This work provides evidence that initial bacterial adhesion and subsequent ability to cause invasive disease is enhanced by pili, long organelles able to extend beyond the polysaccharide capsule, previously unknown to exist in pneumococci. These adhesive pili-like appendages are encoded by the pneumococcal rlrA islet, present in some, but not all, clinical isolates. Introduction of the rlrA islet into an encapsulated rlrA-negative isolate allowed pilus expression, enhanced adherence to lung epithelial cells, and provided a competitive advantage upon mixed intranasal challenge of mice. Furthermore, a pilus-expressing rlrA islet-positive clinical isolate was more virulent than a nonpiliated deletion mutant, and it out-competed the mutant in murine models of colonization, pneumonia, and bacteremia. Additionally, piliated pneumococci evoked a higher TNF response during systemic infection, compared with nonpiliated derivatives, suggesting that pneumococcal pili not only contribute to adherence and virulence but also stimulate the host inflammatory response.


Subject(s)
Fimbriae, Bacterial/physiology , Genes, Bacterial/physiology , Genomic Islands , Pneumonia, Bacterial/microbiology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/pathogenicity , Animals , Bacterial Adhesion/genetics , Bacterial Proteins/genetics , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/ultrastructure , Genes, Bacterial/genetics , Genomic Islands/genetics , Genomic Islands/physiology , Mice , Mice, Inbred C57BL , Mutation , Respiratory Mucosa/microbiology , Streptococcus pneumoniae/ultrastructure , Trans-Activators/genetics , Virulence
11.
Clin Microbiol Infect ; 8(10): 613-22, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12390279

ABSTRACT

When bacteria such as Staphylococcus aureus and Streptococcus pneumoniae are exposed to lytic antibiotics such as penicillin and vancomycin, a self-induced killing process is initiated in the organism. This killing occurs via both non-lytic and lytic processes. Recent data suggest that the non-lytic killing system, which might affect the cytoplasmic membrane, secondarily activates murein hydrolases that eventually lyse the cell. Disturbances in this suicide pathway can lead to antibiotic tolerance, a process whereby the antibiotic still exerts its bacteriostatic effects but the self-induced killing system is impaired. In mutants obtained in vitro, signaling pathways have been affected that show either increased or decreased antibiotic-induced killing. Among clinical isolates of S. pneumoniae that are tolerant to penicillin and/or vancomycin, we do not yet know whether these signaling pathways are affected. We could, however, demonstrate that the activity of murein hydrolases is negatively controlled by the production of capsular polysaccharides in one vancomycin-tolerant isolate. Hence, type and level of capsular expression might constitute one factor that determines the degree of lysis, once the killing signal has been elicited by the antibiotic.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/physiology , Streptococcus pneumoniae/drug effects , Animals , Anti-Bacterial Agents/metabolism , Apoptosis , Bacteriophages/metabolism , Humans , Lactams , Models, Chemical , N-Acetylmuramoyl-L-alanine Amidase/physiology , Penicillins/pharmacology , Peptidoglycan/metabolism , Plasmids/metabolism , Rabbits , Signal Transduction/physiology , Streptococcal Infections/drug therapy , Streptococcus pneumoniae/enzymology , Vancomycin/pharmacology
12.
J Intern Med ; 252(2): 91-106, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12190884

ABSTRACT

Antibiotic resistance is a clinical and socioeconomical problem that is here to stay. Resistance can be natural or acquired. Some bacterial species, such as Pseudomonas aeruginosa, show a high intrinsic resistance to a number of antibiotics whereas others are normally highly antibiotic susceptible such as group A streptococci. Acquired resistance evolve via genetic alterations in the microbes own genome or by horizontal transfer of resistance genes located on various types of mobile DNA elements. Mutation frequencies to resistance can vary dramatically depending on the mechanism of resistance and whether or not the organism exhibits a mutator phenotype. Resistance usually has a biological cost for the microorganism, but compensatory mutations accumulate rapidly that abolish this fitness cost, explaining why many types of resistances may never disappear in a bacterial population. Resistance frequently occurs stepwise making it important to identify organisms with low level resistance that otherwise may constitute the genetic platform for development of higher resistance levels. Self-replicating plasmids, prophages, transposons, integrons and resistance islands all represent DNA elements that frequently carry resistance genes into sensitive organisms. These elements add DNA to the microbe and utilize site-specific recombinases/integrases for their integration into the genome. However, resistance may also be created by homologous recombination events creating mosaic genes where each piece of the gene may come from a different microbe. The selection with antibiotics have informed us much about the various genetic mechanisms that are responsible for microbial evolution.


Subject(s)
Drug Resistance, Bacterial , Mutation , Animals , DNA, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Humans , Mycobacterium tuberculosis/drug effects , Penicillin Resistance/genetics , Pseudomonas aeruginosa/drug effects , beta-Lactam Resistance/genetics
14.
J Infect Dis ; 184(7): 861-9, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11550126

ABSTRACT

In a 10-year period, 1987-1997, there was a >4-fold increase in the rate of pneumococcal bacteremia in Sweden. Invasive pneumococcal isolates (n=1136), which were obtained from 18 Swedish clinical microbiology laboratories from 1987 through 1997, and other national and international isolates were serotyped, and their clonal relationships were determined by molecular typing. The increase in invasive pneumococcal disease in Sweden during this period was associated particularly with an increase in isolates of serotypes 1 and 14. A 3-fold increase of type 14 was seen from 1987 through 1992, and a 10-fold increase of type 1 occurred from 1992 through 1997. One dominating penicillin-susceptible clone of type 14 was responsible for the increase of type 14 during the first 5 years. This clone also was found in Canada and the United States and was shown by multilocus sequence typing to correspond to a previously identified hyper-virulent clone. A novel penicillin-susceptible clone of type 1, which was not found among invasive isolates from 1987 or 1992, was responsible for the increase of serotype 1 during the last 5 years. These results illustrate the ability of virulent penicillin-susceptible pneumococcal clones to emerge and spread rapidly within a country.


Subject(s)
Pneumococcal Infections/microbiology , Streptococcus pneumoniae/genetics , Alleles , Child , DNA, Bacterial/genetics , Electrophoresis, Gel, Pulsed-Field , Electrophoresis, Gel, Two-Dimensional , Humans , Microbial Sensitivity Tests , Molecular Epidemiology , Penicillins/pharmacology , Pneumococcal Infections/epidemiology , Prevalence , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/pathogenicity , Sweden/epidemiology , Virulence
15.
Cell Microbiol ; 3(3): 153-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11260138

ABSTRACT

Mucosal epithelial linings function as physical barriers against microbes. In addition, they participate in the first line of host defence by production of a variety of proinflammatory mediators when exposed to microbes and microbial agents. Here, we use a human urinary tract infection model to demonstrate that organ- and cell-specific innate responses induced by lipopolysaccharides (LPS) present on Gram-negative bacteria correlates with the expression of Toll-like receptor 4 (TLR4). The presence of TLR4 on human bladder epithelial cells enables them to rapidly respond to bacterial infections in vitro and in vivo. In contrast, TLR4 is not expressed on human proximal tubule cells isolated from the renal cortex, which may explain the cortical localization of bacteria in pyelonephritis. TLR4-negative renal epithelial cells, A498, are non-responsive to purified LPS, however, they respond to viable bacteria via a mannose-sensitive attachment-mediated pathway. To identify LPS components recognised by bladder epithelial cells, a bacterial lipid A mutant and LPS of different chemotypes were tested. Full interleukin 8 induction required hexa-acylated lipid A and was decreased by between 50% and 70% in the presence of O-antigen. Taken together, we propose that multiple independent pathways, which are organ- and cell-specifically expressed, mediate bacterial recognition and determine the outcome of innate responses to infection.


Subject(s)
Drosophila Proteins , Escherichia coli/immunology , Lipid A/immunology , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Urothelium/metabolism , Urothelium/microbiology , Cell Line , Escherichia coli/physiology , Humans , Immunity, Innate , Interleukin-8/biosynthesis , Kidney/cytology , Kidney/metabolism , Kidney/microbiology , Lipid A/isolation & purification , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 4 , Toll-Like Receptors , Tumor Cells, Cultured , Urinary Bladder/cytology , Urinary Bladder/metabolism , Urinary Bladder/microbiology
16.
Clin Infect Dis ; 32(4): 552-8, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11181117

ABSTRACT

The ability of Streptococcus pneumoniae to escape lysis and killing by vancomycin, a property termed "tolerance," has recently been noted in a laboratory strain of the species. Vancomycin tolerance in clinical isolates represents a potential new health risk. We determined the prevalence of vancomycin and penicillin tolerance among 116 clinical isolates of pneumococci by monitoring lysis and viability after exposure to the respective antibiotic for 4 hours. Eight percent of the strains were tolerant to penicillin and 3% were tolerant to vancomycin. The 3 vancomycin-tolerant isolates also had a high ratio of minimum bactericidal concentration to minimum inhibitory concentration, in contrast to nontolerant strains. They were of serotype 9V and had reduced susceptibility to penicillin. Only 1 was also tolerant to penicillin. Growth rate and ability to divide were not affected in the 3 vancomycin-tolerant strains, and they all lysed with deoxycholate, which indicates autolysin production. Vancomycin tolerance among clinical isolates of pneumococci will necessitate tracking to determine the magnitude of the evolving health risk, since tolerance may contribute to treatment failure (in particular, cases of meningitis, in which bactericidal activity is critical for eradication) and since it may also be a favored background for acquisition of resistance of vancomycin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Tolerance , Pneumococcal Infections/epidemiology , Protein Kinases/genetics , Streptococcus pneumoniae/drug effects , Vancomycin/pharmacology , Amino Acid Sequence , Bacteremia/epidemiology , Bacteremia/microbiology , Drug Tolerance/genetics , Humans , Microbial Sensitivity Tests , Molecular Sequence Data , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Penicillins/pharmacology , Pneumococcal Infections/microbiology , Protein Kinases/chemistry , Protein Kinases/metabolism , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/isolation & purification , Vancomycin Resistance
17.
J Infect Dis ; 183(4): 612-9, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11170987

ABSTRACT

Septic shock, a major cause of death, is characterized by a pathophysiologic increased production of nitric oxide (NO), which leads to vasodilation and myocardial toxicity. Septic Escherichia coli frequently express proteinaceous curli fibers. In this study, curliated E. coli induced high levels of NO by directly inducing type 2 nitric oxide synthase (NOS2) both in vitro and in vivo. More severe hypotension and higher plasma nitrite/nitrate levels were seen in wild type mice systemically infected with curliated E. coli than in animals infected with E. coli mutants that lacked curli proteins. Blood pressure remained stable in NOS2-deficient mice with curliated bacteria. Increased heart rates, transient hypothermia, and loss of gross activity were seen in all mice, regardless of curli expression. Study results suggest that expression of curli fibers by E. coli activates the NO/NOS2 arm of the innate immune system, which leads to a significant fall in blood pressure.


Subject(s)
Bacterial Proteins/physiology , Escherichia coli Infections/physiopathology , Escherichia coli Proteins , Escherichia coli/pathogenicity , Hypotension/physiopathology , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Animals , Aorta, Thoracic/cytology , Bacterial Proteins/genetics , Blood Pressure , Body Temperature , Cells, Cultured , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Heart Rate , Mice , Mice, Inbred C57BL , Nitrates/blood , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitrites/blood , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Shock, Septic/microbiology , Shock, Septic/physiopathology
19.
J Biol Chem ; 275(51): 40478-82, 2000 Dec 22.
Article in English | MEDLINE | ID: mdl-11010975

ABSTRACT

The use of germ-free mice offers the possibility to study antibacterial components in a gut uncolonized by bacteria. We have developed a method to extract and high pressure liquid chromatography-fractionate the antibacterial factors present in the small intestine of a single mouse. By mass spectrometry and sequence analyses of fractions exhibiting antimicrobial activity, we identified and characterized the defensin region in germ-free mice as well as in colonized mice. Defensins made up around 15% of the total antibacterial activity both in germ-free and colonized mice. The intestine of germ-free mice exhibited the same set of mature enteric defensins (defensins 1, 2, 3, 4, and 6) as mice colonized by a normal microflora. Mature defensins are generated through processing of larger precursors by enzymatic removal of a signal peptide and a propiece. We found that all prodefensins were cleaved at a Ser/Ala-Leu bond, giving 34-residue propiece peptides and only trace amounts of the predicted 39-residue peptide. This first step must be followed by the removal of a residual peptide to render the mature defensins, indicating that the processing is more complex than previously anticipated. The same propieces were found in both germ-free and colonized mice, suggesting that the same processing operates independent of bacterial presence in the intestine.


Subject(s)
Defensins/metabolism , Intestine, Small/metabolism , Protein Precursors/metabolism , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Chromatography, High Pressure Liquid , Defensins/chemistry , Germ-Free Life , Intestine, Small/microbiology , Mice , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Precursors/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...