Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Infect Immun ; 87(10)2019 10.
Article in English | MEDLINE | ID: mdl-31331954

ABSTRACT

Group A Streptococcus (GAS) commonly causes pharyngitis and skin infections. Little is known why streptococcal pharyngitis usually does not lead to pneumonia and why the skin is a favorite niche for GAS. To partially address these questions, the effectiveness of neutrophils in clearing wild-type (wt) M1T1 GAS strain MGAS2221 from the lung and from the skin was examined in murine models of intratracheal pneumonia and subcutaneous infection. Ninety-nine point seven percent of the MGAS2221 inoculum was cleared from the lungs of C57BL/6J mice at 24 h after inoculation, while there was no MGAS2221 clearance from skin infection sites. The bronchial termini had robust neutrophil infiltration, and depletion of neutrophils abolished MGAS2221 clearance from the lung. Phagocyte NADPH oxidase but not myeloperoxidase was required for MGAS2221 clearance. Thus, wt M1T1 GAS can be cleared by neutrophils using an NADPH oxidase-dependent mechanism in the lung. MGAS2221 induced robust neutrophil infiltration at the edge of skin infection sites and throughout infection sites at 24 h and 48 h after inoculation, respectively. Neutrophils within MGAS2221 infection sites had no nuclear staining. Skin infection sites of streptolysin S-deficient MGAS2221 ΔsagA were full of neutrophils with nuclear staining, whereas MGAS2221 ΔsagA infection was not cleared. Gp91phox knockout (KO) and control mice had similar GAS numbers at skin infection sites and similar abilities to select SpeB activity-negative (SpeBA-) variants. These results indicate that phagocyte NADPH oxidase-mediated GAS killing is compromised in the skin. Our findings support a model for GAS skin tropism in which GAS generates an anoxic niche to evade phagocyte NADPH oxidase-mediated clearance.


Subject(s)
Host-Pathogen Interactions/immunology , Lung/enzymology , NADPH Oxidases/immunology , Neutrophils/enzymology , Streptococcal Infections/enzymology , Streptococcus pyogenes/pathogenicity , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Female , Lung/immunology , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/genetics , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/microbiology , Organ Specificity , Phagocytes/enzymology , Phagocytes/immunology , Skin/immunology , Skin/microbiology , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcus pyogenes/immunology , Streptolysins/deficiency , Streptolysins/genetics , Streptolysins/immunology
2.
Folia Microbiol (Praha) ; 61(5): 375-83, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26803756

ABSTRACT

Nasopharyngeal colonization by Streptococcus pneumoniae is an important initial step for the subsequent development of pneumococcal infections. Pneumococci have many virulence factors that play a role in colonization. Pneumolysin (PLY), a pivotal pneumococcal virulence factor for invasive disease, causes severe tissue damage and inflammation with disruption of epithelial tight junctions. In this study, we evaluated the role of PLY in nasal colonization of S. pneumoniae using a mouse colonization model. A reduction of numbers of PLY-deficient pneumococci recovered from nasal tissue, as well as nasal wash, was observed at days 1 and 2 post-intranasal challenges, but not later. The findings strongly support an important role for PLY in the initial establishment nasal colonization. PLY-dependent invasion of local nasal mucosa may be required to establish nasal colonization with S. pneumoniae. The data help provide a rationale to explain why an organism that exists as an asymptomatic colonizer has evolved virulence factors that enable it to occasionally invade and kill its hosts. Thus, the same pneumococcal virulence factor, PLY that can contribute to killing the host, may also play a role early in the establishment of nasopharynx carriage.


Subject(s)
Carrier State/microbiology , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/metabolism , Streptolysins/metabolism , Animals , Bacterial Proteins/metabolism , Female , Mice, Inbred CBA , Streptolysins/deficiency , Virulence Factors/deficiency , Virulence Factors/metabolism
3.
Thorax ; 70(7): 636-46, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25964315

ABSTRACT

RATIONALE: Respiratory tract infections are common in patients suffering from pulmonary fibrosis. The interplay between bacterial infection and fibrosis is characterised poorly. OBJECTIVES: To assess the effect of Gram-positive bacterial infection on fibrosis exacerbation in mice. METHODS: Fibrosis progression in response to Streptococcus pneumoniae was examined in two different mouse models of pulmonary fibrosis. MEASUREMENTS AND MAIN RESULTS: We demonstrate that wild-type mice exposed to adenoviral vector delivery of active transforming growth factor-ß1 (TGFß1) or diphteria toxin (DT) treatment of transgenic mice expressing the DT receptor (DTR) under control of the surfactant protein C (SPC) promoter (SPC-DTR) to induce pulmonary fibrosis developed progressive fibrosis following infection with Spn, without exhibiting impaired lung protective immunity against Spn. Antibiotic treatment abolished infection-induced fibrosis progression. The cytotoxin pneumolysin (Ply) of Spn caused this phenomenon in a TLR4-independent manner, as Spn lacking Ply (SpnΔply) failed to trigger progressive fibrogenesis, whereas purified recombinant Ply did. Progressive fibrogenesis was also observed in AdTGFß1-exposed Ply-challenged TLR4 KO mice. Increased apoptotic cell death of alveolar epithelial cells along with an attenuated intrapulmonary release of antifibrogenic prostaglandin E2 was found to underlie progressive fibrogenesis in Ply-challenged AdTGFß1-exposed mice. Importantly, vaccination of mice with the non-cytotoxic Ply derivative B (PdB) substantially attenuated Ply-induced progression of lung fibrosis in AdTGFß1-exposed mice. CONCLUSIONS: Our data unravel a novel mechanism by which infection with Spn through Ply release induces progression of established lung fibrosis, which can be attenuated by protein-based vaccination of mice.


Subject(s)
Pneumonia, Pneumococcal/complications , Pulmonary Fibrosis/microbiology , Streptolysins/physiology , Animals , Anti-Bacterial Agents/therapeutic use , Apoptosis/drug effects , Bacterial Proteins/pharmacology , Bacterial Proteins/physiology , Bronchoalveolar Lavage Fluid/immunology , Diphtheria Toxin , Disease Models, Animal , Disease Progression , Epithelial Cells/drug effects , Female , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pneumococcal Vaccines , Pneumonia, Pneumococcal/drug therapy , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/metabolism , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/prevention & control , Streptolysins/deficiency , Streptolysins/pharmacology , Transforming Growth Factor beta1/metabolism
4.
Infect Immun ; 79(10): 4122-30, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21788389

ABSTRACT

Streptococcus pneumoniae is a leading cause of human diseases such as pneumonia, bacteremia, meningitis, and otitis media. Pneumolysin (Ply) is an important virulence factor of S. pneumoniae and a promising future vaccine target. However, the expansion of clones carrying ply alleles with reduced hemolytic activity has been observed in serotypes associated with outbreaks of invasive disease and includes an allele identified in a highly virulent serotype 1 isolate (ply4496). The virulence of Ply-deficient and ply allelic-replacement derivatives of S. pneumoniae D39 was compared with that of wild-type D39. In addition, the protective immunogenicity of Ply against pneumococci with low versus high hemolytic activity was also investigated. Replacement of D39 ply with ply4496 resulted in a small but statistically significant reduction of virulence. However, both native Ply- and Ply4496-expressing strains were significantly more virulent than a Ply-deficient mutant. While the numbers of both Ply- and Ply4496-expressing isolate cells were higher in the blood than the numbers of Ply-deficient mutant cells, the growth of the Ply4496-expressing strain was superior to that of the wild type in the first 15 h postchallenge. Ply immunization provided protection regardless of the hemolytic activity of the challenge strain. In summary, we show that low-hemolytic-activity Ply alleles contribute to systemic virulence and may provide a survival advantage in the blood. Moreover, pneumococci expressing such alleles remain vulnerable to Ply-based vaccines.


Subject(s)
Bacteremia/microbiology , Blood/microbiology , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/pathogenicity , Streptolysins/genetics , Amino Acid Sequence , Animals , Animals, Outbred Strains , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Female , Hemolysis , Humans , Immunization , Mice , Molecular Sequence Data , Mutation , Pneumococcal Infections/epidemiology , Pneumococcal Infections/prevention & control , Sequence Alignment , Sequence Analysis, DNA , Streptococcus pneumoniae/immunology , Streptolysins/deficiency , Streptolysins/immunology , Virulence
5.
J Immunol ; 187(1): 434-40, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21646297

ABSTRACT

Streptococcus pneumoniae is a leading cause of pneumonia, meningitis, and sepsis. Pneumococci can be divided into >90 serotypes that show differences in the pathogenicity and invasiveness. We tested the hypotheses that the innate immune inflammasome pathway is involved in fighting pneumococcal pneumonia and that some invasive pneumococcal types are not recognized by this pathway. We show that human and murine mononuclear cells responded to S. pneumoniae expressing hemolytic pneumolysin by producing IL-1ß. This IL-1ß production depended on the NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome. Some serotype 1, serotype 8, and serotype 7F bacteria, which have previously been associated with increased invasiveness and with production of toxins with reduced hemolytic activity, or bacterial mutants lacking pneumolysin did not stimulate notable IL-1ß production. We further found that NLRP3 was beneficial for mice during pneumonia caused by pneumococci expressing hemolytic pneumolysin and was involved in cytokine production and maintenance of the pulmonary microvascular barrier. Overall, the inflammasome pathway is protective in pneumonia caused by pneumococci expressing hemolytic toxin but is not activated by clinically important pneumococcal sequence types causing invasive disease. The study indicates that a virulence factor polymorphism may substantially affect the recognition of bacteria by the innate immune system.


Subject(s)
Carrier Proteins/physiology , Genetic Variation/immunology , Inflammasomes/metabolism , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/pathology , Streptolysins/genetics , Animals , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Female , Genetic Predisposition to Disease , Humans , Immunity, Innate/genetics , Inflammasomes/physiology , Interleukin-18/physiology , Interleukin-1beta/biosynthesis , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , NLR Family, Pyrin Domain-Containing 3 Protein , Nod2 Signaling Adaptor Protein/physiology , Pneumonia, Pneumococcal/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Streptolysins/biosynthesis , Streptolysins/deficiency , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/physiology
6.
Microb Pathog ; 48(3-4): 124-30, 2010.
Article in English | MEDLINE | ID: mdl-20096771

ABSTRACT

Biofilm formation has been suggested to play an important role during Streptococcus pneumoniae nasopharyngeal colonization and may facilitate progression to pneumonia. To test whether the ability of S. pneumoniae to form biofilms was important for virulence we screened the ability of 30 invasive and 22 non-invasive clinical isolates of serotype 6A and 6B to form early biofilms on polystyrene microtiter plates and infect mice following intranasal and intratracheal challenge. We first determined that no correlation existed between the ability to form early biofilms and whether isolates were collected from healthy carriers or individuals with invasive disease. A disconnect between biofilm forming ability and the capacity to colonize the nasopharynx, cause pneumonia, and enter the bloodstream was also observed in mice. Importantly, S. pneumoniae mutants deficient in the established virulence determinants pneumolysin, CbpA, and hydrogen peroxide formed biofilms normally. Incidentally, we determined that robust biofilm production was dependent on the formation and coalescing of bacterial aggregates on a thin layer of bacteria attached to the plate surface. In summary, these studies suggest that the ability to form early biofilms in vitro does not reflect virulence potential. More complex studies are required to determine if biofilm formation is important for virulence.


Subject(s)
Bacteremia/microbiology , Biofilms/growth & development , Carrier State/microbiology , Pneumonia, Pneumococcal/microbiology , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/pathogenicity , Animals , Bacterial Proteins/genetics , Bacterial Typing Techniques , Female , Humans , Hydrogen Peroxide/metabolism , Mice , Mice, Inbred BALB C , Nasopharynx/microbiology , Serotyping , Streptococcus pneumoniae/isolation & purification , Streptolysins/deficiency , Virulence , Virulence Factors/deficiency
7.
Immunobiology ; 215(4): 251-63, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20071053

ABSTRACT

The human nasopharynx is an important ecological niche for Streptococcus pneumoniae, and asymptomatic nasopharyngeal carriage is a common precursor to invasive disease. However, knowledge of the immunological events, which occur during carriage, both on a cellular and humoral level, remains limited. Here, we present a long-term stable model of asymptomatic nasopharyngeal carriage using outbred naïve mice, in which we have investigated the effect of previous nasopharyngeal exposure to pneumococci, in the prevention of subsequent carriage and invasive disease. Carriage of D39 wildtype pneumococci restricted to the nasopharynx could be detected for at least 28 days post-infection, whereas nasopharyngeal carriage of a pneumolysin negative isogenic mutant (PLN-A) was cleared in 7-14 days. Both carriage events induced total and capsule specific IgA mucosal antibodies and increased levels of systemic antibodies (IgG against pneumococcal surface protein A (PspA) and IgM capsular polysaccharide), which increased over time and correlated to reduced nasopharyngeal pneumococcal numbers. Prior nasopharyngeal colonisation with PLN-A significantly reduced the duration of subsequent D39 wildtype carriage, and significantly increased survival following invasive pneumococcal challenge. In this case systemic anti-PspA and anti-capsular antibody IgM concentrations showed a strong correlation with reduced bacterial numbers in the lungs and nasopharynx, respectively and also with increased levels of IL17A and CD4+ T cells in lungs of pre-colonised mice. Prior nasopharyngeal colonisation with PLN-A also resulted in significant cross-serotype protection with mice protected from invasive disease with serotype 3 strain (A66) after pre-colonisation with a serotype 2 strain (D39). Our results suggest that both mucosal and systemic antibody as well as cellular host factors have a role in long-term protection against both colonisation and invasive pneumococcal challenge.


Subject(s)
Carrier State/immunology , Nasopharynx/immunology , Nasopharynx/microbiology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Bacterial Capsules/blood , Bacterial Capsules/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , Carrier State/microbiology , Female , Immunoglobulin A/analysis , Immunoglobulin G/blood , Interleukin-17/immunology , Lung/immunology , Mice , Mutation , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/genetics , Streptolysins/deficiency , Streptolysins/genetics , Virulence/immunology
8.
J Immunol ; 183(9): 5823-9, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19812205

ABSTRACT

Macrophages play a crucial role in the innate immune response against the human pathogen Streptococcus pyogenes, yet the innate immune response against the bacterium is poorly characterized. In the present study, we show that caspase-1 activation and IL-1beta secretion were induced by live, but not killed, S. pyogenes, and required expression of the pore-forming toxin streptolysin O. Using macrophages deficient in inflammasome components, we found that both NLR family pyrin domain-containing 3 (Nlrp3) and apoptosis-associated speck-like protein (Asc) were crucial for caspase-1 activation and IL-1beta secretion, but dispensable for pro-IL-1beta induction, in response to S. pyogenes infection. Conversely, macrophages deficient in the essential TLR adaptors Myd88 and Trif showed normal activation of caspase-1, but impaired induction of pro-IL-1beta and secretion of IL-1beta. Notably, activation of caspase-1 by TLR2 and TLR4 ligands in the presence of streptolysin O required Myd88/Trif, whereas that induced by S. pyogenes was blocked by inhibition of NF-kappaB. Unlike activation of the Nlrp3 inflammasome by TLR ligands, the induction of caspase-1 activation by S. pyogenes did not require exogenous ATP or the P2X7R. In vivo experiments revealed that Nlrp3 was critical for the production of IL-1beta but was not important for survival in a mouse model of S. pyogenes peritoneal infection. These results indicate that caspase-1 activation in response to S. pyogenes infection requires NF-kappaB and the virulence factor streptolysin O, but proceeds independently of P2X7R and TLR signaling.


Subject(s)
Carrier Proteins/metabolism , Inflammation Mediators/physiology , NF-kappa B/metabolism , Receptors, Purinergic P2/physiology , Signal Transduction/immunology , Streptococcus pyogenes/immunology , Streptolysins/physiology , Toll-Like Receptors/physiology , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Carrier Proteins/genetics , Carrier Proteins/physiology , Caspase 1/metabolism , Cells, Cultured , Disease Models, Animal , Inflammation Mediators/metabolism , Interleukin-1beta/biosynthesis , Interleukin-1beta/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Multiprotein Complexes/physiology , NF-kappa B/physiology , NLR Family, Pyrin Domain-Containing 3 Protein , Peritonitis/immunology , Peritonitis/microbiology , Peritonitis/mortality , Protein Precursors/biosynthesis , Protein Precursors/metabolism , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7 , Signal Transduction/genetics , Streptococcal Infections/immunology , Streptococcal Infections/metabolism , Streptococcal Infections/mortality , Streptococcus pyogenes/genetics , Streptolysins/deficiency , Streptolysins/metabolism , Survival Analysis
9.
J Immunol ; 175(3): 1813-9, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16034123

ABSTRACT

Streptococcus pneumoniae is a common cause of septicemia in the immunocompetent host. To establish infection, S. pneumoniae has to overcome host innate immune responses, one component of which is the complement system. Using isogenic bacterial mutant strains and complement-deficient immune naive mice, we show that the S. pneumoniae virulence factor pneumolysin prevents complement deposition on S. pneumoniae, mainly through effects on the classical pathway. In addition, using a double pspA-/ply- mutant strain we demonstrate that pneumolysin and the S. pneumoniae surface protein PspA act in concert to affect both classical and alternative complement pathway activity. As a result, the virulence of the pspA-/ply- strain in models of both systemic and pulmonary infection is greatly attenuated in wild-type mice but not complement deficient mice. The sensitivity of the pspA-/ply- strain to complement was exploited to demonstrate that although early innate immunity to S. pneumoniae during pulmonary infection is partially complement-dependent, the main effect of complement is to prevent spread of S. pneumoniae from the lungs to the blood. These data suggest that inhibition of complement deposition on S. pneumoniae by pneumolysin and PspA is essential for S. pneumoniae to successfully cause septicemia. Targeting mechanisms of complement inhibition could be an effective therapeutic strategy for patients with septicemia due to S. pneumoniae or other bacterial pathogens.


Subject(s)
Bacterial Proteins/physiology , Complement Inactivator Proteins/physiology , Complement System Proteins/metabolism , Heat-Shock Proteins/physiology , Pneumococcal Infections/immunology , Sepsis/immunology , Streptococcus pneumoniae/immunology , Streptolysins/physiology , Animals , Bacterial Proteins/pharmacology , Complement C1q/deficiency , Complement C1q/genetics , Complement C1q/physiology , Complement C3/deficiency , Complement C3/metabolism , Complement C3/physiology , Complement Factor B/deficiency , Complement Factor B/genetics , Complement Inactivator Proteins/deficiency , Complement Inactivator Proteins/pharmacology , Complement Pathway, Classical/genetics , Complement Pathway, Classical/immunology , Complement System Proteins/deficiency , Complement System Proteins/physiology , Drug Synergism , Heat-Shock Proteins/deficiency , Heat-Shock Proteins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumococcal Infections/genetics , Pneumococcal Infections/microbiology , Sepsis/genetics , Sepsis/microbiology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Streptolysins/deficiency , Streptolysins/pharmacology , Virulence Factors/deficiency , Virulence Factors/pharmacology , Virulence Factors/physiology
10.
Infect Immun ; 66(4): 1671-9, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9529097

ABSTRACT

Streptolysin S (SLS) is a potent cytolytic toxin produced by nearly all group A streptococci (GAS). SLS-deficient Tn916 insertional mutants were generated from two clinical isolates of GAS, MGAS166s and T18Ps (M serotypes 1 and 18, respectively), by transposon mutagenesis using Tn916 donor strain Enterococcus faecalis CG110. Representative nonhemolytic transconjugants SBNH5 and SB30-2 each harbored a single Tn916 insertion in identical loci. The insertion in SBNH5 was located in the promoter region of an open reading frame, designated sagA, rendering it transcriptionally inactive. Protease, streptolysin O, and DNase activities and the production of M protein remained the same in the nonhemolytic mutants and the wild-type strains, as did the growth rates and exoprotein profiles. Transconjugants were evaluated in an established murine model by injecting the organisms subcutaneously and monitoring the mice for alterations in weight and the development of necrotic lesions. Animals infected with SBNH5, compared to those infected with MGAS166s, gained weight during the first 24 h (+1.15 versus -1.16 g; P < 0.05) and had fewer necrotic lesions (0 versus 7; P = 0.0007). Animals infected with SB30-2, compared to those infected with T18Ps, also gained weight within the first 24 h (+0.54 versus -0.66 g; P < 0.05) and produced fewer necrotic lesions (1 versus 8; P = 0.001). Revertants of the mutants in which Tn916 had been excised regained the hemolytic phenotype and the virulence profile of the wild-type strains. This study demonstrates that SLS-deficient mutants of GAS, belonging to different M serotypes and containing identical Tn916 mutations, are markedly less virulent than their isogenic parents.


Subject(s)
Bacterial Proteins , DNA Transposable Elements , Streptococcus pyogenes/pathogenicity , Streptolysins/deficiency , Amino Acid Sequence , Animals , Base Sequence , Conjugation, Genetic , Hemolysis , Mice , Molecular Sequence Data , Mutation , Streptococcal Infections/pathology , Virulence
11.
Infect Immun ; 66(2): 839-42, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9453652

ABSTRACT

Pneumolysin-deficient mutant strains of Streptococcus pneumoniae are known to cause less-severe sepsis than wild-type pneumococcal strains that produce pneumolysin. This difference is associated with greater host resistance in mice infected with the pneumolysin-deficient strains. These studies show that the host resistance developed during the first 1 to 2 days after infection with a pneumolysin-deficient mutant strain is dependent on tumor necrosis factor alpha but is apparently independent of interleukin 1beta (IL-1beta) or IL-6. Survival beyond 5 days appeared to depend on the ability of the mice to produce IL-1beta.


Subject(s)
Bacteremia/immunology , Pneumococcal Infections/immunology , Streptolysins/deficiency , Tumor Necrosis Factor-alpha/physiology , Animals , Bacterial Proteins , Interleukin-1/blood , Interleukin-1/physiology , Interleukin-6/blood , Mice , Mice, Inbred CBA , Tumor Necrosis Factor-alpha/analysis
12.
Microb Pathog ; 25(6): 337-42, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9895272

ABSTRACT

The universal and highly conserved production of pneumolysin, the major pneumococcal cytolysin, among clinical isolates of Streptococcus pneumoniae and the previously reported association of pneumolysin production with increased pneumococcal adherence to respiratory epithelium in organ cultures suggest that this toxin might be important for nasopharyngeal colonization. We confirmed that pneumolysin-deficient mutant pneumococcal strains had decreased adherence to respiratory epithelial cells in vitro compared with their isogeneic wild-type strains. However, neither early nor sustained colonization by type 14 S. pneumoniae in an established murine model was dependent on bacterial production of pneumolysin. We conclude that pneumolysin production is not a major determinant of successful nasopharyngeal colonization by pneumococci.


Subject(s)
Bacterial Adhesion/physiology , Streptococcus pneumoniae/physiology , Streptolysins/physiology , Animals , Bacterial Proteins , Cell Line , Epithelial Cells/microbiology , Female , Humans , Mice , Mice, Knockout , Mutation , Nose/microbiology , Pharynx/microbiology , Specific Pathogen-Free Organisms , Streptolysins/deficiency
13.
Infect Immun ; 63(2): 442-7, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7822008

ABSTRACT

Streptococcus pneumoniae is the most common cause of community-acquired pneumonia, and pneumolysin, a hemolytic toxin, is thought to be an important virulence factor. We have studied the interaction of a pneumolysin-sufficient type II S. pneumoniae strain (PL+) and an otherwise identical pneumolysin-deficient derivative (PL-) with human respiratory mucosa in an organ culture with an air interface for up to 48 h. Ciliary beat frequency (CBF) was measured by a photometric technique, and adherence to and invasion of the epithelium were assessed by scanning and transmission electron microscopy. PL+ and PL- caused a progressive fall in CBF compared with the control which became significant (P < 0.01) at 24 h for PL+ and at 48 h for PL-. At 24 h, there was a significant increase in the percentage of the mucosa of the organ culture that was damaged for PL+ compared with the control (P < 0.01) and PL- (P < 0.02). At 48 h, there was a significant increase in mucosal damage for both PL+ (P < 0.005) and PL- (P < 0.05) compared with the control. At 24 and 48 h, PL+ and PL- adhered predominantly to mucus and damaged cells. PL+ infection alone caused separation of tight junctions between epithelial cells, and at 48 h PL+ cells were adherent to the separated edges of otherwise healthy unciliated cells. PL+ and PL- both caused damage to the epithelial cell ultrastructure. S. pneumoniae infection caused patchy damage to the respiratory mucosa and a lowered CBF. These changes were more severe and occurred earlier with the pneumolysin-sufficient variant.


Subject(s)
Adenoids/microbiology , Streptococcus pneumoniae/pathogenicity , Streptolysins/physiology , Bacterial Adhesion , Bacterial Proteins , Cilia/ultrastructure , Microscopy, Electron, Scanning , Mucous Membrane/microbiology , Mucus , Organ Culture Techniques , Streptolysins/deficiency
14.
Infect Immun ; 63(2): 448-55, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7822009

ABSTRACT

Pneumolysin is a cytoplasmic virulence factor of Streptococcus pneumoniae that can interfere with phagocyte function in vitro. We have examined the effects of pneumolysin in vitro and in vivo and have found that it protects intravenously injected pneumococci against infection-induced host resistance. We employed a virulent capsular type 2 pneumococcal strain, D39, and its isogenic pneumolysin-negative mutant, PLN. Strain D39 exhibited exponential net growth in mice (doubling time, 1.4 h); 24 to 28 h after infection with 10(4) CFU, the numbers of pneumococci reached 10(9) to 10(10) CFU/ml and the mice died. Strain PLN yielded identical net growth in mice until reaching 10(6) to 10(7) CFU/ml at 12 to 18 h postinfection. At this time, the increase in the level of PLN CFU per milliliter ceased and remained constant for several days. PLN exhibited wild-type growth kinetics in mice when coinfected simultaneously with strain D39. This observation suggests that pneumolysin exerts its effects at a distance. By 12 to 18 h postinfection with PLN, mice exhibited the following evidence of an induced inflammatory response: (i) elevated plasma interleukin-6, (ii) a halt in the net growth of PLN, and (iii) control of the net growth of pneumolysin-producing D39 pneumococci upon subsequent challenge. Our data suggest that pneumolysin plays a critical role in sepsis during the first few hours after infection by enabling pneumococci to cause acute sepsis rather than a chronic bacteremia. However, once chronic bacteremia was established, it appeared that pneumolysin was no longer able to act as a virulence factor.


Subject(s)
Neutrophils/microbiology , Streptococcal Infections/immunology , Streptococcus pneumoniae/pathogenicity , Streptolysins/physiology , Animals , Bacteremia/etiology , Bacterial Proteins , Chronic Disease , Inflammation/immunology , Interferon-gamma/blood , Interleukin-6/blood , Mice , Mice, Inbred CBA , Phagocytosis , Sepsis/etiology , Streptolysins/deficiency , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...