Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Microbiol Spectr ; 9(2): e0031821, 2021 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-34468195

RESUMO

Streptococcus pneumoniae, the most common cause of community-acquired pneumonia, causes severe invasive infections, including meningitis and bacteremia. The widespread use of macrolides has been reported to increase the prevalence of macrolide-resistant S. pneumoniae (MRSP), thereby leading to treatment failure in patients with pneumococcal pneumonia. However, previous studies have demonstrated that several macrolides and lincosamides have beneficial effects on MRSP infection since they inhibit the production and release of pneumolysin, a pneumococcal pore-forming toxin released during autolysis. In this regard, we previously demonstrated that the mechanisms underlying the inhibition of pneumolysin release by erythromycin involved both the transcriptional downregulation of the gene encoding pneumolysin and the impairment of autolysis in MRSP. Here, using a cell supernatant of the culture, we have shown that clarithromycin inhibits pneumolysin release in MRSP. However, contrary to previous observations in erythromycin-treated MRSP, clarithromycin upregulated the transcription of the pneumococcal autolysis-related lytA gene and enhanced autolysis, leading to the leakage of pneumococcal DNA. On the other hand, compared to erythromycin, clarithromycin significantly downregulated the gene encoding pneumolysin. In a mouse model of MRSP pneumonia, the administration of both clarithromycin and erythromycin significantly decreased the pneumolysin protein level in bronchoalveolar lavage fluid and improved lung injury and arterial oxygen saturation without affecting bacterial load. Collectively, these in vitro and in vivo data reinforce the benefits of macrolides on the clinical outcomes of patients with pneumococcal pneumonia. IMPORTANCE Pneumolysin is a potent intracellular toxin possessing multiple functions that augment pneumococcal virulence. For over 10 years, sub-MICs of macrolides, including clarithromycin, have been recognized to decrease pneumolysin production and release from pneumococcal cells. However, this study indicates that macrolides significantly slowed pneumococcal growth, which may be related to decreased pneumolysin release recorded by previous studies. In this study, we demonstrated that clarithromycin decreases pneumolysin production through downregulation of ply gene transcription, regardless of its inhibitory activity against bacterial growth. Additionally, administration of clarithromycin resulted in the amelioration of lung injury in a mouse model of pneumonia induced by macrolide-resistant pneumococci. Therefore, therapeutic targeting of pneumolysin offers a good strategy to treat pneumococcal pneumonia.


Assuntos
Claritromicina/farmacologia , Eritromicina/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Streptococcus pneumoniae/metabolismo , Estreptolisinas/biossíntese , Animais , Antibacterianos/farmacologia , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Humanos , Lincosamidas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pneumonia/tratamento farmacológico , Pneumonia/microbiologia , Streptococcus pneumoniae/genética , Estreptolisinas/genética , Transcrição Gênica/efeitos dos fármacos
2.
Int J Mol Sci ; 21(22)2020 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-33182614

RESUMO

Bacterial pneumonia is one of the most prevalent infectious diseases and has high mortality in sensitive patients (children, elderly and immunocompromised). Although an infection, the disease alters the alveolar epithelium homeostasis and hinders normal breathing, often with fatal consequences. A special case is hospitalized aged patients, which present a high risk of infection and death because of the community acquired version of the Streptococcus pneumoniae pneumonia. There is evidence that early antibiotics treatment decreases the inflammatory response during pneumonia. Here, we investigate mechanistically this strategy using a multi-level mathematical model, which describes the 24 first hours after infection of a single alveolus from the key signaling networks behind activation of the epithelium to the dynamics of the local immune response. With the model, we simulated pneumonia in aged and young patients subjected to different antibiotics timing. The results show that providing antibiotics to elderly patients 8 h in advance compared to young patients restores in aged individuals the effective response seen in young ones. This result suggests the use of early, probably prophylactic, antibiotics treatment in aged hospitalized people with high risk of pneumonia.


Assuntos
Antibacterianos/administração & dosagem , Modelos Imunológicos , Infiltração de Neutrófilos , Pneumonia Pneumocócica/tratamento farmacológico , Pneumonia Pneumocócica/imunologia , Idoso , Envelhecimento/imunologia , Animais , Proteínas de Bactérias/biossíntese , Simulação por Computador , Citocinas/imunologia , Esquema de Medicação , Humanos , Conceitos Matemáticos , Camundongos , Pneumonia Pneumocócica/microbiologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/microbiologia , Índice de Gravidade de Doença , Streptococcus pneumoniae/imunologia , Streptococcus pneumoniae/patogenicidade , Estreptolisinas/biossíntese , Análise de Sistemas
3.
Bull Exp Biol Med ; 168(4): 485-487, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32146631

RESUMO

Immunogenic and protective activity of recombinant pneumolysin was studied in experiments on male BALB/c mice. The mice were immunized intraperitoneally with recombinant pneumolysin sorbed on Al(OH)3 (200 µg per mouse). In 2 weeks after immunization, the isotypes of antibodies to recombinant pneumolysin in the serum of immunized mice were determined by ELISA. The animals were infected with Streptococcus pneumoniae serotype 3. Immunization with recombinant pneumolysin induced the production of anti-pneumolysin antibodies, mainly of IgG1 subisotype. On day 21 after intraperitoneal infection with S. pneumoniae serotype 3 in a dose of 106 microbial cells, the survival rate of animals immunized with recombinant pneumolysin in a dose of 25 µg/mouse was 67% vs. 0% in the control (p<0.001). Recombinant pneumolysin could be considered as a promising protective antigen for inclusion in the serotype-independent vaccine against S. pneumoniae.


Assuntos
Anticorpos Antibacterianos/biossíntese , Imunoglobulina G/biossíntese , Infecções Pneumocócicas/prevenção & controle , Vacinas Pneumocócicas/administração & dosagem , Streptococcus pneumoniae/imunologia , Estreptolisinas/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Compostos de Alúmen/administração & dosagem , Animais , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/biossíntese , Imunização/métodos , Imunogenicidade da Vacina , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/mortalidade , Infecções Pneumocócicas/patologia , Vacinas Pneumocócicas/biossíntese , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/biossíntese , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/patogenicidade , Estreptolisinas/biossíntese , Análise de Sobrevida
4.
Infect Immun ; 87(12)2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31527126

RESUMO

Severe manifestations of group A Streptococcus (GAS) infections are associated with massive tissue destruction and high mortality. Clindamycin (CLI), a bacterial protein synthesis inhibitor, is recommended for treating patients with severe invasive GAS infection. Nonetheless, the subinhibitory concentration of CLI induces the production of GAS virulent exoproteins, such as streptolysin O (SLO) and NADase, which would enhance bacterial virulence and invasiveness. A better understanding of the molecular mechanism of how CLI triggers GAS virulence factor expression will be critical to develop appropriate therapeutic approaches. The present study shows that CLI activates SLO and NADase expressions in the emm1-type CLI-susceptible wild-type strain but not in covS or control of virulence sensor (CovS) phosphatase-inactivated mutants. Supplementation with Mg2+, which is a CovS phosphatase inhibitor, inhibits the CLI-mediated SLO upregulation in a dose-dependent manner in CLI-susceptible and CLI-resistant strains. These results not only reveal that the phosphorylation of response regulator CovR is essential for responding to CLI stimuli, but also suggest that inhibiting the phosphatase activity of CovS could be a potential strategy for the treatment of invasive GAS infection with CLI.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Clindamicina/farmacologia , Histidina Quinase/metabolismo , Proteínas Repressoras/metabolismo , Streptococcus pyogenes/metabolismo , Estreptolisinas/biossíntese , Proteínas de Bactérias/biossíntese , Histidina Quinase/antagonistas & inibidores , Histidina Quinase/genética , Magnésio/farmacologia , Monoéster Fosfórico Hidrolases/metabolismo , Streptococcus pyogenes/patogenicidade
5.
Infect Immun ; 87(10)2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31308088

RESUMO

Acute otitis media is one of the most common childhood infections worldwide. Currently licensed vaccines against the common otopathogen Streptococcus pneumoniae target the bacterial capsular polysaccharide and confer no protection against nonencapsulated strains or capsular types outside vaccine coverage. Mucosal infections such as acute otitis media remain prevalent, even those caused by vaccine-covered serotypes. Here, we report that a protein-based vaccine, a fusion construct of epitopes of CbpA to pneumolysin toxoid, confers effective protection against pneumococcal acute otitis media for non-PCV-13 serotypes and enhances protection for PCV-13 serotypes when coadministered with PCV-13. Having cross-reactive epitopes, the fusion protein also induces potent antibody responses against nontypeable Haemophilus influenzae and S. pneumoniae, engendering protection against acute otitis media caused by emerging unencapsulated otopathogens. These data suggest that augmenting capsule-based vaccination with conserved, cross-reactive protein-based vaccines broadens and enhances protection against acute otitis media.


Assuntos
Anticorpos Antibacterianos/biossíntese , Infecções por Haemophilus/prevenção & controle , Haemophilus influenzae/imunologia , Otite Média/prevenção & controle , Vacinas Pneumocócicas/biossíntese , Proteínas Recombinantes de Fusão/biossíntese , Streptococcus pneumoniae/imunologia , Doença Aguda , Animais , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Proteção Cruzada , Reações Cruzadas , Feminino , Expressão Gênica , Infecções por Haemophilus/imunologia , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/efeitos dos fármacos , Haemophilus influenzae/patogenicidade , Humanos , Imunogenicidade da Vacina , Camundongos , Camundongos Endogâmicos BALB C , Otite Média/imunologia , Otite Média/microbiologia , Vacinas Pneumocócicas/administração & dosagem , Vacinas Pneumocócicas/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/patogenicidade , Estreptolisinas/biossíntese , Estreptolisinas/genética , Toxoides/biossíntese , Toxoides/genética , Vacinação , Vacinas Sintéticas
6.
Infect Immun ; 84(7): 2086-2093, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27141081

RESUMO

Variable-number tandem-repeat (VNTR) polymorphisms are ubiquitous in bacteria. However, only a small fraction of them has been functionally studied. Here, we report an intergenic VNTR polymorphism that confers an altered level of toxin production and increased virulence in Streptococcus pyogenes The nature of the polymorphism is a one-unit deletion in a three-tandem-repeat locus upstream of the rocA gene encoding a sensor kinase. S. pyogenes strains with this type of polymorphism cause human infection and produce significantly larger amounts of the secreted cytotoxins S. pyogenes NADase (SPN) and streptolysin O (SLO). Using isogenic mutant strains, we demonstrate that deleting one or more units of the tandem repeats abolished RocA production, reduced CovR phosphorylation, derepressed multiple CovR-regulated virulence factors (such as SPN and SLO), and increased virulence in a mouse model of necrotizing fasciitis. The phenotypic effect of the VNTR polymorphism was nearly the same as that of inactivating the rocA gene. In summary, we identified and characterized an intergenic VNTR polymorphism in S. pyogenes that affects toxin production and virulence. These new findings enhance understanding of rocA biology and the function of VNTR polymorphisms in S. pyogenes.


Assuntos
Repetições Minissatélites , Polimorfismo Genético , Streptococcus pyogenes/fisiologia , Estreptolisinas/biossíntese , Transativadores/genética , Animais , Proteínas de Bactérias/metabolismo , Sequência de Bases , Modelos Animais de Doenças , Feminino , Regulação Bacteriana da Expressão Gênica , Camundongos , NAD+ Nucleosidase/metabolismo , Fenótipo , Fosforilação , Deleção de Sequência , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/mortalidade , Streptococcus pyogenes/patogenicidade , Estreptolisinas/metabolismo , Virulência/genética
7.
Recent Results Cancer Res ; 209: 95-110, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28101690

RESUMO

For suicide gene therapy, initially prodrug-converting enzymes (gene-directed enzyme-producing therapy, GDEPT) were employed to intracellularly metabolize non-toxic prodrugs into toxic compounds, leading to the effective suicidal killing of the transfected tumor cells. In this regard, the suicide gene therapy has demonstrated its potential for efficient tumor eradication. Numerous suicide genes of viral or bacterial origin were isolated, characterized, and extensively tested in vitro and in vivo, demonstrating their therapeutic potential even in clinical trials to treat cancers of different entities. Apart from this, growing efforts are made to generate more targeted and more effective suicide gene systems for cancer gene therapy. In this regard, bacterial toxins are an alternative to the classical GDEPT strategy, which add to the broad spectrum of different suicide approaches. In this context, lytic bacterial toxins, such as streptolysin O (SLO) or the claudin-targeted Clostridium perfringens enterotoxin (CPE) represent attractive new types of suicide oncoleaking genes. They permit as pore-forming proteins rapid and also selective toxicity toward a broad range of cancers. In this chapter, we describe the generation and use of SLO as well as of CPE-based gene therapies for the effective tumor cell eradication as promising, novel suicide gene approach particularly for treatment of therapy refractory tumors.


Assuntos
Toxinas Bacterianas/genética , Genes Transgênicos Suicidas , Terapia Genética/métodos , Neoplasias/terapia , Animais , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Toxinas Bacterianas/biossíntese , Morte Celular , Toxina Diftérica/biossíntese , Toxina Diftérica/genética , Enterotoxinas/biossíntese , Enterotoxinas/genética , Regulação da Expressão Gênica , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Estreptolisinas/biossíntese , Estreptolisinas/genética
8.
Wei Sheng Wu Xue Bao ; 56(11): 1755-65, 2016 Nov 04.
Artigo em Chinês | MEDLINE | ID: mdl-29741839

RESUMO

Objective: Streptococcus equi subsp. zooepidemicus (GCS) is mainly used to produce hyaluronic acid (HA) in the industry. GCS secretes the hemolysis toxin (streptolysin S, SLS) that causes hemolysis in the host cells. Therefore, the safety of HA produced by GCS is concerned. We constructed an engineering strain, to produce commercial HA without SLS by knocking out saga. Method: The sagA of GCS was knocked out by the thermosensitive delivery vector system pJR700. The sagA mutant was identified through PCR with primers homologous to the flanking regions and SLS analysis. The yield of HA, HA molecular weight and virulence factors such as streptolysin Hylc, hyaluronate lyase, glyceraldehyde-3-phosphate dehydrogenase and cell surface proteins were determined by spectrophotometer and SDS-PAGE. Result: We constructed successfully the in-frame deletion sagA mutant strain of GCS. In the sagA mutant, HA titer increased more than 30% than that of the wild type strain and no SLS hemolytic activity was detected. Compared to the wild type strain the sagA mutant decreased the quality of surface proteins, hemolytic Hylc activity and glyceraldehyde-3-phosphate dehydrogenase activity. The activities of hyaluronidase and cell were increased in the sagA mutant. Conclusion: The sagA not only expressed hemolysis S but also regulated production of HA, the quality of surface proteins and activities of hyaluronidase, hemolysis Hylc and glyceraldehydes-3-phosphate dehydrogenase in Streptococcus equi subsp. zooepidemicus.


Assuntos
Proteínas de Bactérias/genética , Proteínas Hemolisinas/genética , Ácido Hialurônico/biossíntese , Streptococcus equi/genética , Streptococcus equi/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas Hemolisinas/metabolismo , Mutação , Polissacarídeo-Liases/genética , Polissacarídeo-Liases/metabolismo , Estreptolisinas/biossíntese
9.
mBio ; 6(4): e00622, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-26173696

RESUMO

UNLABELLED: Group A Streptococcus (GAS) genotype emm89 is increasingly recognized as a leading cause of disease worldwide, yet factors that underlie the success of this emm type are unknown. Surveillance identified a sustained nationwide increase in emm89 invasive GAS disease in the United Kingdom, prompting longitudinal investigation of this genotype. Whole-genome sequencing revealed a recent dramatic shift in the emm89 population with the emergence of a new clade that increased to dominance over previous emm89 variants. Temporal analysis indicated that the clade arose in the early 1990s but abruptly increased in prevalence in 2008, coinciding with an increased incidence of emm89 infections. Although standard variable typing regions (emm subtype, tee type, sof type, and multilocus sequence typing [MLST]) remained unchanged, uniquely the emergent clade had undergone six distinct regions of homologous recombination across the genome compared to the rest of the sequenced emm89 population. Two of these regions affected known virulence factors, the hyaluronic acid capsule and the toxins NADase and streptolysin O. Unexpectedly, and in contrast to the rest of the sequenced emm89 population, the emergent clade-associated strains were genetically acapsular, rendering them unable to produce the hyaluronic acid capsule. The emergent clade-associated strains had also acquired an NADase/streptolysin O locus nearly identical to that found in emm12 and modern emm1 strains but different from the rest of the sequenced emm89 population. The emergent clade-associated strains had enhanced expression of NADase and streptolysin O. The genome remodeling in the new clade variant and the resultant altered phenotype appear to have conferred a selective advantage over other emm89 variants and may explain the changes observed in emm89 GAS epidemiology. IMPORTANCE: Sudden upsurges or epidemic waves are common features of group A streptococcal disease. Although the mechanisms behind such changes are largely unknown, they are often associated with an expansion of a single genotype within the population. Using whole-genome sequencing, we investigated a nationwide increase in invasive disease caused by the genotype emm89 in the United Kingdom. We identified a new clade variant that had recently emerged in the emm89 population after having undergone several core genomic recombination-related changes, two of which affected known virulence factors. An unusual finding of the new variant was the loss of the hyaluronic acid capsule, previously thought to be essential for causing invasive disease. A further genomic adaptation in the NADase/streptolysin O locus resulted in enhanced production of these toxins. Recombination-related genome remodeling is clearly an important mechanism in group A Streptococcus that can give rise to more successful and potentially more pathogenic variants.


Assuntos
Cápsulas Bacterianas/genética , Genótipo , Infecções Estreptocócicas/epidemiologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/classificação , Streptococcus pyogenes/isolamento & purificação , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Toxinas Bacterianas/genética , Cromossomos Bacterianos , DNA Bacteriano/química , DNA Bacteriano/genética , Monitoramento Epidemiológico , Expressão Gênica , Recombinação Homóloga , Humanos , Estudos Longitudinais , Epidemiologia Molecular , Dados de Sequência Molecular , Tipagem Molecular , NAD+ Nucleosidase/biossíntese , NAD+ Nucleosidase/genética , Prevalência , Análise de Sequência de DNA , Streptococcus pyogenes/genética , Estreptolisinas/biossíntese , Estreptolisinas/genética , Reino Unido/epidemiologia , Fatores de Virulência/genética
10.
ACS Chem Biol ; 10(5): 1217-26, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25668590

RESUMO

Streptolysin S (SLS) is a post-translationally modified peptide cytolysin that is produced by the human pathogen Streptococcus pyogenes. SLS belongs to a large family of azole-containing natural products that are biosynthesized via an evolutionarily conserved pathway. SLS is an important virulence factor during S. pyogenes infections, but despite an extensive history of study, further investigations are needed to clarify several steps of its biosynthesis. To this end, chemical inhibitors of SLS biosynthesis would be valuable tools to interrogate the various maturation steps of both SLS and biosynthetically related natural products. Such chemical inhibitors could also potentially serve as antivirulence therapeutics, which in theory may alleviate the spread of antibiotic resistance. In this work, we demonstrate that FDA-approved HIV protease inhibitors, especially nelfinavir, block a key proteolytic processing step during SLS production. This inhibition was demonstrated in live S. pyogenes cells and through in vitro protease inhibition assays. A panel of 57 nelfinavir analogs was synthesized, leading to a series of compounds with improved anti-SLS activity while illuminating structure-activity relationships. Nelfinavir was also found to inhibit the maturation of other azole-containing natural products, namely those involved in listeriolysin S, clostridiolysin S, and plantazolicin production. The use of nelfinavir analogs as inhibitors of SLS production has allowed us to begin examining the proteolysis event in SLS maturation and will aid in further investigations of the biosynthesis of SLS and related natural products.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Inibidores da Protease de HIV/farmacologia , Estreptolisinas/antagonistas & inibidores , Sequência de Aminoácidos , Ácido Aspártico Proteases/antagonistas & inibidores , Proteínas de Bactérias/biossíntese , Dados de Sequência Molecular , Inibidores de Proteases/farmacologia , Proteólise , Homologia de Sequência de Aminoácidos , Streptococcus pyogenes/efeitos dos fármacos , Streptococcus pyogenes/metabolismo , Estreptolisinas/biossíntese
11.
BMC Biotechnol ; 14: 1, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24400649

RESUMO

BACKGROUND: Streptococcus pneumoniae (S. pneumoniae) causes several serious diseases including pneumonia, septicemia and meningitis. The World Health Organization estimates that streptococcal pneumonia is the cause of approximately 1.9 million deaths of children under five years of age each year. The large number of serotypes underlying the disease spectrum, which would be reflected in the high production cost of a commercial vaccine effective to protect against all of them and the higher level of amino acid sequence conservation as compared to polysaccharide structure, has prompted us to attempt to use conserved proteins for the development of a simpler vaccine. One of the most prominent proteins is pneumolysin (Ply), present in almost all the serotypes known at the moment, which shows an effective protection against S. pneumoniae infections. RESULTS: We have cloned the pneumolysin gene from S. pneumoniae serotype 14 and studied the effects of eight variables related to medium composition and induction conditions on the soluble expression of rPly in Escherichia coli (E. coli) and a 28-4 factorial design was applied. Statistical analysis was carried out to compare the conditions used to evaluate the expression of soluble pneumolysin; rPly activity was evaluated by hemolytic activity assay and served as the main response to evaluate the proper protein expression and folding. The optimized conditions, validated by the use of triplicates, include growth until an absorbance of 0.8 (measured at 600 nm) with 0.1 mM IPTG during 4 h at 25°C in a 5 g/L yeast extract, 5 g/L tryptone, 10 g/L NaCl, 1 g/L glucose medium, with addition of 30 µg/mL kanamycin. CONCLUSIONS: This experimental design methodology allowed the development of an adequate process condition to attain high levels (250 mg/L) of soluble expression of functional rPly in E. coli, which should contribute to reduce operational costs. It was possible to recover the protein in its active form with 75% homogeneity.


Assuntos
Escherichia coli/metabolismo , Proteínas Recombinantes/biossíntese , Estreptolisinas/biossíntese , Proteínas de Bactérias/biossíntese , Biotecnologia/métodos , Clonagem Molecular , Interpretação Estatística de Dados , Escherichia coli/genética , Análise Multivariada , Reprodutibilidade dos Testes , Projetos de Pesquisa , Streptococcus pneumoniae
12.
PLoS Pathog ; 9(6): e1003394, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23762025

RESUMO

Group A Streptococcus (Streptococcus pyogenes or GAS) causes pharyngitis, severe invasive infections, and the post-infectious syndromes of glomerulonephritis and rheumatic fever. GAS can be internalized and killed by epithelial cells in vitro, a process that may contribute to local innate defense against pharyngeal infection. Secretion of the pore-forming toxin streptolysin O (SLO) by GAS has been reported to stimulate targeted autophagy (xenophagy) upon internalization of the bacteria by epithelial cells. Whereas this process was associated with killing of GAS in HeLa cells, studies in human keratinocytes found SLO production enhanced intracellular survival. To reconcile these conflicting observations, we now report in-depth investigation of xenophagy in response to GAS infection of human oropharyngeal keratinocytes, the predominant cell type of the pharyngeal epithelium. We found that SLO expression was associated with prolonged intracellular survival; unexpectedly, expression of the co-toxin NADase was required for this effect. Enhanced intracellular survival was lost upon deletion of NADase or inactivation of its enzymatic activity. Shortly after internalization of GAS by keratinocytes, SLO-mediated damage to the bacteria-containing vacuole resulted in exposure to the cytosol, ubiquitination of GAS and/or associated vacuolar membrane remnants, and engulfment of GAS in LC3-positive vacuoles. We also found that production of streptolysin S could mediate targeting of GAS to autophagosomes in the absence of SLO, a process accompanied by galectin 8 binding to damaged GAS-containing endosomes. Maturation of GAS-containing autophagosome-like vacuoles to degradative autolysosomes was prevented by SLO pore-formation and by SLO-mediated translocation of enzymatically active NADase into the keratinocyte cytosol. We conclude that SLO stimulates xenophagy in pharyngeal keratinocytes, but the coordinated action of SLO and NADase prevent maturation of GAS-containing autophagosomes, thereby prolonging GAS intracellular survival. This novel activity of NADase to block autophagic killing of GAS in pharyngeal cells may contribute to pharyngitis treatment failure, relapse, and chronic carriage.


Assuntos
Autofagia , Células Epiteliais/metabolismo , NAD+ Nucleosidase/metabolismo , Infecções Estreptocócicas/metabolismo , Streptococcus pyogenes/metabolismo , Estreptolisinas/biossíntese , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Linhagem Celular Transformada , Sobrevivência Celular , Citosol/metabolismo , Citosol/microbiologia , Citosol/patologia , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Células HeLa , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/patologia , Queratinócitos/metabolismo , Queratinócitos/microbiologia , Queratinócitos/patologia , Viabilidade Microbiana/genética , NAD+ Nucleosidase/genética , Faringite/metabolismo , Faringite/microbiologia , Faringite/patologia , Faringe/metabolismo , Faringe/microbiologia , Faringe/patologia , Infecções Estreptocócicas/genética , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidade , Estreptolisinas/genética , Ubiquitinação , Vacúolos/metabolismo , Vacúolos/microbiologia , Vacúolos/patologia
13.
J Clin Microbiol ; 51(6): 1962-5, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23515542

RESUMO

We recovered a non-beta-hemolytic Streptococcus pyogenes strain from a severe soft tissue infection. In this isolate, we detected a premature stop codon within the sagC gene of the streptolysin S (SLS) biosynthetic operon. Reintroduction of full-length sagC gene on a plasmid vector restored the beta-hemolytic phenotype to our clinical isolate, indicating that the point mutation in sagC accounted for loss of hemolytic activity. To the best of our knowledge, this is the first report to demonstrate that a severe soft tissue infection can be caused by a non-beta-hemolytic S. pyogenes strain lacking a functional SagC.


Assuntos
Proteínas de Bactérias/genética , Códon sem Sentido , Infecções dos Tecidos Moles/microbiologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidade , Adulto , Proteínas de Bactérias/biossíntese , Vias Biossintéticas/genética , Teste de Complementação Genética , Hemólise , Humanos , Masculino , Análise de Sequência de DNA , Infecções dos Tecidos Moles/patologia , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/isolamento & purificação , Estreptolisinas/biossíntese , Fatores de Virulência/genética
14.
Eur Respir J ; 41(2): 392-5, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22743667

RESUMO

Although the well-recognised predisposition of cigarette smokers to the development of severe pneumococcal disease may be attributable to impairment of local host defences, less is known about the direct effects of smoke exposure on airway pathogens, or their virulence factors. In the current study, we have investigated the effects of cigarette smoke condensate (CSC) on biofilm formation by Streptococcus pneumoniae, and on the pore-forming activity of its major toxin, pneumolysin. Biofilm formation following exposure of the pneumococcus to CSC (20-160 µg·mL(-1)) was measured using a crystal violet-based spectrophotometric procedure, while the pore-forming activity of recombinant pneumolysin was determined by a fura-2/acetoxymethyl ester-based spectrofluorimetric procedure to monitor the uptake of extracellular Ca(2+) by isolated human neutrophils. Exposure of the pneumococcus or pneumolysin to CSC resulted in significant dose-related augmentation of biofilm formation (p≤0.05 at 80 and 160 µg·mL(-1)) and substantial attenuation of the pore-forming interactions of pneumolysin, respectively. Augmentation of biofilm formation and inactivation of pneumolysin as a consequence of smoking are likely to favour microbial colonisation and persistence, both being essential precursors of pneumococcal disease.


Assuntos
Biofilmes , Neutrófilos/microbiologia , Streptococcus pneumoniae/efeitos dos fármacos , Estreptolisinas/biossíntese , Proteínas de Bactérias/biossíntese , Cálcio/metabolismo , Células Cultivadas , Citosol/metabolismo , Corantes Fluorescentes , Fura-2 , Humanos , Neutrófilos/metabolismo , Fumar/efeitos adversos , Espectrometria de Fluorescência
15.
Anal Biochem ; 420(2): 191-3, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22001374

RESUMO

Group A Streptococcus pyogenes (GAS) is a leading human pathogen that produces a powerful cytolytic bacteriocin known as streptolysin S (SLS). We have developed a bioengineering strategy to successfully reconstitute SLS activity using heterologous expression in laboratory strains of Escherichia coli. Our E. coli-based heterologous expression system will allow more detailed studies into the biosynthesis of other bacteriocin compounds and the production of these natural products in much greater yield.


Assuntos
Proteínas de Bactérias/biossíntese , Escherichia coli/genética , Engenharia Genética/métodos , Estreptolisinas/biossíntese , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/toxicidade , Hemólise/efeitos dos fármacos , Família Multigênica/genética , Ovinos , Estreptolisinas/química , Estreptolisinas/genética , Estreptolisinas/toxicidade
16.
PLoS One ; 6(12): e28738, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22174882

RESUMO

It is unclear whether Streptococcus pneumoniae in biofilms are virulent and contribute to development of invasive pneumococcal disease (IPD). Using electron microscopy we confirmed the development of mature pneumococcal biofilms in a continuous-flow-through line model and determined that biofilm formation occurred in discrete stages with mature biofilms composed primarily of dead pneumococci. Challenge of mice with equal colony forming units of biofilm and planktonic pneumococci determined that biofilm bacteria were highly attenuated for invasive disease but not nasopharyngeal colonization. Biofilm pneumococci of numerous serotypes were hyper-adhesive and bound to A549 type II pneumocytes and Detroit 562 pharyngeal epithelial cells at levels 2 to 11-fold greater than planktonic counterparts. Using genomic microarrays we examined the pneumococcal transcriptome and determined that during biofilm formation S. pneumoniae down-regulated genes involved in protein synthesis, energy production, metabolism, capsular polysaccharide (CPS) production, and virulence. We confirmed these changes by measuring CPS by ELISA and immunoblotting for the toxin pneumolysin and the bacterial adhesins phosphorylcholine (ChoP), choline-binding protein A (CbpA), and Pneumococcal serine-rich repeat protein (PsrP). We conclude that biofilm pneumococci were avirulent due to reduced CPS and pneumolysin production along with increased ChoP, which is known to bind C-reactive protein and is opsonizing. Likewise, biofilm pneumococci were hyper-adhesive due to selection for the transparent phase variant, reduced CPS, and enhanced production of PsrP, CbpA, and ChoP. These studies suggest that biofilms do not directly contribute to development of IPD and may instead confer a quiescent mode of growth during colonization.


Assuntos
Biofilmes/crescimento & desenvolvimento , Infecções Pneumocócicas/microbiologia , Infecções Pneumocócicas/patologia , Streptococcus pneumoniae/patogenicidade , Fatores de Virulência/biossíntese , Animais , Anti-Infecciosos/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Cápsulas Bacterianas/biossíntese , Cápsulas Bacterianas/efeitos dos fármacos , Proteínas de Bactérias/biossíntese , Biofilmes/efeitos dos fármacos , Linhagem Celular , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Fenótipo , Plâncton/efeitos dos fármacos , Plâncton/crescimento & desenvolvimento , Reprodutibilidade dos Testes , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/ultraestrutura , Estreptolisinas/biossíntese , Fatores de Tempo , Virulência/efeitos dos fármacos
17.
J Immunol ; 187(9): 4890-9, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21957143

RESUMO

Streptococcus pneumoniae is a Gram-positive, extracellular bacterium that is responsible for significant mortality and morbidity worldwide. Pneumolysin (PLY), a cytolysin produced by all clinical isolates of the pneumococcus, is one of the most important virulence factors of this pathogen. We have previously reported that PLY is an essential factor for activation of caspase-1 and consequent secretion of IL-1ß and IL-18 in macrophages infected with S. pneumoniae. However, the host molecular factors involved in caspase-1 activation are still unclear. To further elucidate the mechanism of caspase-1 activation in macrophages infected with S. pneumoniae, we examined the involvement of inflammasomes in inducing this cellular response. Our study revealed that apoptosis-associated specklike protein containing a caspase recruitment domain (ASC), an adaptor protein for inflammasome receptors such as nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3 (NLRP3) and absent in melanoma 2 (AIM2), is essentially required for the induction of caspase-1 activation by S. pneumoniae. Caspase-1 activation was partially impaired in NLRP3(-/-) macrophages, whereas knockdown and knockout of AIM2 resulted in a clear decrease in caspase-1 activation in response to S. pneumoniae. These results suggest that ASC inflammasomes, including AIM2 and NLRP3, are critical for caspase-1 activation induced by S. pneumoniae. Furthermore, ASC(-/-) mice were more susceptible than wild-type mice to S. pneumoniae, with impaired secretion of IL-1ß and IL-18 into the bronchoalveolar lavage after intranasal infection, suggesting that ASC inflammasomes contribute to the protection of host from infection with PLY-producing S. pneumoniae.


Assuntos
Caspase 1/metabolismo , Proteínas do Citoesqueleto/fisiologia , Imunidade Inata , Inflamassomos/fisiologia , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/biossíntese , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Transporte/fisiologia , Caspase 1/deficiência , Caspase 1/genética , Linhagem Celular , Linhagem Celular Transformada , Células Cultivadas , Proteínas do Citoesqueleto/deficiência , Proteínas do Citoesqueleto/genética , Proteínas de Ligação a DNA , Resistência à Doença/imunologia , Ativação Enzimática/imunologia , Feminino , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas Nucleares/fisiologia , Infecções Pneumocócicas/enzimologia , Estreptolisinas/antagonistas & inibidores , Estreptolisinas/biossíntese
18.
J Immunol ; 187(1): 434-40, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21646297

RESUMO

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.


Assuntos
Proteínas de Transporte/fisiologia , Variação Genética/imunologia , Inflamassomos/metabolismo , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/patologia , Estreptolisinas/genética , Animais , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Feminino , Predisposição Genética para Doença , Humanos , Imunidade Inata/genética , Inflamassomos/fisiologia , Interleucina-18/fisiologia , Interleucina-1beta/biossíntese , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteína Adaptadora de Sinalização NOD2/fisiologia , Pneumonia Pneumocócica/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Estreptolisinas/biossíntese , Estreptolisinas/deficiência , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética , Receptor Toll-Like 9/fisiologia
19.
Antimicrob Agents Chemother ; 54(1): 98-102, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19805566

RESUMO

The administration of high-dose clindamycin (CLI) along with penicillin is recommended for the treatment of streptococcal toxic shock syndrome. However, the prevalence of CLI-resistant Streptococcus pyogenes strains is increasing worldwide, and the effect of CLI on CLI-resistant S. pyogenes strains remains unknown. We aimed to evaluate the effect of CLI on the in vitro production of three major virulent exoproteins, namely, streptolysin O (Slo), NAD glycohydrolase (Nga), and streptokinase (Ska), by CLI-resistant S. pyogenes strains. After the incubation of M1 serotype CLI-resistant S. pyogenes D2TY in the presence of 1 microg/ml CLI, the amounts of Slo, Nga, and Ska and the levels of slo, nga, and ska mRNA in the supernatant were analyzed by Northern blotting and Western blotting, respectively. The results of both assays showed that the production of Slo, Nga, and Ska was higher with CLI treatment than without CLI treatment. We evaluated the role of the sensor kinase CovS, which is involved in the two-component system of S. pyogenes, in the CLI-induced production of these three exoproteins. Northern blotting analysis revealed that CLI induced the expression of covS mRNA in wild-type strain D2TY. Furthermore, both Northern blotting and Western blotting analyses showed that CLI decreased the levels of expression of Slo, Nga, and Ska in isogenic covS mutant D2TYcovS. These results suggest that CLI increases the production of three virulent exoproteins in CLI-resistant S. pyogenes strains via the action of CovS.


Assuntos
Antibacterianos/farmacologia , Clindamicina/farmacologia , Genes Bacterianos/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , NAD+ Nucleosidase/biossíntese , NAD+ Nucleosidase/genética , Streptococcus pyogenes/efeitos dos fármacos , Streptococcus pyogenes/genética , Estreptoquinase/biossíntese , Estreptoquinase/genética , Estreptolisinas/biossíntese , Estreptolisinas/genética , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Northern Blotting , Western Blotting , Farmacorresistência Bacteriana/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Histidina Quinase , Lincosamidas/farmacologia , Testes de Sensibilidade Microbiana , Mutação , RNA Bacteriano/biossíntese , RNA Bacteriano/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Streptococcus pyogenes/enzimologia
20.
PLoS One ; 3(10): e3455, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18941623

RESUMO

Group A streptococcus (GAS) causes variety of diseases ranging from common pharyngitis to life-threatening severe invasive diseases, including necrotizing fasciitis and streptococcal toxic shock-like syndrome. The characteristic of invasive GAS infections has been thought to attribute to genetic changes in bacteria, however, no clear evidence has shown due to lack of an intriguingly study using serotype-matched isolates from clinical severe invasive GAS infections. In addition, rare outbreaks of invasive infections and their distinctive pathology in which infectious foci without neutrophil infiltration hypothesized us invasive GAS could evade host defense, especially neutrophil functions. Herein we report that a panel of serotype-matched GAS, which were clinically isolated from severe invasive but not from non-invaive infections, could abrogate functions of human polymorphnuclear neutrophils (PMN) in at least two independent ways; due to inducing necrosis to PMN by enhanced production of a pore-forming toxin streptolysin O (SLO) and due to impairment of PMN migration via digesting interleukin-8, a PMN attracting chemokine, by increased production of a serine protease ScpC. Expression of genes was upregulated by a loss of repressive function with the mutation of csrS gene in the all emm49 severe invasive GAS isolates. The csrS mutants from clinical severe invasive GAS isolates exhibited high mortality and disseminated infection with paucity of neutrophils, a characteristic pathology seen in human invasive GAS infection, in a mouse model. However, GAS which lack either SLO or ScpC exhibit much less mortality than the csrS-mutated parent invasive GAS isolate to the infected mice. These results suggest that the abilities of GAS to abrogate PMN functions can determine the onset and severity of invasive GAS infection.


Assuntos
Imunocompetência , Neutrófilos/imunologia , Streptococcus pyogenes/patogenicidade , Fatores de Transcrição , Fatores de Virulência/genética , Animais , Proteínas de Bactérias/biossíntese , Glicoproteínas/biossíntese , Proteínas de Choque Térmico/genética , Humanos , Interleucina-8/análise , Camundongos , Proteínas Mutantes , Receptores Depuradores Classe A/genética , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes/imunologia , Estreptolisinas/biossíntese , Ativação Transcricional , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA