Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
Methods Mol Biol ; 2136: 191-198, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32430822

RESUMO

Light and electron microscopy enables researchers to study the ultrastructure of GAS biofilms formed on abiotic surfaces. Chains of streptococci surrounded by a bluish film are seen under a light microscope after alcian blue staining of preparations grown on coverslips. The extracellular matrix (indicator of biofilm maturity) becomes visible on ultrathin sections in transmission electron microscopy after additional staining with alcian blue; filamentous structures, characteristic of biofilm, are observed in intercellular spaces. The data obtained by scanning electron microscopy also demonstrate the presence of biofilm.


Assuntos
Biofilmes/crescimento & desenvolvimento , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/ultraestrutura , Espaço Extracelular/metabolismo , Microscopia Eletrônica de Varredura/métodos , Microscopia Eletrônica de Transmissão/métodos
3.
PLoS One ; 13(12): e0207897, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30517150

RESUMO

Inactivating mutations in the control of virulence two-component regulatory system (covRS) often account for the hypervirulent phenotype in severe, invasive group A streptococcal (GAS) infections. As CovR represses production of the anti-phagocytic hyaluronic acid capsule, high level capsule production is generally considered critical to the hypervirulent phenotype induced by CovRS inactivation. There have recently been large outbreaks of GAS strains lacking capsule, but there are currently no data on the virulence of covRS-mutated, acapsular strains in vivo. We investigated the impact of CovRS inactivation in acapsular serotype M4 strains using a wild-type (M4-SC-1) and a naturally-occurring CovS-inactivated strain (M4-LC-1) that contains an 11bp covS insertion. M4-LC-1 was significantly more virulent in a mouse bacteremia model but caused smaller lesions in a subcutaneous mouse model. Over 10% of the genome showed significantly different transcript levels in M4-LC-1 vs. M4-SC-1 strain. Notably, the Mga regulon and multiple cell surface protein-encoding genes were strongly upregulated-a finding not observed for CovS-inactivated, encapsulated M1 or M3 GAS strains. Consistent with the transcriptomic data, transmission electron microscopy revealed markedly altered cell surface morphology of M4-LC-1 compared to M4-SC-1. Insertional inactivation of covS in M4-SC-1 recapitulated the transcriptome and cell surface morphology. Analysis of the cell surface following CovS-inactivation revealed that the upregulated proteins were part of the Mga regulon. Inactivation of mga in M4-LC-1 reduced transcript levels of multiple cell surface proteins and reversed the cell surface alterations consistent with the effect of CovS inactivation on cell surface composition being mediated by Mga. CovRS-inactivating mutations were detected in 20% of current invasive serotype M4 strains in the United States. Thus, we discovered that hypervirulent M4 GAS strains with covRS mutations can arise in an acapsular background and that such hypervirulence is associated with profound alteration of the cell surface.


Assuntos
Streptococcus pyogenes/patogenicidade , Animais , Cápsulas Bacterianas/genética , Cápsulas Bacterianas/ultraestrutura , Proteínas de Bactérias/genética , Membrana Celular/genética , Membrana Celular/ultraestrutura , Feminino , Genes Bacterianos , Histidina Quinase , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Microscopia Eletrônica de Transmissão , Mutação , Regulon , Proteínas Repressoras/genética , Sorogrupo , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/ultraestrutura , Virulência/genética , Sequenciamento Completo do Genoma
4.
Bull Exp Biol Med ; 165(1): 110-114, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29797119

RESUMO

Our study confirmed the capacity of S. pyogenes strains to form biofilms on abiotic surfaces. Chains of streptococci surrounded by bluish film were seen under a microscope after alcian blue staining of the preparations grown on slides. On ultrathin sections in transmission electron microscope, the extracellular matrix (indicator of biofilm maturity) became visible after staining with alcian blue. Microscopy of the sections shows structures characteristic of a biofilm in spaces between the cells. Scanning electron microscopy also demonstrates the presence of a biomembrane. Importantly that type 1M strain forming in fact no membranes when cultured on plastic plates (Costar) formed biofilms on the glass. It seems that the conditions for the biofilm formation on the plastic and on the glass differ, due to which the exopolymeric matrices formed on different surfaces vary by biochemical composition.


Assuntos
Biofilmes/crescimento & desenvolvimento , Microscopia Eletrônica de Varredura/métodos , Microscopia/métodos , Streptococcus pyogenes/ultraestrutura
5.
Sci Rep ; 6: 31749, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27526944

RESUMO

Salivaricin B is a 25 amino acid polycyclic peptide belonging to the type AII lantibiotics and first shown to be produced by Streptococcus salivarius. In this study we describe the bactericidal mode of action of salivaricin B against susceptible Gram-positive bacteria. The killing action of salivaricin B required micro-molar concentrations of lantibiotic whereas the prototype lantibiotic nisin A was shown to be potent at nano-molar levels. Unlike nisin A, salivaricin B did not induce pore formation or dissipate the membrane potential in susceptible cells. This was established by measuring the fluorescence of the tryptophan residue at position 17 when salivaricin B interacted with bacterial membrane vesicles. The absence of a fluorescence blue shift indicates a failure of salivaricin B to penetrate the membranes. On the other hand, salivaricin B interfered with cell wall biosynthesis, as shown by the accumulation of the final soluble cell wall precursor UDP-MurNAc-pentapeptide which is the backbone of the bacterial peptidoglycan. Transmission electron microscopy of salivaricin B-treated cells showed a reduction in cell wall thickness together with signs of aberrant septum formation in the absence of visible changes to cytoplasmic membrane integrity.


Assuntos
Antibacterianos/farmacologia , Bacteriocinas/farmacologia , Bactérias Gram-Positivas/efeitos dos fármacos , Sequência de Aminoácidos , Antibacterianos/química , Proteínas de Bactérias/química , Proteínas de Bactérias/farmacologia , Bacteriocinas/química , Parede Celular/efeitos dos fármacos , Parede Celular/ultraestrutura , Bactérias Gram-Positivas/ultraestrutura , Testes de Sensibilidade Microbiana , Micrococcus luteus/efeitos dos fármacos , Micrococcus luteus/ultraestrutura , Microscopia Eletrônica de Transmissão , Streptococcus pyogenes/efeitos dos fármacos , Streptococcus pyogenes/ultraestrutura
6.
J Innate Immun ; 8(3): 243-57, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26641861

RESUMO

Reports have shown that the antimicrobial peptide LL-37 is abundantly expressed but has limited bactericidal effect in Streptococcus pyogenes infections. At sub-inhibitory concentrations, LL-37 has been reported to alter virulence gene expression. Here, we explored the interaction of S. pyogenes strains with LL-37, focusing on bacterial growth, cell surface alterations and pro-inflammatory responses. Bioscreen turbidity measurements of strain 5448 cultured in the presence or absence of LL-37 confirmed the poor antimicrobial effect, and revealed a significant increase in turbidity of bacterial cultures exposed to sub-inhibitory concentrations of LL-37. However, this was not linked to increased bacterial counts. Electron microscopy of LL-37-exposed bacteria revealed the presence of vesicle-like structures on the bacterial surface. The vesicles stained positive for LL-37 and were released from the bacterial surface. Concentrated supernatants enriched in these structures had a broader protein content, including several virulence factors, compared to supernatants from untreated bacteria. The supernatants from LL-37-exposed bacteria were pro-inflammatory and elicited resistin and myeloperoxidase release from neutrophils. This is the first report on S. pyogenes extracellular vesicle-like structures formed at the bacterial surface in response to LL-37. The associated increased pro-inflammatory activity further implicates LL-37 as a potential factor involved in S. pyogenes pathogenesis.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Vesículas Extracelulares/metabolismo , Mediadores da Inflamação/metabolismo , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes/fisiologia , Processos de Crescimento Celular , Vesículas Extracelulares/ultraestrutura , Humanos , Imunização , Ativação de Neutrófilo , Neutrófilos/imunologia , Peroxidase/metabolismo , Resistina/metabolismo , Streptococcus pyogenes/ultraestrutura , Catelicidinas
7.
PLoS Pathog ; 9(8): e1003529, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935504

RESUMO

Previous studies have shown that stimulation of whole blood or peripheral blood mononuclear cells with bacterial virulence factors results in the sequestration of pro-coagulant microvesicles (MVs). These particles explore their clotting activity via the extrinsic and intrinsic pathway of coagulation; however, their pathophysiological role in infectious diseases remains enigmatic. Here we describe that the interaction of pro-coagulant MVs with bacteria of the species Streptococcus pyogenes is part of the early immune response to the invading pathogen. As shown by negative staining electron microscopy and clotting assays, pro-coagulant MVs bind in the presence of plasma to the bacterial surface. Fibrinogen was identified as a linker that, through binding to the M1 protein of S. pyogenes, allows the opsonization of the bacteria by MVs. Surface plasmon resonance analysis revealed a strong interaction between pro-coagulant MVs and fibrinogen with a KD value in the nanomolar range. When performing a mass-spectrometry-based strategy to determine the protein quantity, a significant up-regulation of the fibrinogen-binding integrins CD18 and CD11b on pro-coagulant MVs was recorded. Finally we show that plasma clots induced by pro-coagulant MVs are able to prevent bacterial dissemination and possess antimicrobial activity. These findings were confirmed by in vivo experiments, as local treatment with pro-coagulant MVs dampens bacterial spreading to other organs and improved survival in an invasive streptococcal mouse model of infection. Taken together, our data implicate that pro-coagulant MVs play an important role in the early response of the innate immune system in infectious diseases.


Assuntos
Coagulação Sanguínea/imunologia , Antígeno CD11b/imunologia , Antígenos CD18/imunologia , Micropartículas Derivadas de Células/imunologia , Infecções Estreptocócicas/metabolismo , Streptococcus pyogenes/imunologia , Animais , Antígeno CD11b/metabolismo , Antígenos CD18/metabolismo , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/microbiologia , Micropartículas Derivadas de Células/ultraestrutura , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/ultraestrutura
8.
Indian J Med Res ; 137(5): 963-71, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23760384

RESUMO

BACKGROUND & OBJECTIVES: Subinhibitory concentrations (sub-MICs) of antibiotics, although not able to kill bacteria, but influence bacterial virulence significantly. Fluoroquinolones (FQs) which are used against other bacterial pathogens creates resistance in non-targeted Streptococcus pyogenes. This study was undertaken to characterize the effect of sub-MICs of FQs on S. pyogenes biofilm formation. METHODS: Biofilm forming six M serotypes M56, st38, M89, M65, M100 and M74 of S. pyogenes clinical isolates were challenged against four FQs namely, ciprofloxacin, ofloxacin, levofloxacin and norfloxacin. The antibiofilm potential of these FQs was analysed at their subinhibitory concentrations (1/2 to 1/64 MIC) using biofilm assay, XTT reduction assay, scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). RESULTS: Among the four FQs tested, ofloxacin and levofloxacin at 1/2 MIC showed the maximum inhibition (92%) of biofilm formation against M56 and M74 serotypes. FQs effectively interfered in the microcolony formation of S. pyogenes isolates at 1/2 to 1/8 sub-MICs. Inhibition of biofilm formation was greatly reduced beyond 1/16 MICs and allowed biofilm formation. XTT reduction assay revealed the increase in metabolic activity of S. pyogenes biofilm against the decrease in FQs concentration. SEM and CLSM validated the potential of sub-MICs of FQs against the six S. pyogenes. INTERPRETATION & CONCLUSIONS: Our results showed that the inhibitory effect all four FQs on S. pyogenes biofilm formation was concentration dependent. FQs at proper dosage can be effective against S. pyogenes and lower concentrations may allow the bacteria to form barriers against the antibiotic in the form of biofilm.


Assuntos
Biofilmes/efeitos dos fármacos , Fluoroquinolonas/farmacologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/efeitos dos fármacos , Antibacterianos/uso terapêutico , Biofilmes/crescimento & desenvolvimento , Ciprofloxacina/farmacologia , Humanos , Levofloxacino/farmacologia , Testes de Sensibilidade Microbiana , Microscopia Confocal , Microscopia Eletrônica de Varredura , Norfloxacino/farmacologia , Ofloxacino/farmacologia , Infecções Estreptocócicas/tratamento farmacológico , Streptococcus pyogenes/crescimento & desenvolvimento , Streptococcus pyogenes/patogenicidade , Streptococcus pyogenes/ultraestrutura , Virulência/efeitos dos fármacos
9.
J R Soc Interface ; 10(80): 20120892, 2013 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-23303219

RESUMO

Gram-positive bacteria can transport molecules necessary for their survival through holes in their cell wall. The holes in cell walls need to be large enough to let critical nutrients pass through. However, the cell wall must also function to prevent the bacteria's membrane from protruding through a large hole into the environment and lysing the cell. As such, we hypothesize that there exists a range of cell wall hole sizes that allow for molecule transport but prevent membrane protrusion. Here, we develop and analyse a biophysical theory of the response of a Gram-positive cell's membrane to the formation of a hole in the cell wall. We predict a critical hole size in the range of 15-24 nm beyond which lysis occurs. To test our theory, we measured hole sizes in Streptococcus pyogenes cells undergoing enzymatic lysis via transmission electron microscopy. The measured hole sizes are in strong agreement with our theoretical prediction. Together, the theory and experiments provide a means to quantify the mechanisms of death of Gram-positive cells via enzymatically mediated lysis and provides insights into the range of cell wall hole sizes compatible with bacterial homeostasis.


Assuntos
Estruturas da Membrana Celular/fisiologia , Parede Celular/fisiologia , Streptococcus pyogenes/fisiologia , Transporte Biológico Ativo/fisiologia , Estruturas da Membrana Celular/ultraestrutura , Parede Celular/ultraestrutura , Microscopia Eletrônica de Transmissão , Streptococcus pyogenes/ultraestrutura
10.
J Exp Med ; 209(13): 2367-81, 2012 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-23230002

RESUMO

Several of the most significant bacterial pathogens in humans, including Streptococcus pyogenes, express surface proteins that bind IgG antibodies via their fragment crystallizable (Fc) region, and the dogma is that this protects the bacteria against phagocytic killing in blood. However, analysis of samples from a patient with invasive S. pyogenes infection revealed dramatic differences in the presence and orientation of IgG antibodies at the surface of bacteria from different sites. In the throat, IgG was mostly bound to the bacterial surface via Fc, whereas in the blood IgG was mostly bound via fragment antigen-binding (Fab). In infected and necrotic tissue, the Fc-binding proteins were removed from the bacterial surface. Further investigation showed that efficient bacterial IgGFc-binding occurs only in IgG-poor environments, such as saliva. As a consequence, the bacteria are protected against phagocytic killing, whereas in blood plasma where the concentration of IgG is high, the antibodies preferentially bind via Fab, facilitating opsonization and bacterial killing. IgG-poor environments represent the natural habitat for IgGFc-binding bacteria, and IgGFc-binding proteins may have evolved to execute their function in such environments. The lack of protection in plasma also helps to explain why cases of severe invasive infections with IgGFc-binding bacteria are so rare compared with superficial and uncomplicated infections.


Assuntos
Anticorpos Antibacterianos/metabolismo , Fasciite Necrosante/imunologia , Fasciite Necrosante/microbiologia , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/imunologia , Streptococcus pyogenes/patogenicidade , Adulto , Sequência de Aminoácidos , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/classificação , Anticorpos Antibacterianos/genética , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Região Branquial/imunologia , Região Branquial/microbiologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Membrana Celular/imunologia , Proteínas do Sistema Complemento/metabolismo , Fasciite Necrosante/genética , Feminino , Humanos , Fragmentos Fab das Imunoglobulinas/metabolismo , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulina G/sangue , Imunoglobulina G/classificação , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Microscopia Imunoeletrônica , Dados de Sequência Molecular , Neutrófilos/imunologia , Neutrófilos/microbiologia , Fagocitose , Homologia de Sequência de Aminoácidos , Choque Séptico/genética , Choque Séptico/imunologia , Choque Séptico/microbiologia , Infecções Estreptocócicas/genética , Streptococcus pyogenes/genética , Streptococcus pyogenes/ultraestrutura
11.
J Mol Med (Berl) ; 90(10): 1197-207, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22527883

RESUMO

Streptococcus pyogenes (group A Streptococcus (GAS)) causes ∼700 million human infections each year, resulting in over 500,000 deaths. The development of a commercial GAS vaccine is hampered by the occurrence of many unique GAS serotypes, antigenic variation within the same serotype, differences in serotype geographical distribution, and the production of antibodies cross-reactive with human tissue that may lead to autoimmune disease. Several independent studies have documented a number of GAS cell wall-associated or secreted metabolic enzymes that contain neither N-terminal leader sequences nor C-terminal cell wall anchors. Here, we applied a proteomic analysis of serotype M1T1 GAS cell wall extracts for the purpose of vaccine development. This approach catalogued several anchorless proteins and identified two protective vaccine candidates, arginine deiminase and trigger factor. These surface-exposed enzymes are expressed across multiple GAS serotypes exhibiting ≥99% amino acid sequence identity. Vaccine safety concerns are alleviated by the observation that these vaccine candidates lack human homologs, while sera from human populations suffering repeated GAS infections and high levels of autoimmune complications do not recognize these enzymes. Our study demonstrates anchorless cell surface antigens as promising vaccine candidates for the prevention of GAS disease.


Assuntos
Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Hidrolases/metabolismo , Peptidilprolil Isomerase/metabolismo , Infecções Estreptocócicas/prevenção & controle , Vacinas Estreptocócicas/metabolismo , Streptococcus pyogenes/imunologia , Adolescente , Animais , Proteínas de Bactérias/imunologia , Parede Celular/imunologia , Criança , Feminino , Humanos , Hidrolases/imunologia , Soros Imunes/imunologia , Imunidade Ativa , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia Imunoeletrônica , Peptidilprolil Isomerase/imunologia , Proteoma/imunologia , Proteoma/metabolismo , Coelhos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Infecções Estreptocócicas/imunologia , Vacinas Estreptocócicas/administração & dosagem , Vacinas Estreptocócicas/imunologia , Streptococcus pyogenes/enzimologia , Streptococcus pyogenes/ultraestrutura , Vacinação , Adulto Jovem
12.
J Biol Chem ; 286(43): 37566-77, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-21880740

RESUMO

The human pathogen Streptococcus pyogenes produces diverse pili depending on the serotype. We investigated the assembly mechanism of FCT type 1 pili in a serotype M6 strain. The pili were found to be assembled from two precursor proteins, the backbone protein T6 and ancillary protein FctX, and anchored to the cell wall in a manner that requires both a housekeeping sortase enzyme (SrtA) and pilus-associated sortase enzyme (SrtB). SrtB is primarily required for efficient formation of the T6 and FctX complex and subsequent polymerization of T6, whereas proper anchoring of the pili to the cell wall is mainly mediated by SrtA. Because motifs essential for polymerization of pilus backbone proteins in other Gram-positive bacteria are not present in T6, we sought to identify the functional residues involved in this process. Our results showed that T6 encompasses the novel VAKS pilin motif conserved in streptococcal T6 homologues and that the lysine residue (Lys-175) within the motif and cell wall sorting signal of T6 are prerequisites for isopeptide linkage of T6 molecules. Because Lys-175 and the cell wall sorting signal of FctX are indispensable for substantial incorporation of FctX into the T6 pilus shaft, FctX is suggested to be located at the pilus tip, which was also implied by immunogold electron microscopy findings. Thus, the elaborate assembly of FCT type 1 pili is potentially organized by sortase-mediated cross-linking between sorting signals and the amino group of Lys-175 positioned in the VAKS motif of T6, thereby displaying T6 and FctX in a temporospatial manner.


Assuntos
Parede Celular/metabolismo , Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/metabolismo , Multimerização Proteica/fisiologia , Streptococcus pyogenes/metabolismo , Motivos de Aminoácidos , Sequência de Bases , Parede Celular/genética , Parede Celular/ultraestrutura , Proteínas de Fímbrias/genética , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/ultraestrutura , Humanos , Dados de Sequência Molecular , Streptococcus pyogenes/genética , Streptococcus pyogenes/ultraestrutura
13.
PLoS One ; 6(5): e20345, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21655249

RESUMO

Streptococcus pyogenes is an important human pathogen and surface structures allow it to adhere to, colonize and invade the human host. Proteins containing leucine rich repeats (LRR) have been identified in mammals, viruses, archaea and several bacterial species. The LRRs are often involved in protein-protein interaction, are typically 20-30 amino acids long and the defining feature of the LRR motif is an 11-residue sequence LxxLxLxxNxL (x being any amino acid). The streptococcal leucine rich (Slr) protein is a hypothetical lipoprotein that has been shown to be involved in virulence, but at present no ligands for Slr have been identified. We could establish that Slr is a membrane attached horseshoe shaped lipoprotein by homology modeling, signal peptidase II inhibition, electron microscopy (of bacteria and purified protein) and immunoblotting. Based on our previous knowledge of LRR proteins we hypothesized that Slr could mediate binding to collagen. We could show by surface plasmon resonance that recombinant Slr and purified M1 protein bind with high affinity to collagen I. Isogenic slr mutant strain (MB1) and emm1 mutant strain (MC25) had reduced binding to collagen type I as shown by slot blot and surface plasmon resonance. Electron microscopy using gold labeled Slr showed multiple binding sites to collagen I, both to the monomeric and the fibrillar structure, and most binding occurred in the overlap region of the collagen I fibril. In conclusion, we show that Slr is an abundant membrane bound lipoprotein that is co-expressed on the surface with M1, and that both these proteins are involved in recruiting collagen type I to the bacterial surface. This underlines the importance of S. pyogenes interaction with extracellular matrix molecules, especially since both Slr and M1 have been shown to be virulence factors.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Colágeno Tipo I/metabolismo , Lipoproteínas/metabolismo , Streptococcus pyogenes/metabolismo , Western Blotting , Parede Celular/metabolismo , Microscopia Eletrônica de Transmissão , Ligação Proteica , Streptococcus pyogenes/ultraestrutura , Ressonância de Plasmônio de Superfície
14.
Clin Chim Acta ; 411(17-18): 1172-80, 2010 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-20452338

RESUMO

Group A streptococci (GAS, Streptococcus pyogenes) are exclusive human pathogens that have been extensively studied for many decades. The spectrum of diseases caused by these bacteria ranges from uncomplicated and superficial to severe and invasive infections. In order to give rise to these complications, GAS have evolved a number of surface-bound and secreted virulence factors, of which the M proteins are probably the best characterized. Evidence has emerged that M proteins are multifunctional pathogenic determinants, and over the years many interactions between M proteins and the human host have been reported. The present review article aims to present a state-of-the-art overview of the most important virulence mechanisms employed by M proteins to trigger disease.


Assuntos
Proteínas de Bactérias/fisiologia , Streptococcus pyogenes/patogenicidade , Virulência , Autofagia , Aderência Bacteriana , Proteínas de Bactérias/química , Hemostasia , Humanos , Microscopia Eletrônica de Transmissão , Mimetismo Molecular , Conformação Proteica , Streptococcus pyogenes/ultraestrutura
15.
J Biol Chem ; 285(29): 22666-75, 2010 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-20472552

RESUMO

Streptococcus pyogenes (group A streptococcus (GAS)) is a pathogen that invades non-phagocytic host cells, and causes a variety of acute infections such as pharyngitis. Our group previously reported that intracellular GAS is effectively degraded by the host-cell autophagic machinery, and that a cholesterol-dependent cytolysin, streptolysin O (SLO), is associated with bacterial escape from endosomes in epithelial cells. However, the details of both the intracellular behavior of GAS and the process leading to its autophagic degradation remain unknown. In this study, we found that two host small G proteins, Rab5 and Rab7, were associated with the pathway of autophagosome formation and the fate of intracellular GAS. Rab5 was involved in bacterial invasion and endosome fusion. Rab7 was clearly multifunctional, with roles in bacterial invasion, endosome maturation, and autophagosome formation. In addition, this study showed that the bacterial cytolysin SLO supported the escape of GAS into the cytoplasm from endosomes, and surprisingly, a SLO-deficient mutant of GAS was viable longer than the wild-type strain although it failed to escape the endosomes. This intracellular behavior of GAS is unique and distinct from that of other types of bacterial invaders. Our results provide a new picture of GAS infection and host-cell responses in epithelial cells.


Assuntos
Autofagia , Espaço Intracelular/microbiologia , Estreptolisinas/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Proteínas de Bactérias/metabolismo , Biomarcadores/metabolismo , Endossomos/microbiologia , Endossomos/ultraestrutura , Células HeLa , Humanos , Espaço Intracelular/metabolismo , Lisossomos/microbiologia , Lisossomos/ultraestrutura , Viabilidade Microbiana , Modelos Biológicos , Proteínas Mutantes/metabolismo , Fagossomos/metabolismo , Fagossomos/microbiologia , Fagossomos/ultraestrutura , Transporte Proteico , Streptococcus pyogenes/citologia , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/ultraestrutura , Fatores de Tempo , proteínas de unión al GTP Rab7
16.
J Invest Dermatol ; 130(5): 1365-72, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20107486

RESUMO

Bacterial skin infections, such as erysipelas or cellulitis, are characterized by fever and a painful erythematous rash. Despite the high prevalence of these infections, little is known about the underlying pathogenic mechanisms. This is partly due to the fact that a bacterial diagnosis is often difficult to attain. To gain insight into the pathogenesis of erysipelas, we investigated the samples obtained from infected and noninfected areas of skin from 12 patients with erysipelas. Bacterial cultures, detection of specific streptococcal antibodies in convalescent sera, and immunohistochemical analyses of biopsies indicated group A streptococcal etiology in 11 of the 12 patients. Also, electron micrographs of erythematous skin confirmed the presence of group A streptococcal cells and showed a limited solubilization of the surface-attached M protein. Degradation of high-molecular-weight kininogen and upregulation of the bradykinin-1 receptor in inflamed tissues indicated activation of the contact system in 11 patients. Analyses of release of the vasoactive heparin-binding protein (HBP) showed increased levels in the infected as compared with the noninfected areas. The results suggest that group A streptococci induce contact activation and HBP release during skin infection, which likely contribute to the symptoms seen in erysipelas: fever, pain, erythema, and edema.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas Sanguíneas/metabolismo , Proteínas de Transporte/metabolismo , Erisipela/imunologia , Erisipela/microbiologia , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/metabolismo , Streptococcus pyogenes/metabolismo , Doença Aguda , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Peptídeos Catiônicos Antimicrobianos/imunologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Biópsia , Proteínas Sanguíneas/imunologia , Proteínas de Transporte/imunologia , Erisipela/patologia , Humanos , Cininogênio de Alto Peso Molecular/metabolismo , Microscopia Eletrônica , Pessoa de Meia-Idade , Receptor B1 da Bradicinina/metabolismo , Pele/imunologia , Pele/microbiologia , Pele/patologia , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/ultraestrutura
17.
Autophagy ; 6(2): 288-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20104023

RESUMO

Autophagy functions as a cell-autonomous effector mechanism of innate immunity by separating bacteria from cytosolic resources and delivering them for lysosomal destruction. How cytosolic bacteria are targeted for autophagy is incompletely understood. We recently discovered that Salmonella enterica serotype Typhimurium and Streptococcus pyogenes are detected by NDP52 (nuclear dot protein 52 kDa), after these bacteria enter the cytosol of human cells and become decorated with polyubiquitinated proteins. NDP52 binds the bacterial ubiquitin coat as well as ATG8/LC3 and delivers cytosolic bacteria into autophagosomes. In the absence of NDP52 ubiquitin-coated bacteria accumulate outside ATG8/LC3(+) autophagosomes. Cells lacking NDP52 fail to restrict bacterial proliferation, as do cells depleted of TBK1, an IKK family kinase colocalizing with NDP52 at the bacterial surface. Our findings demonstrate the existence of a receptor for the selective autophagy of cytosolic bacteria, suggesting that cells are able to differentiate between antibacterial and other forms of autophagy.


Assuntos
Autofagia/fisiologia , Proteínas Nucleares/metabolismo , Salmonella typhimurium/metabolismo , Streptococcus pyogenes/metabolismo , Ubiquitina/metabolismo , Citosol/metabolismo , Humanos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Salmonella typhimurium/citologia , Streptococcus pyogenes/ultraestrutura
18.
Antimicrob Agents Chemother ; 54(1): 221-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19858262

RESUMO

Phim46.1, the recognized representative of the most common variant of mobile, prophage-associated genetic elements carrying resistance genes mef(A) (which confers efflux-mediated erythromycin resistance) and tet(O) (which confers tetracycline resistance) in Streptococcus pyogenes, was fully characterized. Sequencing of the Phim46.1 genome (55,172 bp) demonstrated a modular organization typical of tailed bacteriophages. Electron microscopic analysis of mitomycin-induced Phim46.1 revealed phage particles with the distinctive icosahedral head and tail morphology of the Siphoviridae family. The chromosome integration site was within a 23S rRNA uracil methyltransferase gene. BLASTP analysis revealed that the proteins of Phim46.1 had high levels of amino acid sequence similarity to the amino acid sequences of proteins from other prophages, especially Phi10394.4 of S. pyogenes and lambdaSa04 of S. agalactiae. Phage DNA was present in the host cell both as a prophage and as free circular DNA. The lysogeny module appears to have been split due to the insertion of a segment containing tet(O) (from integrated conjugative element 2096-RD.2) and mef(A) (from a Tn1207.1-like transposon) into the unintegrated phage DNA. The phage attachment sequence lies in the region between tet(O) and mef(A) in the unintegrated form. Thus, whereas in this form tet(O) is approximately 5.5 kb upstream of mef(A), in the integrated form, tet(O), which lies close to the right end of the prophage, is approximately 46.3 kb downstream of mef(A), which lies close to the left end of the prophage.


Assuntos
Proteínas de Bactérias/genética , Bacteriófagos/genética , Genes Bacterianos/genética , Proteínas de Membrana/genética , Streptococcus pyogenes/genética , Sítios de Ligação Microbiológicos/genética , Bacteriófagos/ultraestrutura , Sequência de Bases , Elementos de DNA Transponíveis/genética , DNA Circular/genética , DNA Viral/genética , Bases de Dados Genéticas , Genoma Viral , Dados de Sequência Molecular , Prófagos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Streptococcus pyogenes/ultraestrutura
19.
Curr Microbiol ; 60(6): 454-60, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20020301

RESUMO

Streptococcus pyogenes biofilms tend to exhibit significant tolerance to antimicrobials during infections. We screened coral-associated actinomycetes (CAA) for antibiofilm activity against different biofilm forming M serotype of Streptococcus pyogenes. Actinomycetes isolated from the mucus of the coral Acropora digitifera were screened for antibiofilm activity against S. pyogenes biofilms wherein several isolates clearly demonstrated antibiofilm activity. The biofilm inhibitory concentrations (BICs) and the sub-BICs (1/2 and 1/4 BIC) of the extracts significantly prevented biofilm formation up to 60-80%. The extract of Streptomyces akiyoshinensis (A3) displayed efficient antibiofilm activity against all the biofilm forming M serotypes. All the five extracts efficiently reduced the cell surface hydrophobicity (a crucial factor for biofilm formation in S. pyogenes) of three M types and thus may inhibit biofilm formation. CAA represent an interesting source of marine invertebrates-derived antibiofilm agents in the development of new strategies to combat Streptococcal biofilms.


Assuntos
Actinobacteria/química , Antozoários/microbiologia , Anti-Infecciosos/farmacologia , Biofilmes/efeitos dos fármacos , Streptococcus pyogenes/efeitos dos fármacos , Actinobacteria/isolamento & purificação , Actinobacteria/fisiologia , Animais , Biofilmes/crescimento & desenvolvimento , Interações Hidrofóbicas e Hidrofílicas , Testes de Sensibilidade Microbiana , Streptococcus pyogenes/fisiologia , Streptococcus pyogenes/ultraestrutura
20.
Immunol Cell Biol ; 87(5): 391-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19417770

RESUMO

Infections caused by group A streptococcus (GAS) represent a public health problem in both developing and developed countries. The current available methods of prevention are either inadequate or ineffective, which is highlighted by the resurgence in invasive GAS infections over the past two decades. The management of GAS and associated diseases requires new and improved approaches. This review discusses various potential approaches in controlling GAS infections, ranging from prophylactic vaccines to antibody immunotherapy.


Assuntos
Infecções Estreptocócicas/imunologia , Vacinas Estreptocócicas/imunologia , Streptococcus pyogenes/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Humanos , Imunização/métodos , Imunização/tendências , Imunoterapia/métodos , Imunoterapia/tendências , Microscopia Eletrônica , Infecções Estreptocócicas/prevenção & controle , Infecções Estreptocócicas/terapia , Vacinas Estreptocócicas/administração & dosagem , Streptococcus pyogenes/ultraestrutura , Vacinas de Subunidades Antigênicas/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...