Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Benef Microbes ; 9(3): 429-439, 2018 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-29633637

RESUMO

Probiotics, mainly lactic acid bacteria (LAB), are widely focused on gastrointestinal applications. However, recent microbiome studies indicate that LAB can be endogenous members of other human body sites such as the upper respiratory tract (URT). Interestingly, DNA-based microbiome research suggests an inverse correlation between the presence of LAB and the occurrence of potential pathogens, such as Moraxella catarrhalis, an important URT pathogen linked to otitis media, sinusitis and chronic obstructive pulmonary disease. However, a direct interaction between these microbes has not been explored in detail. This study investigated the direct antipathogenic effects of Lactobacillus species, including several well-documented probiotic strains, on M. catarrhalis using agar-based assays, time course analysis, biofilm assays and minimal inhibitory concentration (MIC) testing. These assays were performed using spent culture supernatans (SCS) at two pHs (4.3 and 7) and D- and/or L-lactic acid at three pHs (2, 4 and 7). In addition, cell line assays for adhesion competition and immunomodulation were used to substantiate the inhibitory effect of lactobacilli against M. catarrhalis. A proportion of Lactobacillus strains, including the model probiotic Lactobacillus rhamnosus GG, showed a strong and direct activity against M. catarrhalis. Screening of the activity of the SCS after different treatments demonstrated that lactic acid has an important antimicrobial activity against this pathogen - at least in vitro - with mean MIC values for D- and L-lactic acid varying between 0.5 and 27 g/l depending on the pH. Furthermore, L. rhamnosus GG also decreased the adhesion of M. catarrhalis to human airway epithelial Calu-3 cells with more than 50%, and the expression of mucin MUC5AC, pro-inflammatory cytokines interleukin (IL)-8, IL-1ß, and tumor necrosis factor-α at least 1.2 fold. This study suggests that several lactobacilli and their key metabolite lactic acid are possible candidates for probiotic therapeutic interventions against URT infections.


Assuntos
Antibiose , Biofilmes/crescimento & desenvolvimento , Lactobacillus/crescimento & desenvolvimento , Moraxella catarrhalis/crescimento & desenvolvimento , Antibacterianos/metabolismo , Aderência Bacteriana , Linhagem Celular , Humanos , Ácido Láctico/metabolismo , Lactobacillus/metabolismo , Técnicas Microbiológicas
2.
Eur J Clin Microbiol Infect Dis ; 35(9): 1441-7, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27234592

RESUMO

Recent studies have demonstrated that anti-staphylococcal beta-lactam antibiotics, like nafcillin, render methicillin-resistant Staphylococcus aureus (MRSA) more susceptible to killing by innate host defense peptides (HDPs), such as cathelicidin LL-37. We compared the effects of growth in 1/4 minimum inhibitory concentration (MIC) of nafcillin or vancomycin on the LL-37 killing of 92 methicillin-susceptible S. aureus (MSSA) isolates. For three randomly selected strains among these, we examined the effects of nafcillin, vancomycin, daptomycin, or linezolid on LL-37 killing and autolysis. Growth in the presence of subinhibitory nafcillin significantly enhanced LL-37 killing of MSSA compared to vancomycin and antibiotic-free controls. Nafcillin also reduced MSSA production of the golden staphylococcal pigment staphyloxanthin in 39 % of pigmented strains vs. 14 % for vancomycin. Among the antibiotics tested, only nafcillin resulted in significantly increased MSSA autolysis. These studies point to additional mechanisms of anti-staphylococcal activity of nafcillin beyond direct bactericidal activity, properties that vancomycin and other antibiotic classes do not exhibit. The ability of nafcillin to enhance sensitivity to innate HDPs may contribute to its superior effectiveness against MSSA, as suggested by studies comparing clinical outcomes to vancomycin treatment.


Assuntos
Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Bacteriemia/microbiologia , Viabilidade Microbiana/efeitos dos fármacos , Nafcilina/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Vancomicina/farmacologia , Bacteriólise/efeitos dos fármacos , Interações Medicamentosas , Humanos , Testes de Sensibilidade Microbiana , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/isolamento & purificação , Staphylococcus aureus/fisiologia , Catelicidinas
3.
EBioMedicine ; 4: 95-103, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26981574

RESUMO

Successful treatment of bacterial infections requires the timely administration of appropriate antimicrobial therapy. The failure to initiate the correct therapy in a timely fashion results in poor clinical outcomes, longer hospital stays, and higher medical costs. Current approaches to antibiotic susceptibility testing of cultured pathogens have key limitations ranging from long run times to dependence on prior knowledge of genetic mechanisms of resistance. We have developed a rapid antimicrobial susceptibility assay for Staphylococcus aureus based on bacterial cytological profiling (BCP), which uses quantitative fluorescence microscopy to measure antibiotic induced changes in cellular architecture. BCP discriminated between methicillin-susceptible (MSSA) and -resistant (MRSA) clinical isolates of S. aureus (n = 71) within 1-2 h with 100% accuracy. Similarly, BCP correctly distinguished daptomycin susceptible (DS) from daptomycin non-susceptible (DNS) S. aureus strains (n = 20) within 30 min. Among MRSA isolates, BCP further identified two classes of strains that differ in their susceptibility to specific combinations of beta-lactam antibiotics. BCP provides a rapid and flexible alternative to gene-based susceptibility testing methods for S. aureus, and should be readily adaptable to different antibiotics and bacterial species as new mechanisms of resistance or multidrug-resistant pathogens evolve and appear in mainstream clinical practice.


Assuntos
Técnicas de Tipagem Bacteriana/métodos , Farmacorresistência Bacteriana , Staphylococcus aureus Resistente à Meticilina/citologia , Antibacterianos/farmacologia , Daptomicina/farmacologia , Staphylococcus aureus Resistente à Meticilina/classificação , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos
4.
Nature ; 496(7444): 238-42, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-23535595

RESUMO

Macrophages activated by the Gram-negative bacterial product lipopolysaccharide switch their core metabolism from oxidative phosphorylation to glycolysis. Here we show that inhibition of glycolysis with 2-deoxyglucose suppresses lipopolysaccharide-induced interleukin-1ß but not tumour-necrosis factor-α in mouse macrophages. A comprehensive metabolic map of lipopolysaccharide-activated macrophages shows upregulation of glycolytic and downregulation of mitochondrial genes, which correlates directly with the expression profiles of altered metabolites. Lipopolysaccharide strongly increases the levels of the tricarboxylic-acid cycle intermediate succinate. Glutamine-dependent anerplerosis is the principal source of succinate, although the 'GABA (γ-aminobutyric acid) shunt' pathway also has a role. Lipopolysaccharide-induced succinate stabilizes hypoxia-inducible factor-1α, an effect that is inhibited by 2-deoxyglucose, with interleukin-1ß as an important target. Lipopolysaccharide also increases succinylation of several proteins. We therefore identify succinate as a metabolite in innate immune signalling, which enhances interleukin-1ß production during inflammation.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Interleucina-1beta/biossíntese , Transdução de Sinais , Ácido Succínico/metabolismo , Animais , Células da Medula Óssea/citologia , Ciclo do Ácido Cítrico/efeitos dos fármacos , Desoxiglucose/farmacologia , Regulação para Baixo/efeitos dos fármacos , Genes Mitocondriais/efeitos dos fármacos , Genes Mitocondriais/genética , Glutamina/metabolismo , Glicólise/efeitos dos fármacos , Glicólise/genética , Humanos , Imunidade Inata/efeitos dos fármacos , Inflamação/metabolismo , Interleucina-1beta/genética , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Regulação para Cima/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo
5.
Br J Dermatol ; 158(3): 442-55, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18275522

RESUMO

Microbes found on the skin are usually regarded as pathogens, potential pathogens or innocuous symbiotic organisms. Advances in microbiology and immunology are revising our understanding of the molecular mechanisms of microbial virulence and the specific events involved in the host-microbe interaction. Current data contradict some historical classifications of cutaneous microbiota and suggest that these organisms may protect the host, defining them not as simple symbiotic microbes but rather as mutualistic. This review will summarize current information on bacterial skin flora including Staphylococcus, Corynebacterium, Propionibacterium, Streptococcus and Pseudomonas. Specifically, the review will discuss our current understanding of the cutaneous microbiota as well as shifting paradigms in the interpretation of the roles microbes play in skin health and disease.


Assuntos
Metagenoma/imunologia , Viabilidade Microbiana/imunologia , Pele/microbiologia , Corynebacterium diphtheriae/imunologia , Feminino , Infecções por Bactérias Gram-Positivas/imunologia , Cocos Gram-Positivos/crescimento & desenvolvimento , Cocos Gram-Positivos/imunologia , Humanos , Masculino , Metagenoma/fisiologia , Propionibacterium acnes/imunologia
6.
J Immunol ; 167(12): 7069-76, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11739528

RESUMO

Group B streptococcus (GBS) imposes a major health threat to newborn infants. Little is known about the molecular basis of GBS-induced sepsis. Both heat-inactivated whole GBS bacteria and a heat-labile soluble factor released by GBS during growth (GBS-F) induce nuclear translocation of NF-kappaB, the secretion of TNF-alpha, and the formation of NO in mouse macrophages. Macrophages from mice with a targeted disruption of MyD88 failed to secrete TNF-alpha in response to both heat-inactivated whole bacteria and GBS-F, suggesting that Toll-like receptors (TLRs) are involved in different aspects of GBS recognition. Immune cell activation by whole bacteria differed profoundly from that by secreted GBS-F. Whole GBS activated macrophages independently of TLR2 and TLR6, whereas a response to the secreted GBS-F was not observed in macrophages from TLR2-deficient animals. In addition to TLR2, TLR6 and CD14 expression were essential for GBS-F responses, whereas TLR1 and TLR4 or MD-2 did not appear to be involved. Heat lability distinguished GBS-F from peptidoglycan and lipoproteins. GBS mutants deficient in capsular polysaccharide or beta-hemolysin had GBS-F activity comparable to that of wild-type streptococci. We suggest that CD14 and TLR2 and TLR6 function as coreceptors for secreted microbial products derived from GBS and that cell wall components of GBS are recognized by TLRs distinct from TLR1, 2, 4, or 6.


Assuntos
Proteínas de Drosophila , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/imunologia , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Interleucina-1 , Streptococcus agalactiae/fisiologia , Animais , Antígenos de Superfície/fisiologia , Fatores Biológicos/metabolismo , Células CHO , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Células Cultivadas , Cricetinae , Humanos , Mediadores da Inflamação/metabolismo , Antígeno 96 de Linfócito , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Knockout , Modelos Imunológicos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Sepse/imunologia , Infecções Estreptocócicas/imunologia , Receptor 1 Toll-Like , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Receptor 6 Toll-Like , Receptores Toll-Like , Transfecção , Fator de Necrose Tumoral alfa/biossíntese
7.
Nature ; 414(6862): 454-7, 2001 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-11719807

RESUMO

In mammals, several gene families encode peptides with antibacterial activity, such as the beta-defensins and cathelicidins. These peptides are expressed on epithelial surfaces and in neutrophils, and have been proposed to provide a first line of defence against infection by acting as 'natural antibiotics'. The protective effect of antimicrobial peptides is brought into question by observations that several of these peptides are easily inactivated and have diverse cellular effects that are distinct from antimicrobial activity demonstrated in vitro. To investigate the function of a specific antimicrobial peptide in a mouse model of cutaneous infection, we applied a combined mammalian and bacterial genetic approach to the cathelicidin antimicrobial gene family. The mature human (LL-37) and mouse (CRAMP) peptides are encoded by similar genes (CAMP and Cnlp, respectively), and have similar alpha-helical structures, spectra of antimicrobial activity and tissue distribution. Here we show that cathelicidins are an important native component of innate host defence in mice and provide protection against necrotic skin infection caused by Group A Streptococcus (GAS).


Assuntos
Peptídeos Catiônicos Antimicrobianos , Proteínas/imunologia , Dermatopatias Bacterianas/imunologia , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes , Animais , Catelicidinas , Farmacorresistência Bacteriana/genética , Feminino , Imunidade Inata , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Proteínas/genética , Proteínas/farmacologia , Proteínas Recombinantes de Fusão , Streptococcus pyogenes/genética
8.
J Infect ; 42(4): 272-7, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11545571

RESUMO

Necrotizing fasciitis (NF) is a life-threatening infection involving rapid necrosis of subcutaneous and fascial tissues. Streptococcus pneumoniae (SPN) soft tissue infection is exceedingly uncommon, reported primarily in patients with immunosuppression or other underlying conditions. We report a case of NF and septic shock in a healthy 32-year-old man, whose only predisposing factor was antecedent blunt trauma. Pathological examination and culture of the extensive tissue debridement were positive only for SPN. The serotype 9V isolate was penicillin (PCN)-resistant (MIC=2.0), and closely-related by pulse field gel electrophoresis and multilocus fingerprinting to clone France 9V-3, an important genetic reservoir for increasing PCN-resistance worldwide. This unique case has implications for our pathogenic under-standing and empiric management of NF.


Assuntos
Fasciite Necrosante/diagnóstico , Fasciite Necrosante/microbiologia , Resistência às Penicilinas , Streptococcus pneumoniae/efeitos dos fármacos , Adulto , Antibacterianos , Quimioterapia Combinada/uso terapêutico , Eletroforese em Gel de Campo Pulsado , Fasciite Necrosante/patologia , Fasciite Necrosante/terapia , Humanos , Imunocompetência , Masculino , Choque Séptico/microbiologia , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/isolamento & purificação , Ferimentos não Penetrantes/complicações
9.
J Invest Dermatol ; 117(1): 91-7, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11442754

RESUMO

Cathelicidins are a family of peptides thought to provide an innate defensive barrier against a variety of potential microbial pathogens. The human and mouse cathelicidins (LL-37 and CRAMP, respectively) are expressed at select epithelial interfaces where they have been proposed to kill a number of gram-negative and gram-positive bacteria. To determine if these peptides play a part in the protection of skin against wound infections, the anti-microbial activity of LL-37 and CRAMP was determined against the common wound pathogen group A Streptococcus, and their expression was examined after cutaneous injury. We observed a large increase in the expression of cathelicidins in human and murine skin after sterile incision, or in mouse following infection by group A Streptococcus. The appearance of cathelicidins in skin was due to both synthesis within epidermal keratinocytes and deposition from granulocyctes that migrate to the site of injury. Synthesis and deposition in the wound was accompanied by processing from the inactive prostorage form to the mature C-terminal peptide. Analysis of anti-microbial activity of this C-terminal peptide against group A Streptococcus revealed that both LL-37 and CRAMP potently inhibited bacterial growth. Action against group A Streptococcus occurred in conditions that typically abolish the activity of anti-microbial peptides against other organisms. Thus, cathelicidins are well suited to provide defense against infections due to group A Streptococcus, and represent an important element of cutaneous innate immunity.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas/metabolismo , Pele/lesões , Infecções Estreptocócicas/metabolismo , Streptococcus pyogenes , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Catelicidinas , Feminino , Expressão Gênica/fisiologia , Humanos , Queratinócitos/metabolismo , Queratinócitos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas/genética , RNA Mensageiro/análise , Pele/microbiologia , Cicatrização/fisiologia
10.
Infect Immun ; 69(4): 1994-2000, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11254550

RESUMO

Streptococcus iniae causes meningoencephalitis and death in commercial fish species and has recently been identified as an emerging human pathogen producing fulminant soft tissue infection. As identified by pulsed-field gel electrophoresis (PFGE), strains causing disease in either fish or humans belong to a single clone, whereas isolates from nondiseased fish are genetically diverse. In this study, we used in vivo and in vitro models to examine the pathogenicity of disease-associated isolates. Strains with the clonal (disease-associated) PFGE profile were found to cause significant weight loss and bacteremia in a mouse model of subcutaneous infection. As little as 10(2) CFU of a disease-associated strain was sufficient to establish bacteremia, with higher inocula (10(7)) resulting in increased mortality. In contrast, non-disease-associated (commensal) strains failed to cause bacteremia and weight loss, even at inocula of 10(8) CFU. In addition, disease-associated strains were more resistant to phagocytic clearance in a human whole blood killing assay compared to commensal strains, which were almost entirely eradicated. Disease-associated strains were also cytotoxic to human endothelial cells as measured by lactate dehydrogenase release from host cells. However, both disease-associated and commensal strains adhered to and invaded cultured human epithelial and endothelial cells equally well. While cellular invasion may still contribute to the pathogenesis of invasive S. iniae disease, resistance to phagocytic clearance and direct cytotoxicity appear to be discriminating virulence attributes of the disease-associated clone.


Assuntos
Streptococcus/patogenicidade , Animais , Aderência Bacteriana , Atividade Bactericida do Sangue , Linhagem Celular , Feminino , Camundongos , Camundongos Pelados , Streptococcus/genética , Virulência
11.
Mol Microbiol ; 39(2): 236-47, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11136446

RESUMO

Group B streptococci (GBS) express a beta-haemolysin/cytolysin that contributes to disease pathogenesis. We report an independent discovery and extension of a genetic locus encoding the GBS beta-haemolysin/cytolysin activity. A plasmid library of GBS chromosomal DNA was cloned into Escherichia coli, and a transformant was identified as beta-haemolytic on blood agar. The purified plasmid contained a 4046 bp insert of GBS DNA encoding two complete open reading frames (ORFs). A partial upstream ORF (cylB) and the first complete ORF (cylE) represent the 3' end of a newly reported genetic locus (cyl) required for GBS haemolysin/cytolysin activity. ORF cylE is predicted to encode a 78.3 kDa protein without GenBank homologies. The GBS DNA fragment also includes a previously unreported ORF, cylF, with homology to bacterial aminomethyltransferases, and the 5' end of cylH, with homology to 3-ketoacyl-ACP synthases. Southern analysis demonstrated that the cyl locus was conserved among GBS of all common serotypes. Targeted plasmid integrational mutagenesis was used to disrupt cylB, cylE, cylF and cylH in three wild-type GBS strains representing serotypes Ia, III and V. Targeted integrations in cylB, cylF and cylH retaining wild-type haemolytic activity were identified in all strains. In contrast, targeted integrations in cylE were invariably non-haemolytic and non-cytolytic, a finding confirmed by in frame allelic exchange of the cylE gene. The haemolytic/cytolytic activity of the cylE allelic exchange mutants could be restored by reintroduction of cylE on a plasmid vector. Inducible expression of cylE, cylF and cylEF demonstrated that it is CylE that confers haemolytic activity in E. coli. We conclude that cylE probably represents the structural gene for the GBS haemolysin/cytolysin, a novel bacterial toxin.


Assuntos
Citotoxinas/genética , Citotoxinas/metabolismo , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Streptococcus agalactiae/genética , Alelos , Proteínas de Bactérias , Células Cultivadas , Células Epiteliais/microbiologia , Escherichia coli/genética , Genes Bacterianos/genética , Teste de Complementação Genética , Humanos , Pulmão/citologia , Mutagênese , Óperon , Plasmídeos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/classificação , Streptococcus agalactiae/metabolismo , Transformação Bacteriana
12.
J Infect Dis ; 182(3): 824-32, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10950777

RESUMO

Septic arthritis is a clinical manifestation of group B streptococcal (GBS) infection in neonates and adults. To examine the potential role of GBS beta-hemolysin in joint injury, mice were infected with 2 wild-type strains or with nonhemolytic (NH) or hyperhemolytic (HH) variants derived by transposon mutagenesis. Compared with mice infected with the parent strains, mice infected with the NH mutants had decreased mortality and bacterial proliferation. A reduced LD(50) and a higher microbial load were obtained in mice infected with the HH mutants. Greater degrees of joint inflammation and damage were observed in the HH mutant-infected animals than in those infected with the parental strains. NH mutant-infected mice manifested only a mild and transient arthritis. Systemic and local levels of interleukin-6 mirrored the observed differences in virulence and severity of arthritis. These data support a direct correlation of GBS beta-hemolysin expression with mortality and severity of articular lesions.


Assuntos
Artrite Infecciosa/metabolismo , Proteínas Hemolisinas/biossíntese , Infecções Estreptocócicas/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Inflamação/microbiologia , Interleucina-6/metabolismo , Dose Letal Mediana , Masculino , Camundongos , Streptococcus/genética , Streptococcus/metabolismo , Streptococcus/patogenicidade
13.
J Infect Dis ; 182(1): 150-7, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10882592

RESUMO

Group B streptococcus (GBS) is the leading cause of sepsis in neonates. Nitric oxide (NO) release plays a role in the hypotension that characterizes septic shock. To examine the role of the GBS beta-hemolysin in NO production, the murine macrophage line RAW 264. 7 was exposed to a wild-type (WT) GBS isolate and to hyperhemolytic (HH) and nonhemolytic (NH) transposon mutants derived from that isolate. After activation of macrophages by the WT strain, the HH mutant, or cell-free extracts of beta-hemolysin, nitrite release into the supernatant increased >10-fold and inducible NO synthase (iNOS) levels in cell lysates increased up to 10-fold compared with treatment with the NH mutant or extracts from that mutant. Hemolysin-induced NO production was dependent on protein tyrosine kinases and NF-kappaB, but not on extracellular signal-related kinase-1/2-mitogen-activated kinases or protein kinase A. These results indicate that GBS beta-hemolysin induces murine macrophage iNOS via intracellular pathways similar to those that mediate lipopolysaccharide-induced iNOS activation.


Assuntos
Proteínas Hemolisinas/farmacologia , Macrófagos/efeitos dos fármacos , Óxido Nítrico/metabolismo , Animais , Proteínas de Bactérias , Células Cultivadas , Relação Dose-Resposta a Droga , Indução Enzimática , Hemólise , Interferon gama/farmacologia , Macrófagos/enzimologia , Macrófagos/metabolismo , Camundongos , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase Tipo II , Nitritos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
14.
Infect Immun ; 68(7): 4245-54, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10858242

RESUMO

Group A streptococcus (GAS) is an important human pathogen that causes pharyngitis and invasive infections, including necrotizing fasciitis. Streptolysin S (SLS) is the cytolytic factor that creates the zone of beta-hemolysis surrounding GAS colonies grown on blood agar. We recently reported the discovery of a potential genetic determinant involved in SLS production, sagA, encoding a small peptide of 53 amino acids (S. D. Betschel, S. M. Borgia, N. L. Barg, D. E. Low, and J. C. De Azavedo, Infect. Immun. 66:1671-1679, 1998). Using transposon mutagenesis, chromosomal walking steps, and data from the GAS genome sequencing project (www.genome.ou.edu/strep. html), we have now identified a contiguous nine-gene locus (sagA to sagI) involved in SLS production. The sag locus is conserved among GAS strains regardless of M protein type. Targeted plasmid integrational mutagenesis of each gene in the sag operon resulted in an SLS-negative phenotype. Targeted integrations (i) upstream of the sagA promoter and (ii) downstream of a terminator sequence after sagI did not affect SLS production, establishing the functional boundaries of the operon. A rho-independent terminator sequence between sagA and sagB appears to regulate the amount of sagA transcript produced versus transcript for the entire operon. Reintroduction of the nine-gene sag locus on a plasmid vector restored SLS activity to the nonhemolytic sagA knockout mutant. Finally, heterologous expression of the intact sag operon conferred the SLS beta-hemolytic phenotype to the nonhemolytic Lactococcus lactis. We conclude that gene products of the GAS sag operon are both necessary and sufficient for SLS production. Sequence homologies of sag operon gene products suggest that SLS is related to the bacteriocin family of microbial toxins.


Assuntos
Proteínas de Bactérias , Genes Bacterianos , Streptococcus pyogenes/genética , Streptococcus pyogenes/metabolismo , Estreptolisinas/biossíntese , Estreptolisinas/genética , Sequência de Aminoácidos , Sequência de Bases , Mapeamento Cromossômico , Passeio de Cromossomo , Sequência Conservada , Primers do DNA/genética , DNA Bacteriano/genética , Hemólise , Humanos , Dados de Sequência Molecular , Mutagênese Insercional , Óperon , Fenótipo , Homologia de Sequência de Aminoácidos , Streptococcus pyogenes/patogenicidade
15.
Infect Immun ; 68(2): 637-43, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10639427

RESUMO

Streptococcus suis serotype 2 is a worldwide causative agent of many forms of swine infection and is also recognized as a zoonotic agent causing human disease, including meningitis. The pathogenesis of S. suis infections is poorly understood. Bacteria circulate in the bloodstream in the nonimmune host until they come in contact with brain microvascular endothelial cells (BMEC) forming the blood-brain barrier. The bacterial polysaccharide capsule confers antiphagocytic properties. It is known that group B streptococci (GBS) invade and damage BMEC, which may be a primary step in the pathogenesis of neonatal meningitis. Interactions between S. suis and human endothelial cells were studied to determine if they differ from those between GBS and endothelial cells. Invasion assays performed with BMEC and human umbilical vein endothelial cells demonstrated that unlike GBS, S. suis serotype 2 could not invade either type of cell. Adherence assays showed that S. suis adhered only to BMEC, whereas GBS adhered to both types of cell. These interactions were not affected by the presence of a capsule, since acapsular mutants from both bacterial species adhered similarly compared to the wild-type strains. Lactate dehydrogenase release measurements indicated that some S. suis strains were highly cytotoxic for BMEC, even more than GBS, whereas others were not toxic at all. Cell damage was related to suilysin (S. suis hemolysin) production, since only suilysin-producing strains were cytotoxic and cytotoxicity could be inhibited by cholesterol and antisuilysin antibodies. It is possible that hemolysin-positive S. suis strains use adherence and suilysin-induced BMEC injury, as opposed to direct cellular invasion, to proceed from the circulation to the central nervous system.


Assuntos
Encéfalo/microbiologia , Endotélio Vascular/microbiologia , Streptococcus suis/patogenicidade , Aderência Bacteriana , Células Cultivadas , Endotélio Vascular/ultraestrutura , Proteínas Hemolisinas/toxicidade , Humanos , Microscopia Eletrônica , Compostos Orgânicos , Polissacarídeos Bacterianos/fisiologia , Sorotipagem , Virulência
16.
Pediatr Res ; 45(5 Pt 1): 626-34, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10231855

RESUMO

Group B streptococci (GBS) are the leading cause of pneumonia and sepsis in human newborns. Exudative pulmonary edema and alveolar hemorrhage seen in GBS pneumonia indicate vascular damage, and we reported that GBS injure lung microvascular endothelial cells (LMvEC) both in vivo and in vitro. The specific GBS factors causing LMvEC injury are uncertain, but GBS beta-hemolysin activity is associated with lung epithelial cell injury. We hypothesized that GBS beta-hemolysin contributes to LMvEC injury and exudative pulmonary edema. To test this hypothesis we used isogenic nonhemolytic and hyperhemolytic GBS mutants derived by transposon insertional mutagenesis from three different wild-type strains. Hemolytic titers for each strain were calculated using live GBS and Tween 80/starch-stabilized extracts of log-phase GBS. All nonhemolytic mutants lacked detectable hemolytic activity, whereas hyperhemolytic mutants produced 4-16 times the hemolytic activity of their parent strains. LMvEC injury was assayed by light microscopy, the release of lactate dehydrogenase, trypan blue nuclear staining and Evans blue-albumin flux. Compared with the parent strains, all nonhemolytic mutants caused significantly reduced, and all hyperhemolytic mutants caused significantly greater lactate dehydrogenase release from and trypan blue nuclear staining of LMvEC. Moreover, a nonhemolytic mutant caused reduced and a hyperhemolytic mutant caused increased Evans-blue albumin flux across polar LMvEC monolayers. These findings were corroborated by light microscopic evidence of hemolysin-associated damage to the LMvEC monolayers. We conclude that GBS beta-hemolysin promotes LMvEC injury and increases permeability in vitro, and speculate that GBS beta-hemolysin contributes to the pathogenesis of alveolar edema and hemorrhage in early onset GBS pneumonia.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Proteínas Hemolisinas/toxicidade , Microcirculação/efeitos dos fármacos , Circulação Pulmonar , Streptococcus agalactiae , Animais , Proteínas de Bactérias , Morte Celular/efeitos dos fármacos , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/patologia , Proteínas Hemolisinas/genética , Hemólise , Humanos , Recém-Nascido , L-Lactato Desidrogenase , Mutagênese Insercional , Proteínas Recombinantes/toxicidade , Soroalbumina Bovina/farmacocinética , Streptococcus agalactiae/genética , Streptococcus agalactiae/isolamento & purificação , Suínos
17.
Infect Immun ; 65(12): 5074-81, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9393798

RESUMO

Group B streptococci (GBS) are the leading cause of meningitis in newborns. Although meningitis develops following bacteremia, the precise mechanism or mechanisms whereby GBS leave the bloodstream and gain access to the central nervous system (CNS) are not known. We hypothesized that GBS produce meningitis because of a unique capacity to invade human brain microvascular endothelial cells (BMEC), the single-cell layer which constitutes the blood-brain barrier. In order to test this hypothesis, we developed an in vitro model with BMEC isolated from a human, immortalized by simian virus 40 transformation, and propagated in tissue culture monolayers. GBS invasion of BMEC monolayers was demonstrated by electron microscopy. Intracellular GBS were found within membrane-bound vacuoles, suggesting the organism induced its own endocytic uptake. GBS invasion of BMEC was quantified with a gentamicin protection assay. Serotype III strains, which account for the majority of CNS isolates, invaded BMEC more efficiently than strains from other common GBS serotypes. GBS survived within BMEC for up to 20 h without significant intracellular replication. GBS invasion of BMEC required active bacterial DNA, RNA, and protein synthesis, as well as microfilament and microtubule elements of the eukaryotic cytoskeleton. The polysaccharide capsule of GBS attenuated the invasive ability of the organism. At high bacterial densities, GBS invasion of BMEC was accompanied by evidence of cellular injury; this cytotoxicity was correlated to beta-hemolysin production by the bacterium. Finally, GBS demonstrated transcytosis across intact, polar BMEC monolayers grown on Transwell membranes. GBS invasion of BMEC may be a primary step in the pathogenesis of meningitis, allowing bacteria access to the CNS by transcytosis or by injury and disruption of the endothelial blood-brain barrier.


Assuntos
Endotélio Vascular/microbiologia , Microcirculação/microbiologia , Streptococcus/fisiologia , Encéfalo/irrigação sanguínea , Células Cultivadas , Endotélio Vascular/ultraestrutura , Humanos , Microscopia Eletrônica , Streptococcus/patogenicidade
19.
Adv Exp Med Biol ; 418: 627-30, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9331730

RESUMO

There is a direct correlation between the level of GBS beta-hemolysin expression and the ability of GBS to injury lung epithelial cells. Electron microscopy suggest the hemolysin acts as a pore-forming cytolysin. beta-hemolysin-associated lung epithelial cell injury is inhibited by surfactant phospholipid, a substance in which high-risk premature infants are deficient. We have now shown that loss of GBS hemolysin activity is associated with decreased animal virulence following intrathoracic inoculation of the organism. Further, a knockout of a putative GBS beta-hemolysin gene from the literature suggests it is not the major GBS hemolysin determinant. Cloning and sequencing analysis of the Tn916 (or Tn916DE) insertions in three of our nonhemolytic GBS mutants show identical integration sites in a distinct chromosomal locus. Finally, a putative 11-kd hemolysin species is identified by comparative analysis of protein extracts from isogenic hemolysin mutants.


Assuntos
Proteínas Hemolisinas/toxicidade , Lesão Pulmonar , Streptococcus agalactiae/patogenicidade , Animais , Animais Recém-Nascidos , Proteínas de Bactérias , Modelos Animais de Doenças , Genes Bacterianos , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/isolamento & purificação , Humanos , Técnicas In Vitro , Recém-Nascido , Mutação , Pneumonia Bacteriana/etiologia , Ratos , Ratos Sprague-Dawley , Infecções Estreptocócicas/etiologia , Streptococcus agalactiae/genética , Virulência/genética
20.
Infect Immun ; 64(9): 3818-26, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8751934

RESUMO

Group B streptococci (GBS) are the leading cause of serious bacterial infection in newborns. Early-onset disease is heralded by pneumonia and lung injury, and the lung may serve as a portal of entry for GBS into the bloodstream. To examine a potential role for GBS beta-hemolysin in lung epithelial injury, five wild-type strains varying in beta-hemolysin expression were chosen, along with five nonhemolytic (NH) and five hyperhemolytic (HH) variants of these strains derived by chemical or transposon mutagenesis. Monolayers of A549 alveolar epithelial cells were exposed to log-phase GBS or stabilized hemolysin extracts of GBS cultures, and cellular injury was assessed by lactate dehydrogenase (LDH) release and trypan blue nuclear staining. Whereas NH strains produced no detectable injury beyond baseline (medium alone), hemolysin-producing strains induced LDH release from A549 cells in direct correlation to their ability to lyse sheep erythrocytes. HH strains were also associated with marked increases in trypan blue nuclear staining of A549 monolayers. The extent of LDH release produced by HH strains was significantly reduced in the presence of dipalmitoyl phosphatidylcholine, a known inhibitor of hemolysin and the major phospholipid component of human surfactant. Electron microscopic studies of A549 cell monolayers exposed to HH GBS mutants revealed global loss of microvillus architecture, disruption of cytoplasmic and nuclear membranes, and marked swelling of the cytoplasm and organelles. We conclude that GBS hemolysin expression correlates with lung epithelial cell injury and may be important in the initial pathogenesis of early-onset disease, particularly when pulmonary surfactant is deficient.


Assuntos
Proteínas Hemolisinas/metabolismo , Alvéolos Pulmonares/microbiologia , Streptococcus agalactiae/patogenicidade , 1,2-Dipalmitoilfosfatidilcolina/metabolismo , Proteínas de Bactérias , Ligação Competitiva , Células Cultivadas , Elementos de DNA Transponíveis , DNA Bacteriano/genética , Epitélio/patologia , Humanos , L-Lactato Desidrogenase/metabolismo , Microscopia Eletrônica de Varredura , Mutagênese Insercional , Fenótipo , Pigmentos Biológicos , Polimorfismo de Fragmento de Restrição , Alvéolos Pulmonares/patologia , Surfactantes Pulmonares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...