Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Cell Int ; 19: 346, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31889898

RESUMO

BACKGROUND: Treatments that generate T cell-mediated immunity to a patient's unique neoantigens are the current holy grail of cancer immunotherapy. In particular, treatments that do not require cumbersome and individualized ex vivo processing or manufacturing processes are especially sought after. Here we report that AGI-134, a glycolipid-like small molecule, can be used for coating tumor cells with the xenoantigen Galα1-3Galß1-4GlcNAc (α-Gal) in situ leading to opsonization with pre-existing natural anti-α-Gal antibodies (in short anti-Gal), which triggers immune cascades resulting in T cell mediated anti-tumor immunity. METHODS: Various immunological effects of coating tumor cells with α-Gal via AGI-134 in vitro were measured by flow cytometry: (1) opsonization with anti-Gal and complement, (2) antibody-dependent cell-mediated cytotoxicity (ADCC) by NK cells, and (3) phagocytosis and antigen cross-presentation by antigen presenting cells (APCs). A viability kit was used to test AGI-134 mediated complement dependent cytotoxicity (CDC) in cancer cells. The anti-tumoral activity of AGI-134 alone or in combination with an anti-programmed death-1 (anti-PD-1) antibody was tested in melanoma models in anti-Gal expressing galactosyltransferase knockout (α1,3GT-/-) mice. CDC and phagocytosis data were analyzed by one-way ANOVA, ADCC results by paired t-test, distal tumor growth by Mantel-Cox test, C5a data by Mann-Whitney test, and single tumor regression by repeated measures analysis. RESULTS: In vitro, α-Gal labelling of tumor cells via AGI-134 incorporation into the cell membrane leads to anti-Gal binding and complement activation. Through the effects of complement and ADCC, tumor cells are lysed and tumor antigen uptake by APCs increased. Antigen associated with lysed cells is cross-presented by CD8α+ dendritic cells leading to activation of antigen-specific CD8+ T cells. In B16-F10 or JB/RH melanoma models in α1,3GT-/- mice, intratumoral AGI-134 administration leads to primary tumor regression and has a robust abscopal effect, i.e., it protects from the development of distal, uninjected lesions. Combinations of AGI-134 and anti-PD-1 antibody shows a synergistic benefit in protection from secondary tumor growth. CONCLUSIONS: We have identified AGI-134 as an immunotherapeutic drug candidate, which could be an excellent combination partner for anti-PD-1 therapy, by facilitating tumor antigen processing and increasing the repertoire of tumor-specific T cells prior to anti-PD-1 treatment.

2.
Wound Repair Regen ; 26(6): 403-412, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30264418

RESUMO

The cholinergic anti-inflammatory pathway can directly affect skin antibacterial responses via nicotinic acetylcholine receptors (nAChRs). In particular, α7 nAChR (CHRNA7) present in the epidermis modulates the host response to wounding and/or wound bacterial infection. While physiologic inflammation is required to initiate normal wound repair and can be triggered by Toll-like receptor (TLR) activation, chronic inflammation is frequently observed in diabetic wounds and can occur, in part, via excessive TLR2 activation or production. Consequently, this can delay physiologic wound healing responses and increase diabetic host susceptibility to bacterial infection. In this study, we demonstrate that topical nAChR activation diminishes bacterial survival and systemic dissemination in a mouse model of diabetic wound infection, while reducing wound TLR2 production, relative to control mice. We further determined that the antimicrobial peptide activity of diabetic mouse wounds is increased compared to control mice, but this effect is lost following topical nAChR activation. Finally, we observed that human diabetic wounds exhibit impaired α7 nAChR (CHRNA7) abundance and localization relative to human control (nondiabetic) skin. These findings suggest that topical administration of nAChR agonists may improve wound healing and infection outcomes in diabetic wounds by dampening TLR2-mediated inflammation and antimicrobial peptide response, and that the diabetic microenvironment may promote aberrant CHRNA7 production/activation that likely contributes to diabetic wound pathogenesis.


Assuntos
Infecções Bacterianas/tratamento farmacológico , Agonistas Nicotínicos/farmacologia , Receptores Nicotínicos/metabolismo , Receptor 2 Toll-Like/efeitos dos fármacos , Cicatrização/fisiologia , Infecção dos Ferimentos/tratamento farmacológico , Animais , Infecções Bacterianas/patologia , Modelos Animais de Doenças , Mediadores da Inflamação , Camundongos , Camundongos Endogâmicos NOD , Agonistas Nicotínicos/administração & dosagem , Infecção dos Ferimentos/patologia
4.
PLoS One ; 11(5): e0154616, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27171010

RESUMO

A proof-of-concept study evaluating the potential of Streptococcus pneumoniae Pneumococcal Surface Protein A (PspA) as a passive immunization target was conducted. We describe the generation and isolation of several broadly reactive mouse anti-PspA monoclonal antibodies (mAbs). MAb 140H1 displayed (i) 98% strain coverage, (ii) activity in complement deposition and opsonophagocytic killing (OPK) assays, which are thought to predict the in vivo efficacy of anti-pneumococcal mAbs, (iii) efficacy in mouse sepsis models both alone and in combination with standard-of-care antibiotics, and (iv) therapeutic activity in a mouse pneumonia model. Moreover, we demonstrate that antibody engineering can significantly enhance anti-PspA mAb effector function. We believe that PspA has promising potential as a target for the therapy of invasive pneumococcal disease by mAbs, which could be used alone or in conjunction with standard-of-care antibiotics.


Assuntos
Anticorpos Monoclonais/imunologia , Proteínas de Bactérias/imunologia , Streptococcus pneumoniae/imunologia , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Complemento C3/metabolismo , Modelos Animais de Doenças , Mapeamento de Epitopos , Feminino , Humanos , Imunoglobulina G/sangue , Pneumopatias/imunologia , Pneumopatias/microbiologia , Camundongos Endogâmicos BALB C , Proteínas Opsonizantes/metabolismo , Fagócitos/metabolismo , Fagocitose , Infecções Pneumocócicas/tratamento farmacológico , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/microbiologia , Ligação Proteica , Sepse/tratamento farmacológico , Sepse/imunologia , Sepse/microbiologia , Resultado do Tratamento
5.
PLoS One ; 10(8): e0136605, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26305782

RESUMO

BlaI is a repressor of BlaZ, the beta-lactamase responsible for penicillin resistance in Staphylococcus aureus. Through screening a transposon library in S. aureus Newman for susceptibility to cathelicidin antimicrobial peptide, we discovered BlaI as a novel cathelicidin resistance factor. Additionally, through integrational mutagenesis in S. aureus Newman and MRSA Sanger 252 strains, we confirmed the role of BlaI in resistance to human and murine cathelidicin and showed that it contributes to virulence in human whole blood and murine infection models. We further demonstrated that BlaI could be a target for innate immune-based antimicrobial therapies; by removing BlaI through subinhibitory concentrations of 6-aminopenicillanic acid, we were able to sensitize S. aureus to LL-37 killing.


Assuntos
Peptídeos Catiônicos Antimicrobianos/administração & dosagem , Infecções Estafilocócicas/genética , Staphylococcus aureus/genética , beta-Lactamases/genética , Animais , Elementos de DNA Transponíveis/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Ácido Penicilânico/administração & dosagem , Ácido Penicilânico/análogos & derivados , Resistência às Penicilinas/genética , Infecções Estafilocócicas/sangue , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade , Catelicidinas
6.
J Mol Med (Berl) ; 93(6): 619-31, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25940316

RESUMO

UNLABELLED: The ever-increasing threat of multi-drug resistant bacterial infections has spurred renewed interest in alternative approaches to classical antibiotic therapy. In contrast to other mammals, humans do not express the galactose-α-1,3-galactosyl-ß-1,4-N-acetyl-glucosamine (α-Gal) epitope. As a result of exposure of humans to α-Gal in the environment, a large proportion of circulating antibodies are specific for the trisaccharide. In this study, we examine whether these anti-Gal antibodies can be recruited and redirected to exert anti-bacterial activity. We show that a specific DNA aptamer conjugated to an α-Gal epitope at its 5' end, herein termed an alphamer, can bind to group A Streptococcus (GAS) bacteria by recognition of a conserved region of the surface-anchored M protein. The anti-GAS alphamer was shown to recruit anti-Gal antibodies to the streptococcal surface in an α-Gal-specific manner, elicit uptake and killing of the bacteria by human phagocytes, and slow growth of invasive GAS in human whole blood. These studies provide a first in vitro proof of concept that alphamers have the potential to redirect pre-existing antibodies to bacteria in a specific manner and trigger an immediate antibacterial immune response. Further validation of this novel therapeutic approach of applying α-Gal technology in in vivo models of bacterial infection is warranted. KEY MESSAGES: . α-Gal-tagged aptamers lead to GAS opsonization with anti-Gal antibodies. . α-Gal-tagged aptamers confer phagocytosis and killing of GAS cells by human phagocytes. . α-Gal-tagged aptamers reduces replication of GAS in human blood. . α-Gal-tagged aptamers may have the potential to be used as novel passive immunization drugs.


Assuntos
Acetilglucosamina/farmacologia , Antibacterianos/farmacologia , Anticorpos Antibacterianos/imunologia , Aptâmeros de Nucleotídeos/farmacologia , Infecções Estreptocócicas/tratamento farmacológico , Streptococcus/efeitos dos fármacos , Acetilglucosamina/química , Acetilglucosamina/imunologia , Animais , Antibacterianos/química , Antibacterianos/imunologia , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/imunologia , Sequência de Bases , Descoberta de Drogas , Epitopos/química , Epitopos/imunologia , Epitopos/farmacologia , Humanos , Dados de Sequência Molecular , Fagocitose/efeitos dos fármacos , Infecções Estreptocócicas/imunologia , Streptococcus/imunologia
7.
J Leukoc Biol ; 86(5): 1159-69, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19638500

RESUMO

Neutrophils kill invading pathogens by AMPs, including cathelicidins, ROS, and NETs. The human pathogen Staphylococcus aureus exhibits enhanced resistance to neutrophil AMPs, including the murine cathelicidin CRAMP, in part, as a result of alanylation of teichoic acids by the dlt operon. In this study, we took advantage of the hypersusceptible phenotype of S. aureus DeltadltA against cationic AMPs to study the impact of the murine cathelicidin CRAMP on staphylococcal killing and to identify its key site of action in murine neutrophils. We demonstrate that CRAMP remained intracellular during PMN exudation from blood and was secreted upon PMA stimulation. We show first evidence that CRAMP was recruited to phagolysosomes in infected neutrophils and exhibited intracellular activity against S. aureus. Later in infection, neutrophils produced NETs, and immunofluorescence revealed association of CRAMP with S. aureus in NETs, which similarly killed S. aureus wt and DeltadltA, indicating that CRAMP activity was reduced when associated with NETs. Indeed, the presence of DNA reduced the antimicrobial activity of CRAMP, and CRAMP localization in response to S. aureus was independent of the NADPH oxidase, whereas killing was partially dependent on a functional NADPH oxidase. Our study indicates that neutrophils use CRAMP in a timed and locally coordinated manner in defense against S. aureus.


Assuntos
Peptídeos Catiônicos Antimicrobianos/fisiologia , Infecções Estafilocócicas/sangue , Animais , Peptídeos Catiônicos Antimicrobianos/deficiência , Sobrevivência Celular , Citometria de Fluxo , Fluoresceína-5-Isotiocianato , Humanos , Lipopolissacarídeos/fisiologia , Lisossomos/microbiologia , Lisossomos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/microbiologia , Neutrófilos/patologia , Fagocitose , Infecções Estafilocócicas/patologia , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/crescimento & desenvolvimento , Catelicidinas
8.
J Bacteriol ; 191(7): 2023-32, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19114476

RESUMO

Group B Streptococcus (GBS) is major cause of invasive disease in newborn infants and the leading cause of neonatal meningitis. To gain access to the central nervous system (CNS), GBS must not only subvert host defenses in the bloodstream but also invade and survive within brain microvascular endothelial cells (BMEC), the principal cell layer composing the blood-brain barrier (BBB). While several GBS determinants that contribute to the invasion of BMEC have been identified, little is known about the GBS factors that are required for intracellular survival and ultimate disease progression. In this study we sought to identify these factors by screening a random GBS mutant library in an in vitro survival assay. One mutant was identified which contained a disruption in a two-component regulatory system homologous to CiaR/CiaH, which is present in other streptococcal pathogens. Deletion of the putative response regulator, ciaR, in GBS resulted in a significant decrease in intracellular survival within neutrophils, murine macrophages, and human BMEC, which was linked to increased susceptibility to killing by antimicrobial peptides, lysozyme, and reactive oxygen species. Furthermore, competition experiments with mice showed that wild-type GBS had a significant survival advantage over the GBS DeltaciaR mutant in the bloodstream and brain. Microarray analysis comparing gene expression between wild-type and DeltaciaR mutant GBS bacteria revealed several CiaR-regulated genes that may contribute to stress tolerance and the subversion of host defenses by GBS. Our results identify the GBS CiaR response regulator as a crucial factor in GBS intracellular survival and invasive disease pathogenesis.


Assuntos
Proteínas de Bactérias/imunologia , Imunidade Inata , Proteínas Quinases/imunologia , Infecções Estreptocócicas/imunologia , Streptococcus agalactiae/fisiologia , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Linhagem Celular , Células Cultivadas , Células Endoteliais/microbiologia , Regulação Bacteriana da Expressão Gênica , Humanos , Macrófagos/microbiologia , Masculino , Camundongos , Dados de Sequência Molecular , Neutrófilos/microbiologia , Proteínas Quinases/química , Proteínas Quinases/genética , Alinhamento de Sequência , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/genética , Streptococcus agalactiae/imunologia , Streptococcus agalactiae/patogenicidade , Virulência
9.
BMC Microbiol ; 8: 85, 2008 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-18518949

RESUMO

BACKGROUND: Modification of teichoic acids with D-alanine by the products of the dlt operon protects Gram-positive bacteria against major antimicrobial host defense molecules such as defensins, cathelicidins, myeloperoxidase or phospholipase. The graRS regulatory genes have recently been implicated in the control of D-alanylation in Staphylococcus aureus. RESULTS: To determine the impact of the GraRS regulatory system on resistance to antimicrobial host defense mechanisms and virulence of S. aureus, we compared inactivation of S. aureus SA113 wild type and its isogenic graRS deletion mutant by the human cathelicidin LL-37 or human neutrophil granulocytes in vitro, and the ability to cause infection in vivo. We show here that graRS deletion considerably alters bacterial surface charge, increases susceptibility to killing by human neutrophils or the defense peptide LL-37, and attenuates virulence of S. aureus in a mouse infection model. CONCLUSION: Our results indicate that S. aureus can regulate its surface properties in order to overcome innate host defenses.


Assuntos
Genes Reguladores , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidade , Alanina/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/imunologia , Catelicidinas , Membrana Celular/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Feminino , Deleção de Genes , Genes Bacterianos , Humanos , Imunidade Inata , Rim/microbiologia , Lisina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/imunologia , Óperon , Fosfatidilgliceróis/metabolismo , Ácidos Teicoicos/metabolismo , Virulência
10.
J Infect Dis ; 197(7): 1028-35, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18419540

RESUMO

BACKGROUND: Biofilm formation is considered to be an important virulence factor of the opportunistic pathogen Staphylococcus epidermidis. We hypothesized that biofilm formation could interfere with the deposition of immunoglobulins and complement on the bacterial surface, leading to diminished activation of the complement system and protection from killing by human phagocytes. METHODS: The killing of biofilm-encased and planktonically grown wild-type (wt) S. epidermidis and the killing of an isogenic biofilm-negative ica mutant (ica(-)) by human polymorphonuclear neutrophils (PMNs) were compared. C3a induction and deposition of C3b and immunoglobulin G (IgG) on the bacteria after opsonization with human serum were assessed by enzyme-linked immunosorbent assay, flow cytometry, and electron microscopy. The virulence of the bacterial strains was compared in a mouse model of catheter-associated infection. RESULTS: Biofilm-embedded wt S. epidermidis was killed less well by human PMNs and induced more C3a than planktonically grown wt and ica(-) S. epidermidis. However, the deposition of C3b and IgG on the bacterial surface was diminished in biofilm-encased staphylococci. wt S. epidermidis was more virulent in implant-associated infections and was killed more slowly than ica(-) in ex vivo assays of killing by PMNs. CONCLUSIONS: The results indicate that prevention of C3b and IgG deposition on the bacterial surface contributes to the biofilm-mediated protection of S. epidermidis from killing by PMNs.


Assuntos
Biofilmes/crescimento & desenvolvimento , Complemento C3/imunologia , Imunoglobulina G/imunologia , Viabilidade Microbiana , Neutrófilos/imunologia , Staphylococcus epidermidis/imunologia , Staphylococcus epidermidis/patogenicidade , Animais , Cateteres de Demora/microbiologia , Contagem de Colônia Microbiana , Complemento C3/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imunoglobulina G/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Proteínas Opsonizantes/metabolismo , Infecções Estafilocócicas/microbiologia
11.
FASEB J ; 21(4): 1107-16, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17215486

RESUMO

Antibiotics are designed to support host defense in controlling infection. Here we describe a paradoxical inhibitory effect of bacteriostatic antibiotics on key mediators of mammalian innate immunity. When growth of species including Escherichia coli and Staphylococcus aureus is suppressed by chloramphenicol or erythromycin, the susceptibility of the bacteria to cathelicidin antimicrobial peptides or serum complement was markedly diminished. Survival of the bacteria in human whole blood, human wound fluid, or a mouse wound infection model was in turn increased after antibiotic-induced bacteriostasis. These findings provide a further rationale against the indiscriminate use of antibiotics.


Assuntos
Anti-Infecciosos/farmacologia , Farmacorresistência Bacteriana , Escherichia coli/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/farmacologia , Catelicidinas , Cloranfenicol/farmacologia , Proteínas Inativadoras do Complemento/farmacologia , Eritromicina/farmacologia , Feminino , Humanos , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C
12.
Mol Microbiol ; 62(1): 15-25, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16942605

RESUMO

Serum opacity factor (SOF) is a bifunctional cell surface protein expressed by 40-50% of group A streptococcal (GAS) strains comprised of a C-terminal domain that binds fibronectin and an N-terminal domain that mediates opacification of mammalian sera. The sof gene was recently discovered to be cotranscribed in a two-gene operon with a gene encoding another fibronectin-binding protein, sfbX. We compared the ability of a SOF(+) wild-type serotype M49 GAS strain and isogenic mutants lacking SOF or SfbX to invade cultured HEp-2 human pharyngeal epithelial cells. Elimination of SOF led to a significant decrease in HEp-2 intracellular invasion while loss of SfbX had minimal effect. The hypoinvasive phenotype of the SOF(-) mutant could be restored upon complementation with the sof gene on a plasmid vector, and heterologous expression of sof49 in M1 GAS or Lactococcus lactis conferred marked increases in HEp-2 cell invasion. Studies using a mutant sof49 gene lacking the fibronectin-binding domain indicated that the N-terminal opacification domain of SOF contributes to HEp-2 invasion independent of the C-terminal fibronectin binding domain, findings corroborated by observations that a purified SOF N-terminal peptide could promote latex bead adherence to HEp-2 cells and inhibit GAS invasion of HEp-2 cells in a dose-dependent manner. Finally, the first in vivo studies to employ a single gene allelic replacement mutant of SOF demonstrate that this protein contributes to GAS virulence in a murine model of necrotizing skin infection.


Assuntos
Células Epiteliais/metabolismo , Peptídeo Hidrolases/metabolismo , Streptococcus pyogenes/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Células Epiteliais/citologia , Células Epiteliais/microbiologia , Fibronectinas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Mutação/genética , Peptídeo Hidrolases/genética , Ligação Proteica , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidade , Virulência/genética
13.
Curr Biol ; 16(4): 396-400, 2006 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-16488874

RESUMO

The innate immune response plays a crucial role in satisfactory host resolution of bacterial infection. In response to chemotactic signals, neutrophils are early responding cells that migrate in large numbers to sites of infection. The recent discovery of secreted neutrophil extracellular traps (NETs) composed of DNA and histones opened a novel dimension in our understanding of the microbial killing capacity of these specialized leukocytes. M1 serotype strains of the pathogen Group A Streptococcus (GAS) are associated with invasive infections including necrotizing fasciitis (NF) and express a potent DNase (Sda1). Here we apply a molecular genetic approach of allelic replacement mutagenesis, single gene complementation, and heterologous expression to demonstrate that DNase Sda1 is both necessary and sufficient to promote GAS neutrophil resistance and virulence in a murine model of NF. Live fluorescent microscopic cell imaging and histopathological analysis are used to establish for the first time a direct linkage between NET degradation and bacterial pathogenicity. Inhibition of GAS DNase activity with G-actin enhanced neutrophil clearance of the pathogen in vitro and reduced virulence in vivo. The results demonstrate a significant role for NETs in neutrophil-mediated innate immunity, and at the same time identify a novel therapeutic target against invasive GAS infection.


Assuntos
Desoxirribonucleases/metabolismo , Neutrófilos/fisiologia , Streptococcus pyogenes/patogenicidade , Actinas/farmacologia , Animais , Atividade Bactericida do Sangue , Bovinos , Desoxirribonucleases/antagonistas & inibidores , Modelos Animais de Doenças , Fasciite Necrosante/imunologia , Fasciite Necrosante/microbiologia , Teste de Complementação Genética , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Ativação de Neutrófilo , Streptococcus pyogenes/enzimologia , Transformação Bacteriana
14.
Cell Microbiol ; 8(2): 207-17, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16441432

RESUMO

The chemotactic migration of phagocytes to sites of infection, guided by gradients of microbial molecules, plays a key role in the first line of host defence. Bacteria are distinguished from eukaryotes by initiation of protein synthesis with formyl methionine. Synthetic formylated peptides (FPs) have been shown to be chemotactic for phagocytes, leading to the concept of FPs as pathogen-associated molecular patterns (PAMPs). However, it remains unclear whether FPs are major chemoattractants released by bacteria and whether further chemoattractants are produced. A Staphylococcus aureus mutant whose formyltransferase gene was inactivated (Deltafmt) produced no FPs and the in vitro and in vivo ability of Deltafmt culture supernatants to recruit neutrophils was considerably reduced compared with those of the parental strain. However, some chemotactic activity was retained, indicating that bacteria produce also unknown, non-FP chemoattractants. The activity of these novel PAMPs was sensitive to pertussis toxin but insensitive to the formyl peptide receptor inhibitor CHIPS. Deltafmt culture supernatants caused reduced calcium ion fluxes and reduced CD11b upregulation in neutrophils compared with wild-type supernatants. These data demonstrate an important role of FPs in innate immunity against bacterial infections and indicate that host chemotaxis receptors recognize a larger set of bacterial molecules than previously thought.


Assuntos
Fatores Quimiotáticos/fisiologia , Quimiotaxia de Leucócito , Neutrófilos/fisiologia , Peptídeos/fisiologia , Staphylococcus aureus/fisiologia , Animais , Antígeno CD11b/biossíntese , Cálcio/metabolismo , Feminino , Humanos , Hidroximetil e Formil Transferases/genética , Hidroximetil e Formil Transferases/metabolismo , Imunidade Inata , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Mutação , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Receptores de Formil Peptídeo/antagonistas & inibidores , Receptores de Formil Peptídeo/metabolismo , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/genética
15.
J Bacteriol ; 187(19): 6719-25, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16166534

RESUMO

Group A streptococcus (GAS) is a leading cause of severe, invasive human infections, including necrotizing fasciitis and toxic shock syndrome. An important element of the mammalian innate defense system against invasive bacterial infections such as GAS is the production of antimicrobial peptides (AMPs) such as cathelicidins. In this study, we identify a specific GAS phenotype that confers resistance to host AMPs. Allelic replacement of the dltA gene encoding d-alanine-d-alanyl carrier protein ligase in an invasive serotype M1 GAS isolate led to loss of teichoic acid d-alanylation and an increase in net negative charge on the bacterial surface. Compared to the wild-type (WT) parent strain, the GAS DeltadltA mutant exhibited increased susceptibility to AMP and lysozyme killing and to acidic pH. While phagocytic uptake of WT and DeltadltA mutants by human neutrophils was equivalent, neutrophil-mediated killing of the DeltadltA strain was greatly accelerated. Furthermore, we observed the DeltadltA mutant to be diminished in its ability to adhere to and invade cultured human pharyngeal epithelial cells, a likely proximal step in the pathogenesis of invasive infection. Thus, teichoic acid d-alanylation may contribute in multiple ways to the propensity of invasive GAS to bypass mucosal defenses and produce systemic infection.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Neutrófilos/microbiologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/imunologia , Streptococcus pyogenes/metabolismo , Ácidos Teicoicos/metabolismo , Alanina/metabolismo , Aderência Bacteriana/imunologia , Sobrevivência Celular/imunologia , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/microbiologia , Humanos , Concentração de Íons de Hidrogênio , Muramidase/metabolismo , Mutagênese , Neutrófilos/citologia , Óperon/fisiologia , Fagocitose/imunologia , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidade , Virulência , Fatores de Virulência/genética
16.
J Infect Dis ; 191(10): 1771-7, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15838806

RESUMO

Wall teichoic acids (WTAs) are major surface components of gram-positive bacteria that have recently been shown to play a key role in nasal colonization by Staphylococcus aureus. In the present study, we assessed the impact that WTAs have on endovascular infections by using a WTA-deficient S. aureus mutant ( Delta tagO). There were no significant differences detected between the isogenic parental strain (SA113) and the Delta tagO mutant in polymorphonuclear leukocyte-mediated opsonophagocytosis; killing by a prototypic platelet microbicidal protein; or binding to platelets, fibronectin, or fibrinogen. However, compared with the parental strain, the Delta tagO mutant adhered considerably less well to human endothelial cells, especially under flow conditions (70.3% reduction; P<.05). Beads coated with WTA bound to endothelium in a dose-dependent manner, suggesting that WTA contributes specifically to this interaction. These in vitro data closely paralleled those from a rabbit model of infective endocarditis in which the Delta tagO mutant was compared with the parental strain. Clearances of staphylococcus from the bloodstream were equivalent, but the Delta tagO mutant showed a significantly reduced capacity to both colonize sterile cardiac vegetations (P<.05) and proliferate within these vegetations, the kidneys, and the spleen (P<.001). We conclude that WTA is an important factor in the induction and progression of endovascular S. aureus infection, likely through a specific interaction with endothelial cells.


Assuntos
Endocardite Bacteriana/microbiologia , Células Endoteliais/fisiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade , Ácidos Teicoicos/metabolismo , Animais , Adesão Celular , Parede Celular/metabolismo , Humanos , Mutação , Neutrófilos , Fagocitose , Coelhos , Staphylococcus aureus/genética , Ácidos Teicoicos/genética , Virulência
17.
Microb Pathog ; 36(5): 237-45, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15043859

RESUMO

The virulence of Staphylococcus aureus Sa113 (SA113) and an isogenic ica deletion mutant (ica-), deficient in the production of polysaccharide intercellular adhesin (PIA), which is crucial for biofilm formation, was compared in a mouse tissue cage infection model. The minimal infective doses for the induction of persistent tissue infections in C57BL/6 mice were 10(3) CFU for both SA113 and the ica- mutant. Bacterial growth, initial adherence to surfaces within the implants and the course of inflammation including growth-dependent host TNF and MIP-2 release, influx of phagocytes and an accumulation of dead leukocytes were similar as well. Since SA113 expressed PIA in vivo, we could demonstrate that PIA and the lack of biofilm formation did not influence the capacity of S. aureus to induce persistent infections and did not modulate host responses in the mouse tissue cage model.


Assuntos
Biofilmes/crescimento & desenvolvimento , Polissacarídeos Bacterianos/biossíntese , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade , Animais , Aderência Bacteriana/genética , Aderência Bacteriana/fisiologia , Contagem de Células , Quimiocina CXCL2 , Quimiocinas/análise , Quimiocinas/imunologia , Contagem de Colônia Microbiana , Cultura em Câmaras de Difusão , Camundongos , Camundongos Endogâmicos C57BL , Fagócitos/imunologia , Polissacarídeos Bacterianos/genética , Staphylococcus aureus/genética , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/imunologia , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/imunologia , Virulência
18.
Nat Med ; 10(3): 243-5, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14758355

RESUMO

Colonization of the anterior nares in approximately 37% of the population is a major risk factor for severe Staphylococcus aureus infections. Here we show that wall teichoic acid (WTA), a surface-exposed staphylococcal polymer, is essential for nasal colonization and mediates interaction with human nasal epithelial cells. WTA-deficient mutants were impaired in their adherence to nasal cells, and were completely unable to colonize cotton rat nares. This study describes the first essential factor for S. aureus nasal colonization.


Assuntos
Infecção Hospitalar/microbiologia , Mucosa Nasal/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/crescimento & desenvolvimento , Ácidos Teicoicos/metabolismo , Animais , Aderência Bacteriana , Células Cultivadas , Células Epiteliais/microbiologia , Humanos , Estrutura Molecular , Mucosa Nasal/citologia , Ratos , Fatores de Risco , Sigmodontinae , Staphylococcus aureus/fisiologia , Ácidos Teicoicos/química
19.
Curr Drug Targets ; 4(8): 643-9, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14577655

RESUMO

Increasing bacterial resistance to virtually all available antibiotics causes an urgent need for new antimicrobial drugs, drug targets and therapeutic concepts. This review focuses on strategies to render bacteria highly susceptible to the antimicrobial arsenal of the immune system by targeting bacterial immune escape mechanisms that are conserved in a major number of pathogens. Virtually all innate molecules that inactivate bacteria, ranging from antimicrobial peptides such as defensins and cathelicidins to bacteriolytic enzymes such as lysozyme and group IIA phospholipase A2, are highly cationic in order to facilitate binding to the anionic bacterial cell envelopes. Bacteria have found ways to modulate their anionic cell wall polymers such as peptidoglycan, lipopolysaccharide, teichoic acid or phospholipids by introducing positively charged groups. Two of these mechanisms involving the transfer of D-alanine into teichoic acids and of L-lysine into phospholipids, respectively, have been identified and characterized in Staphylococcus aureus, a major human pathogen in community- and hospital-acquired infections. Inactivation of the responsible genes, dltABCD for alanylation of teichoic acids and mprF for lysinylation of phosphatidylglycerol, renders S. aureus highly susceptible to many human antimicrobial molecules and leads to profoundly attenuated virulence in several animal models. dltABCD- and mprF-related genes are found in the genomes of many bacterial pathogens indicating that the escape from human host defenses by modulation of the cell envelope is a general trait in pathogenic bacteria. This review suggests that inhibitors of DltABCD or MprF should have great potential in complementing or replacing the conventional antibiotic therapies.


Assuntos
Anti-Infecciosos/administração & dosagem , Anti-Infecciosos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Resistência Microbiana a Medicamentos/genética , Animais , Resistência a Medicamentos/genética , Humanos
20.
J Infect Dis ; 188(3): 414-23, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12870123

RESUMO

Staphylococcus aureus is inherently resistant to cationic antimicrobial peptides because of alanylation of cell envelope teichoic acids. To test the effect of alanylated teichoic acids on virulence and host defense mediated by Toll-like receptor 2 (TLR2), wild-type (wt) S. aureus ATCC35556 (S.a.113) and its isogenic mutant expressing unalanylated teichoic acids (dlt(-)) were compared in a tissue cage infection model that used C57BL/6 wt and TLR2-deficient mice. The minimum infective doses (MID) to establish persistent infection with S.a.113 were 10(3) and 10(2) colony-forming units (cfu) in wt and TLR2(-/-) mice, respectively. The corresponding MID for dlt(-) were 5x105 and 10(3) cfu in wt and TLR2(-/-) mice, respectively. Both mouse strains showed bacterial-load-dependent inflammation with elevations in tumor necrosis factor, macrophage inflammatory protein 2, and leukocytes, with increasing proportions of dead cells. These findings indicate that alanylated teichoic acids contribute to virulence of S. aureus, and TLR2 mediates host defense, which partly targets alanylated teichoic acids.


Assuntos
Glicoproteínas de Membrana/fisiologia , Peptídeos , Receptores de Superfície Celular/fisiologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/patogenicidade , Ácidos Teicoicos/imunologia , Animais , Antibacterianos/farmacologia , Bacteriocinas , Quimiocina CXCL2 , Modelos Animais de Doenças , Feminino , Inflamação/imunologia , Inflamação/microbiologia , Contagem de Leucócitos , Leucócitos/imunologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monocinas/análise , Mutação , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Infecções Estafilocócicas/sangue , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Ácidos Teicoicos/metabolismo , Receptor 2 Toll-Like , Receptores Toll-Like , Fator de Necrose Tumoral alfa/análise , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...