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

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 113(20): 5718-23, 2016 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-27140614

RESUMO

A hallmark of Staphylococcus aureus disease in humans is persistent infections without development of protective immune responses. Infected patients generate VH3 plasmablast expansions and increased VH3 idiotype Ig; however, the mechanisms for staphylococcal modification of immune responses are not known. We report here that S. aureus-infected mice generate VH3 antibody expansions via a mechanism requiring MHC-restricted antigen presentation to CD4(+) T cells and staphylococcal protein A (SpA), a cell wall-anchored surface molecule that binds Fcγ and VH3 variant heavy chains of Ig. VH3 expansion occurred with peptidoglycan-linked SpA from the bacterial envelope but not with recombinant SpA, and optimally required five tandem repeats of its Ig-binding domains. Signaling via receptor-interacting serine/threonine protein kinase 2 (RIPK2) was essential for implementing peptidoglycan-linked SpA superantigen activity. VH3 clan IgG from S. aureus-infected or SpA-treated animals was not pathogen-specific, suggesting that SpA cross-linking of VH3 idiotype B-cell receptors and activation via attached peptidoglycan are the determinants of staphylococcal escape from adaptive immune responses.


Assuntos
Anticorpos Antibacterianos/biossíntese , Infecções Estafilocócicas/imunologia , Proteína Estafilocócica A/imunologia , Staphylococcus aureus/imunologia , Linfócitos T/metabolismo , Animais , Anticorpos Antibacterianos/sangue , Formação de Anticorpos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Peptidoglicano/imunologia , Infecções Estafilocócicas/sangue , Infecções Estafilocócicas/microbiologia , Linfócitos T/imunologia , Linfócitos T/microbiologia
2.
Mol Cell Proteomics ; 11(4): M111.015206, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22199230

RESUMO

Very few studies have so far been dedicated to the systematic analysis of protein interactions occurring between surface and/or secreted proteins in bacteria. Such interactions are expected to play pivotal biological roles that deserve investigation. Taking advantage of the availability of a detailed map of surface and secreted proteins in Streptococcus pyogenes (group A Streptococcus (GAS)), we used protein array technology to define the "surface interactome" in this important human pathogen. Eighty-three proteins were spotted on glass slides in high density format, and each of the spotted proteins was probed for its capacity to interact with any of the immobilized proteins. A total of 146 interactions were identified, 25 of which classified as "reciprocal," namely, interactions that occur irrespective of which of the two partners was immobilized on the chip or in solution. Several of these interactions were validated by surface plasmon resonance and supported by confocal microscopy analysis of whole bacterial cells. By this approach, a number of interesting interactions have been discovered, including those occurring between OppA, DppA, PrsA, and TlpA, proteins known to be involved in protein folding and transport. These proteins, all localizing at the septum, might be part, together with HtrA, of the recently described ExPortal complex of GAS. Furthermore, SpeI was found to strongly interact with the metal transporters AdcA and Lmb. Because SpeI strictly requires zinc to exert its function, this finding provides evidence on how this superantigen, a major player in GAS pathogenesis, can acquire the metal in the host environment, where it is largely sequestered by carrier proteins. We believe that the approach proposed herein can lead to a deeper knowledge of the mechanisms underlying bacterial invasion, colonization, and pathogenesis.


Assuntos
Proteínas de Bactérias/metabolismo , Streptococcus pyogenes/metabolismo , Cloretos/farmacologia , Análise Serial de Proteínas , Ligação Proteica/efeitos dos fármacos , Compostos de Zinco/farmacologia
3.
Mol Cell Proteomics ; 11(6): M111.015693, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22286755

RESUMO

We propose an experimental strategy for highly accurate selection of candidates for bacterial vaccines without using in vitro and/or in vivo protection assays. Starting from the observation that efficacious vaccines are constituted by conserved, surface-associated and/or secreted components, the strategy contemplates the parallel application of three high throughput technologies, i.e. mass spectrometry-based proteomics, protein array, and flow-cytometry analysis, to identify this category of proteins, and is based on the assumption that the antigens identified by all three technologies are the protective ones. When we tested this strategy for Group A Streptococcus, we selected a total of 40 proteins, of which only six identified by all three approaches. When the 40 proteins were tested in a mouse model, only six were found to be protective and five of these belonged to the group of antigens in common to the three technologies. Finally, a combination of three protective antigens conferred broad protection against a panel of four different Group A Streptococcus strains. This approach may find general application as an accelerated and highly accurate path to bacterial vaccine discovery.


Assuntos
Antígenos de Bactérias/imunologia , Vacinas Bacterianas/administração & dosagem , Infecções Estreptocócicas/prevenção & controle , Streptococcus pyogenes/imunologia , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Análise por Conglomerados , Feminino , Citometria de Fluxo , Hemólise , Humanos , Camundongos , Faringite/sangue , Faringite/imunologia , Faringite/microbiologia , Análise Serial de Proteínas , Proteoma/imunologia , Proteoma/metabolismo , Ovinos , Infecções Estreptocócicas/sangue , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/metabolismo , Vacinação
4.
Biochem J ; 455(3): 273-84, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23895222

RESUMO

In the human pathogen Staphylococcus aureus, there exists an enormous diversity of proteins containing DUFs (domains of unknown function). In the present study, we characterized the family of conserved staphylococcal antigens (Csa) classified as DUF576 and taxonomically restricted to Staphylococci. The 18 Csa paralogues in S. aureus Newman are highly similar at the sequence level, yet were found to be expressed in multiple cellular locations. Extracellular Csa1A was shown to be post-translationally processed and released. Molecular interaction studies revealed that Csa1A interacts with other Csa paralogues, suggesting that these proteins are involved in the same cellular process. The structures of Csa1A and Csa1B were determined by X-ray crystallography, unveiling a peculiar structure with limited structural similarity to other known proteins. Our results provide the first detailed biological characterization of this family and confirm the uniqueness of this family also at the structural level. We also provide evidence that Csa family members elicit protective immunity in in vivo animal models of staphylococcal infections, indicating a possible important role for these proteins in S. aureus biology and pathogenesis. These findings identify the Csa family as new potential vaccine candidates, and underline the importance of mining the bacterial unknown proteome to identify new targets for preventive vaccines.


Assuntos
Antígenos de Bactérias/química , Proteínas de Bactérias/química , Proteoma/química , Staphylococcus aureus/metabolismo , Animais , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , Mineração de Dados , Camundongos , Camundongos Endogâmicos , Proteoma/genética , Proteoma/metabolismo , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/imunologia
5.
Artigo em Inglês | MEDLINE | ID: mdl-24100558

RESUMO

Streptococcus pyogenes (Group A streptococcus; GAS) is an important human pathogen against which an effective vaccine does not yet exist. The S. pyogenes protein SpyCEP (S. pyogenes cell-envelope proteinase) is a surface-exposed subtilisin-like serine protease of 1647 amino acids. In addition to its auto-protease activity, SpyCEP is capable of cleaving interleukin 8 and related chemokines, contributing to GAS immune-evasion strategies. SpyCEP is immunogenic and confers protection in animal models of GAS infections. In order to structurally characterize this promising vaccine candidate, several SpyCEP protein-expression constructs were designed, cloned, produced in Escherichia coli, purified by affinity chromatography and subjected to crystallization trials. Crystals of a selenomethionyl form of a near-full-length SpyCEP ectodomain were obtained. The crystals diffracted X-rays to 3.3 Šresolution and belonged to space group C2, with unit-cell parameters a=139.2, b=120.4, c=104.3 Å, ß=111°.


Assuntos
Antígenos de Bactérias/química , Antígenos de Bactérias/isolamento & purificação , Peptídeo Hidrolases/química , Peptídeo Hidrolases/isolamento & purificação , Vacinas Estreptocócicas/imunologia , Streptococcus pyogenes/imunologia , Difração de Raios X , Clonagem Molecular , Cristalização , Cristalografia por Raios X , Humanos , Peptídeo Hidrolases/imunologia , Estrutura Terciária de Proteína , Selenometionina/química
6.
J Infect Dis ; 206(7): 1041-9, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22829645

RESUMO

Iron availability plays an essential role in staphylococcal pathogenesis. We selected FhuD2, a lipoprotein involved in iron-hydroxamate uptake, as a novel vaccine candidate against Staphylococcus aureus. Unprecedented for staphylococcal lipoproteins, the protein was demonstrated to have a discrete, punctate localization on the bacterial surface. FhuD2 vaccination generated protective immunity against diverse clinical S. aureus isolates in murine infection models. Protection appeared to be associated with functional antibodies that were shown to mediate opsonophagocytosis, to be effective in passive transfer experiments, and to potentially block FhuD2-mediated siderophore uptake. Furthermore, the protein was found to be up-regulated in infected tissues and was required for staphylococcal dissemination and abscess formation. Herein we show that the staphylococcal iron-hydroxamate uptake system is important in invasive infection and functions as an efficacious vaccine target.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/metabolismo , Vacinação , Abscesso/imunologia , Abscesso/prevenção & controle , Sequência de Aminoácidos , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Compostos Férricos/metabolismo , Regulação Bacteriana da Expressão Gênica , Técnicas de Inativação de Genes , Células HL-60 , Humanos , Ácidos Hidroxâmicos/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/imunologia , Camundongos , Dados de Sequência Molecular , Transporte Proteico , Coelhos , Sepse/imunologia , Sepse/prevenção & controle , Infecções Estafilocócicas/imunologia , Vacinas Antiestafilocócicas/administração & dosagem , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/imunologia
7.
Infect Immun ; 80(10): 3460-70, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22825452

RESUMO

Staphylococcus aureus is a leading cause of human soft tissue infections and bacterial sepsis. The emergence of antibiotic-resistant strains (methicillin-resistant S. aureus [MRSA]) has prompted research into staphylococcal vaccines and preventive measures. The envelope of S. aureus is decorated with staphylococcal protein A (SpA), which captures the Fcγ portion of immunoglobulins to prevent opsonophagocytosis and associates with the Fab portion of V(H)3-type B cell receptors to trigger B cell superantigen activity. Nontoxigenic protein A (SpA(KKAA)), when used as an immunogen in mice, stimulates humoral immune responses that neutralize the Fcγ and the V(H)3(+) Fab binding activities of SpA and provide protection from staphylococcal abscess formation in mice. Here, we isolated monoclonal antibodies (MAbs) against SpA(KKAA) that, by binding to the triple-helical bundle fold of its immunoglobulin binding domains (IgBDs), neutralize the Fcγ and Fab binding activities of SpA. SpA(KKAA) MAbs promoted opsonophagocytic killing of MRSA in mouse and human blood, provided protection from abscess formation, and stimulated pathogen-specific immune responses in a mouse model of staphylococcal disease. Thus, SpA(KKAA) MAbs may be useful for the prevention and therapy of staphylococcal disease in humans.


Assuntos
Anticorpos Antibacterianos/sangue , Anticorpos Monoclonais/sangue , Staphylococcus aureus Resistente à Meticilina/imunologia , Infecções Estafilocócicas/imunologia , Proteína Estafilocócica A/metabolismo , Abscesso/microbiologia , Abscesso/prevenção & controle , Animais , Especificidade de Anticorpos , Sítios de Ligação de Anticorpos , Humanos , Nefropatias/microbiologia , Nefropatias/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Terciária de Proteína , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/prevenção & controle , Proteína Estafilocócica A/genética , Proteína Estafilocócica A/imunologia
8.
Microbiol Spectr ; 10(2): e0271621, 2022 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-35389241

RESUMO

Staphylococcus aureus remains a leading cause of skin and soft tissue infections (SSTIs) globally. In the United States, many of these infections are caused by isolates classified as USA300. Our understanding of the success of USA300 as a human pathogen is due in part to data obtained from animal infection models, including rabbit SSTI models. These animal models have been used to study S. aureus virulence and pathogenesis and to gain an enhanced understanding of the host response to infection. Although significant knowledge has been gained, the need to use a relatively high inoculum of USA300 (1 × 108 to 5 × 108 CFU) is a caveat of these infection models. As a step toward addressing this issue, we created mutations in USA300 that mimic those found in S. aureus strains with naturally occurring rabbit tropism-namely, single nucleotide polymorphisms in dltB and/or deletion of rot. We then developed a rabbit SSTI model that utilizes an inoculum of 106 USA300 CFU to cause reproducible disease and tested whether primary SSTI protects rabbits against severe reinfection caused by the same strain. Although there was modest protection against severe reinfection, primary infection and reinfection with rabbit-tropic USA300 strains failed to increase the overall level of circulating anti-S. aureus antibodies significantly. These findings provide additional insight into the host response to S. aureus. More work is needed to further develop a low-inoculum infection model that can be used to better test the potential of new therapeutics or vaccine target antigens. IMPORTANCE Animal models of S. aureus infection are important for evaluating bacterial pathogenesis and host immune responses. These animal infection models are often used as an initial step in the testing of vaccine antigens and new therapeutics. The extent to which animal models of S. aureus infection approximate human infections remains a significant consideration for translation of results to human clinical trials. Although significant progress has been made with rabbit models of S. aureus infection, one concern is the high inoculum needed to cause reproducible disease. Here, we generated USA300 strains that have tropism for rabbits and developed a rabbit SSTI model that uses fewer CFU than previous models.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Infecções dos Tecidos Moles , Infecções Estafilocócicas , Infecções Cutâneas Estafilocócicas , Vacinas , Animais , Staphylococcus aureus Resistente à Meticilina/genética , Coelhos , Reinfecção , Infecções Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/microbiologia , Staphylococcus aureus , Estados Unidos
9.
FASEB J ; 24(8): 2839-48, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20339024

RESUMO

SpyCEP is a 170-kDa multidomain serine protease expressed on the surface of the human pathogen Streptococcus pyogenes, which plays an important role in infection by catalyzing cleavage and inactivation of the neutrophil chemoattractant interleukin-8. In this study, we investigated the biochemical features and maturation process of SpyCEP, starting from a recombinant form of the protease expressed and purified from Escherichia coli. We show that active recombinant SpyCEP differs from other bacterial proteases in that it is constituted by 2 noncovalently linked fragments derived from autocatalytic processing, an N-terminal fragment of 210 aa bearing one of the 3 catalytic triad residues, and a 1369-residue C-terminal polypeptide containing the remaining 2 catalytic amino acids. The same type of organization is present in the enzyme obtained from S. pyogenes. Furthermore, N-terminal SpyCEP is not involved in the folding of the mature enzyme. The 2 protease fragments were separately expressed in E. coli as soluble polypeptides that, when combined, reconstituted a fully active enzyme complex. Therefore, SpyCEP appears to possess a completely new structural architecture that has not been described so far for other microbial proteases.


Assuntos
Fragmentos de Peptídeos/química , Peptídeo Hidrolases/química , Clonagem Molecular , Humanos , Proteínas Recombinantes , Streptococcus pyogenes
10.
Mol Microbiol ; 68(6): 1378-94, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18452511

RESUMO

Group A streptococci (GAS) are the most frequent cause of bacterial pharyngitis. The first obstacle to GAS colonization of the pharynx is saliva. As well as forming a physical barrier, saliva contains components of innate and acquired immunity. Previous work has shown that saliva induces bacterial aggregation, which may serve as a clearance mechanism. As the aggregation of some oral streptococci in saliva is mediated by long proteinaceous appendages, we hypothesized that pili of GAS might behave similarly. Wild-type GAS M1 strain SF370 aggregated in saliva, while pilus-defective mutants did not. Similarly, heterologous expression of diverse GAS pili on the surface of Lactococcus lactis induced aggregation in saliva, while control strains were unaffected. Further studies revealed that aggregating bacteria bound salivary component gp340. Purified gp340 aggregated wild-type GAS and L. lactis expressing GAS pili, but not control strains. GAS pilus-defective mutants were abrogated in gp340 binding and aggregation. Furthermore, gp340-mediated aggregation reduced bacterial adhesion to human epithelial cells, suggesting a role in host defence.


Assuntos
Aderência Bacteriana , Receptores de Superfície Celular/metabolismo , Proteínas e Peptídeos Salivares/metabolismo , Streptococcus pyogenes/fisiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Ligação ao Cálcio , Linhagem Celular , Proteínas de Ligação a DNA , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/metabolismo , Humanos , Lactococcus lactis/genética , Lactococcus lactis/fisiologia , Camundongos , Receptores de Superfície Celular/isolamento & purificação , Saliva/metabolismo , Proteínas e Peptídeos Salivares/isolamento & purificação , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/genética , Proteínas Supressoras de Tumor
11.
Infect Immun ; 76(3): 978-85, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18174342

RESUMO

Circumvention of the host innate immune response is critical for bacterial pathogens to infect and cause disease. Here we demonstrate that the group A Streptococcus (GAS; Streptococcus pyogenes) protease SpyCEP (S. pyogenes cell envelope protease) cleaves granulocyte chemotactic protein 2 (GCP-2) and growth-related oncogene alpha (GROalpha), two potent chemokines made abundantly in human tonsils. Cleavage of GCP-2 and GROalpha by SpyCEP abrogated their abilities to prime neutrophils for activation, detrimentally altering the innate immune response. SpyCEP expression is negatively regulated by the signal transduction system CovR/S. Purified recombinant CovR bound the spyCEP gene promoter region in vitro, indicating direct regulation. Immunoreactive SpyCEP protein was present in the culture supernatants of covR/S mutant GAS strains but not in supernatants from wild-type strains. However, wild-type GAS strains do express SpyCEP, where it is localized to the cell wall. Strain MGAS2221, an organism representative of the highly virulent and globally disseminated M1T1 GAS clone, differed significantly from its isogenic spyCEP mutant derivative strain in a mouse soft tissue infection model. Interestingly, and in contrast to previous studies, the isogenic mutant strain generated lesions of larger size than those formed following infection with the parent strain. The data indicate that SpyCEP contributes to GAS virulence in a strain- and disease-dependent manner.


Assuntos
Quimiocina CXCL1/metabolismo , Quimiocina CXCL6/metabolismo , Peptídeo Hidrolases/metabolismo , Streptococcus pyogenes/enzimologia , Streptococcus pyogenes/imunologia , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Parede Celular/química , DNA Bacteriano/metabolismo , Feminino , Deleção de Genes , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Humanos , Camundongos , Ativação de Neutrófilo , Neutrófilos/imunologia , Peptídeo Hidrolases/genética , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Repressoras/metabolismo , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidade , Virulência , Fatores de Virulência/genética
12.
Clin Vaccine Immunol ; 23(6): 442-50, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27030589

RESUMO

Staphylococcus aureus alpha-hemolysin (Hla) assembles into heptameric pores on the host cell membrane, causing lysis, apoptosis, and junction disruption. Herein, we present the design of a newly engineered S. aureus alpha-toxin, HlaPSGS, which lacks the predicted membrane-spanning stem domain. This protein is able to form heptamers in aqueous solution in the absence of lipophilic substrata, and its structure, obtained by transmission electron microscopy and single-particle reconstruction analysis, resembles the cap of the wild-type cytolytic Hla pore. HlaPSGS was found to be impaired in binding to host cells and to its receptor ADAM10 and to lack hemolytic and cytotoxic activity. Immunological studies using human sera as well as sera from mice convalescent from S. aureus infection suggested that the heptameric conformation of HlaPSGS mimics epitopes exposed by the cytolytic Hla pore during infection. Finally, immunization with this newly engineered Hla generated high protective immunity against staphylococcal infection in mice. Overall, this study provides unprecedented data on the natural immune response against Hla and suggests that the heptameric HlaPSGS is a highly valuable vaccine candidate against S. aureus.


Assuntos
Toxinas Bacterianas/química , Toxinas Bacterianas/imunologia , Proteínas Hemolisinas/química , Proteínas Hemolisinas/imunologia , Mimetismo Molecular , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus , Proteína ADAM10/metabolismo , Animais , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/genética , Linhagem Celular , Citotoxinas , Epitopos/imunologia , Escherichia coli/genética , Proteínas Hemolisinas/administração & dosagem , Proteínas Hemolisinas/genética , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Engenharia de Proteínas , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação , Vacinas Antiestafilocócicas/imunologia , Staphylococcus aureus/química , Staphylococcus aureus/metabolismo , Vacinação
13.
mBio ; 6(1)2015 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-25564466

RESUMO

UNLABELLED: Staphylococcus aureus infection is not associated with the development of protective immunity, and disease relapses occur frequently. We hypothesize that protein A, a factor that binds immunoglobulin Fcγ and cross-links V(H)3 clan B cell receptors (IgM), is the staphylococcal determinant for host immune suppression. To test this, vertebrate IgM was examined for protein A cross-linking. High V(H)3 binding activity occurred with human and guinea immunoglobulin, whereas mouse and rabbit immunoglobulins displayed little and no binding, respectively. Establishing a guinea pig model of S. aureus bloodstream infection, we show that protein A functions as a virulence determinant and suppresses host B cell responses. Immunization with SpA(KKAA), which cannot bind immunoglobulin, elicits neutralizing antibodies that enable guinea pigs to develop protective immunity. IMPORTANCE: Staphylococcus aureus is the leading cause of soft tissue and bloodstream infections; however, a vaccine with clinical efficacy is not available. Using mice to model staphylococcal infection, earlier work identified protective antigens; however, corresponding human clinical trials did not reach their endpoints. We show that B cell receptor (IgM) cross-linking by protein A is an important immune evasion strategy of S. aureus that can be monitored in a guinea pig model of bloodstream infection. Further, immunization with nontoxigenic protein A enables infected guinea pigs to elicit antibody responses that are protective against S. aureus. Thus, the guinea pig model may support preclinical development of staphylococcal vaccines.


Assuntos
Formação de Anticorpos , Bacteriemia/imunologia , Infecções Estafilocócicas/imunologia , Proteína Estafilocócica A/imunologia , Staphylococcus aureus/imunologia , Animais , Anticorpos Antibacterianos/química , Anticorpos Antibacterianos/imunologia , Bacteriemia/microbiologia , Feminino , Cobaias , Humanos , Evasão da Resposta Imune , Imunoglobulina M/química , Imunoglobulina M/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Coelhos , Infecções Estafilocócicas/microbiologia , Proteína Estafilocócica A/genética , Staphylococcus aureus/genética
14.
J Exp Med ; 211(12): 2331-9, 2014 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-25348152

RESUMO

Staphylococcus aureus bacterial infection commonly results in chronic or recurrent disease, suggesting that humoral memory responses are hampered. Understanding how S. aureus subverts the immune response is critical for the rescue of host natural humoral immunity and vaccine development. S. aureus expresses the virulence factor Protein A (SpA) on all clinical isolates, and SpA has been shown in mice to expand and ablate variable heavy 3 (VH3) idiotype B cells. The effects of SpA during natural infection, however, have not been addressed. Acutely activated B cells, or plasmablasts (PBs), were analyzed to dissect the ongoing immune response to infection through the production of monoclonal antibodies (mAbs). The B cells that were activated by infection had a highly limited response. When screened against multiple S. aureus antigens, only high-affinity binding to SpA was observed. Consistently, PBs underwent affinity maturation, but their B cell receptors demonstrated significant bias toward the VH3 idiotype. These data suggest that the superantigenic activity of SpA leads to immunodominance, limiting host responses to other S. aureus virulence factors that would be necessary for protection and memory formation.


Assuntos
Evasão da Resposta Imune/imunologia , Infecções Estafilocócicas/imunologia , Proteína Estafilocócica A/imunologia , Staphylococcus aureus/imunologia , Adulto , Idoso , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Citometria de Fluxo , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Plasmócitos/imunologia , Plasmócitos/metabolismo , Células Precursoras de Linfócitos B/imunologia , Células Precursoras de Linfócitos B/metabolismo , Análise de Sequência de DNA , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Staphylococcus aureus/fisiologia , Fatores de Virulência/imunologia , Adulto Jovem
15.
mBio ; 4(5): e00575-13, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23982075

RESUMO

UNLABELLED: Heritable defects in human B cell/antibody development are not associated with increased susceptibility to Staphylococcus aureus infection. Protein A (SpA), a surface molecule of S. aureus, binds the Fcγ domain of immunoglobulin (Ig) and cross-links the Fab domain of VH3-type B cell receptors (IgM). Here we generated S. aureus spa variants harboring amino acid substitutions at four key residues in each of the five Ig-binding domains of SpA. Wild-type S. aureus required SpA binding to Ig to resist phagocytosis and SpA-mediated B cell receptor cross-linking to block antibody development in mice. The spaKKAA mutant, which cannot bind Ig or IgM, was phagocytosed and elicited B cell responses to key virulence antigens that protected animals against lethal S. aureus challenge. The immune evasive attributes of S. aureus SpA were abolished in µMT mice lacking mature B cells and antibodies. Thus, while wild-type S. aureus escapes host immune surveillance, the spaKKAA variant elicits adaptive responses that protect against recurrent infection. IMPORTANCE: Staphylococcus aureus causes recurrent skin and bloodstream infections without eliciting immunity. Heritable defects in neutrophil and T cell function, but not B cell or antibody development, are associated with increased incidence of S. aureus infection, and efforts to develop antibody-based S. aureus vaccines have thus far been unsuccessful. We show here that the Fcγ and VH3-type Fab binding activities of staphylococcal protein A (SpA) are essential for S. aureus escape from host immune surveillance in mice. The virulence attributes of SpA in mice required mature B cells and immunoglobulin. These results suggest that antibodies and B cells play a key role in the pathogenesis of staphylococcal infections and provide insights into the development of a vaccine against S. aureus.


Assuntos
Imunidade Adaptativa , Evasão da Resposta Imune , Infecções Estafilocócicas/imunologia , Proteína Estafilocócica A/imunologia , Staphylococcus aureus/imunologia , Animais , Linfócitos B/imunologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Infecções Estafilocócicas/microbiologia , Proteína Estafilocócica A/genética , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidade , Virulência
16.
mBio ; 4(1): e00387-12, 2013 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-23300245

RESUMO

UNLABELLED: Streptolysin O is a potent pore-forming toxin produced by group A Streptococcus. The aims of the present study were to dissect the relative contributions of different structural domains of the protein to hemolytic activity, to obtain a detoxified form of streptolysin O amenable to human vaccine formulation, and to investigate the role of streptolysin O-specific antibodies in protection against group A Streptococcus infection. On the basis of in silico structural predictions, we introduced two amino acid substitutions, one in the proline-rich domain 1 and the other in the conserved undecapeptide loop in domain 4. The resulting streptolysin O derivative showed no toxicity, was highly impaired in binding to eukaryotic cells, and was unable to form organized oligomeric structures on the cell surface. However, it was fully capable of conferring consistent protection in a murine model of group A Streptococcus infection. When we engineered a streptococcal strain to express the double-mutated streptolysin O, a drastic reduction in virulence as well as a diminished capacity to kill immune cells recruited at the infection site was observed. Furthermore, when mice immunized with the toxoid were challenged with the wild-type and mutant strains, protection only against the wild-type strain, not against the strain expressing the double-mutated streptolysin O, was obtained. We conclude that protection occurs by antibody-mediated neutralization of active toxin. IMPORTANCE: We present a novel example of structural design of a vaccine antigen optimized for human vaccine use. Having previously demonstrated that immunization of mice with streptolysin O elicits a protective immune response against infection with group A Streptococcus strains of different serotypes, we developed in this study a double-mutated nontoxic derivative that represents a novel tool for the development of protective vaccine formulations against this important human pathogen. Furthermore, the innovative construction of an isogenic strain expressing a functionally inactive toxin and its use in infection and opsonophagocytosis experiments allowed us to investigate the mechanism by which streptolysin O mediates protection against group A Streptococcus. Finally, the ability of this toxin to directly attack and kill host immune cells during infection was studied in an air pouch model, which allowed parallel quantification of cellular recruitment, vitality, and cytokine release at the infection site.


Assuntos
Substituição de Aminoácidos , Streptococcus pyogenes/patogenicidade , Estreptolisinas/genética , Estreptolisinas/toxicidade , Fatores de Virulência/genética , Fatores de Virulência/toxicidade , Animais , Anticorpos Antibacterianos/sangue , Antitoxinas/sangue , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/toxicidade , Modelos Animais de Doenças , Camundongos , Modelos Moleculares , Proteínas Mutantes/genética , Proteínas Mutantes/imunologia , Proteínas Mutantes/toxicidade , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Infecções Estreptocócicas/prevenção & controle , Streptococcus pyogenes/genética , Streptococcus pyogenes/imunologia , Estreptolisinas/imunologia , Análise de Sobrevida , Virulência , Fatores de Virulência/imunologia
17.
PLoS One ; 4(7): e6332, 2009 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-19623252

RESUMO

The human pathogen Group A Streptococcus (Streptococcus pyogenes, GAS) is widely recognized as a major cause of common pharyngitis as well as of severe invasive diseases and non-suppurative sequelae associated with the existence of GAS antigens eliciting host autoantibodies. It has been proposed that a subset of paediatric disorders characterized by tics and obsessive-compulsive symptoms would exacerbate in association with relapses of GAS-associated pharyngitis. This hypothesis is however still controversial. In the attempt to shed light on the contribution of GAS infections to the onset of neuropsychiatric or behavioral disorders affecting as many as 3% of children and adolescents, we tested the antibody response of tic patient sera to a representative panel of GAS antigens. In particular, 102 recombinant proteins were spotted on nitrocellulose-coated glass slides and probed against 61 sera collected from young patients with typical tic neuropsychiatric symptoms but with no overt GAS infection. Sera from 35 children with neither tic disorder nor overt GAS infection were also analyzed. The protein recognition patterns of these two sera groups were compared with those obtained using 239 sera from children with GAS-associated pharyngitis. This comparative analysis identified 25 antigens recognized by sera of the three patient groups and 21 antigens recognized by tic and pharyngitis sera, but poorly or not recognized by sera from children without tic. Interestingly, these antigens appeared to be, in quantitative terms, more immunogenic in tic than in pharyngitis patients. Additionally, a third group of antigens appeared to be preferentially and specifically recognized by tic sera. These findings provide the first evidence that tic patient sera exhibit immunological profiles typical of individuals who elicited a broad, specific and strong immune response against GAS. This may be relevant in the context of one of the hypothesis proposing that GAS antigen-dependent induction of autoantibodies in susceptible individuals may be involved the occurrence of tic disorders.


Assuntos
Antígenos de Bactérias/imunologia , Faringite/imunologia , Análise Serial de Proteínas , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes/imunologia , Tiques/sangue , Estudos de Casos e Controles , Criança , Humanos , Faringite/sangue , Infecções Estreptocócicas/sangue
18.
J Infect Dis ; 198(12): 1834-41, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18928376

RESUMO

BACKGROUND: We previously reported that group A Streptococcus (GAS) pili are the T antigens described by Rebecca Lancefield. We also showed that these pili, constituted by backbone, ancillary 1, and ancillary 2 proteins, confer protection against GAS challenge in a mouse model. METHODS: We evaluated pilus distribution and conservation by sequencing the subunits of 39 new GAS isolates and used immunoblot analysis and agglutination assays to define the specificity of T sera to pilus subunits. RESULTS: GAS pili are encoded by 9 different islands within which backbone protein, ancillary protein 1, and ancillary protein 2 cluster in 15, 16, and 5 variants, respectively. Immunoblot and agglutination assays revealed that T type is determined by the backbone variant. This observation enabled us to set up a simple polymerase chain reaction assay to define the T type of GAS isolates. CONCLUSIONS: We propose the use of a tee gene sequence typing, analogous to the emm gene typing, as a valuable molecular tool that could substitute for the serological T classification of GAS strains. From our sequence analysis and from recent epidemiological data, we estimate that a vaccine comprising a combination of 12 backbone variants would protect against > 90% of currently circulating strains.


Assuntos
Fímbrias Bacterianas/genética , Variação Genética , Streptococcus pyogenes/genética , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fímbrias Bacterianas/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Genes Bacterianos , Filogenia , Streptococcus pyogenes/classificação
19.
Mol Microbiol ; 64(4): 968-83, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17501921

RESUMO

Group A Streptococcus (GAS, Streptococcus pyogenes) is a Gram-positive human pathogen responsible for several acute diseases and autoimmune sequelae that account for half a million deaths worldwide every year. GAS infections require the capacity of the pathogen to adhere to host tissues and assemble in cell aggregates. Furthermore, a role for biofilms in GAS pathogenesis has recently been proposed. Here we investigated the role of GAS pili in biofilm formation. We demonstrated that GAS pilus-negative mutants, in which the genes encoding either the pilus backbone structural protein or the sortase C1 have been deleted, showed an impaired capacity to attach to a pharyngeal cell line. The same mutants were much less efficient in forming cellular aggregates in liquid culture and microcolonies on human cells. Furthermore, mutant strains were incapable of producing the typical three-dimensional layer with bacterial microcolonies embedded in a carbohydrate polymeric matrix. Complemented mutants had an adhesion and aggregation phenotype similar to the wild-type strain. Finally, in vivo expression of pili was indirectly confirmed by demonstrating that most of the sera from human patients affected by GAS-mediated pharyngitis recognized recombinant pili proteins. These data support the role of pili in GAS adherence and colonization and suggest a general role of pili in all pathogenic streptococci.


Assuntos
Adesinas Bacterianas/fisiologia , Aderência Bacteriana/fisiologia , Biofilmes/crescimento & desenvolvimento , Fímbrias Bacterianas/fisiologia , Faringe/microbiologia , Streptococcus pyogenes/patogenicidade , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Aminoaciltransferases/genética , Anticorpos Antibacterianos/sangue , Aderência Bacteriana/genética , Proteínas de Bactérias/genética , Linhagem Celular , Cisteína Endopeptidases/genética , Células Epiteliais/microbiologia , Epitélio/microbiologia , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/imunologia , Deleção de Genes , Teste de Complementação Genética , Humanos , Microscopia Confocal , Microscopia Imunoeletrônica , Faringe/citologia , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/fisiologia
20.
Proc Natl Acad Sci U S A ; 102(43): 15641-6, 2005 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-16223875

RESUMO

Although pili have long been recognized in Gram-negative pathogens as important virulence factors involved in adhesion and invasion, very little is known about extended surface organelles in Gram-positive pathogens. Here we report that Group A Streptococcus (GAS), a Gram-positive human-specific pathogen that causes pharyngitis, impetigo, invasive disease, necrotizing fasciitis, and autoimmune sequelae has long, surface-exposed, pilus-like structures composed of members of a family of extracellular matrix-binding proteins. We describe four variant pili and show that each is recognized by a specific serum of the Lancefield T-typing system, which has been used for over five decades to characterize GAS isolates. Furthermore, we show that immunization of mice with a combination of recombinant pilus proteins confers protection against mucosal challenge with virulent GAS bacteria. The data indicate that induction of a protective immune response against these structures may be a useful strategy for development of a vaccine against disease caused by GAS infection.


Assuntos
Antígenos de Bactérias/análise , Fímbrias Bacterianas/fisiologia , Streptococcus pyogenes/imunologia , Animais , Sequência de Bases , Feminino , Proteínas de Fímbrias/imunologia , Imunização , Camundongos , Dados de Sequência Molecular , Infecções Estreptocócicas/prevenção & controle , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidade , Fatores de Virulência/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA