Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-37886572

RESUMO

The spherical bacterium Staphylococcus aureus, a leading cause of nosocomial infections, undergoes binary fission by dividing in two alternating orthogonal planes, but the mechanism by which S. aureus correctly selects the next cell division plane is not known. To identify cell division placement factors, we performed a chemical genetic screen that revealed a gene which we termed pcdA. We show that PcdA is a member of the McrB family of AAA+ NTPases that has undergone structural changes and a concomitant functional shift from a restriction enzyme subunit to an early cell division protein. PcdA directly interacts with the tubulin-like central divisome component FtsZ and localizes to future cell division sites before membrane invagination initiates. This parallels the action of another McrB family protein, CTTNBP2, which stabilizes microtubules in animals. We show that PcdA also interacts with the structural protein DivIVA and propose that the DivIVA/PcdA complex recruits unpolymerized FtsZ to assemble along the proper cell division plane. Deletion of pcdA conferred abnormal, non-orthogonal division plane selection, increased sensitivity to cell wall-targeting antibiotics, and reduced virulence in a murine infection model. Targeting PcdA could therefore highlight a treatment strategy for combatting antibiotic-resistant strains of S. aureus.

2.
Proc Natl Acad Sci U S A ; 116(39): 19659-19664, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31413191

RESUMO

Rickettsial diseases have long been diagnosed with serum antibodies cross-reactive against Proteus vulgaris (Weil-Felix reaction). Although Weil-Felix antibodies are associated with the development of immunity, their rickettsial target and contribution to disease pathogenesis are not established. Here, we developed a transposon for insertional mutagenesis of Rickettsia conorii, isolating variants defective for replication in cultured cells and in spotted fever pathogenesis. Mutations in the polysaccharide synthesis operon (pso) abolish lipopolysaccharide O-antigen synthesis and Weil-Felix serology and alter outer-membrane protein assembly. Unlike wild-type R. conorii, pso mutants cannot elicit bactericidal antibodies that bind O antigen. The pso operon is conserved among rickettsial pathogens, suggesting that bactericidal antibodies targeting O antigen may generate universal immunity that could be exploited to develop vaccines against rickettsial diseases.


Assuntos
Reações Cruzadas/imunologia , Antígenos O/imunologia , Rickettsia conorii/imunologia , Antibacterianos , Anticorpos Antibacterianos/imunologia , Lipopolissacarídeos/imunologia , Rickettsia/imunologia , Rickettsia/patogenicidade , Infecções por Rickettsia/imunologia , Rickettsia conorii/patogenicidade
3.
Microbiol Spectr ; 7(4)2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31267890

RESUMO

The highly cross-linked peptidoglycan represents the rigid layer of the bacterial envelope and protects bacteria from osmotic lysis. In Gram-positive bacteria, peptidoglycan also functions as a scaffold for the immobilization of capsular polysaccharide, wall teichoic acid (WTA), and surface proteins. This chapter captures recent development on the assembly of the envelope of Staphylococcus aureus including mechanisms accounting for immobilization of molecules to peptidoglycan as well as hydrolysis of peptidoglycan for the specific release of bound molecules, facilitation of protein secretion across the envelope and cell division. Peptidoglycan, WTA and capsular polysaccharide are directly synthesized onto undecaprenol. Surface proteins are anchored by Sortase A, a membrane-embedded transpeptidase that scans secreted polypeptides for the C-terminal LPXTG motif of sorting signals. The resulting acyl enzyme intermediate is resolved by lipid II, the undecaprenol-bound peptidoglycan precursor. While these pathways share membrane diffusible undecaprenol, assembly of these molecules occurs either at the cross-walls or the cell poles. In S. aureus, the cross-wall represents the site of de novo peptidoglycan synthesis which is eventually split to complete the cell cycle yielding newly divided daughter cells. Peptidoglycan synthesized at the cross-wall is initially devoid of WTA. Conversely, lipoteichoic acid (LTA) synthesis which does not require bactoprenol is seemingly restricted to septal membranes. Similarly, S. aureus distinguishes two types of surface protein precursors. Polypeptides with canonical signal peptides are deposited at the cell poles, whereas precursors with conserved YSIRK-GXXS motif signal peptides traffic to the cross-wall. A model for protein trafficking in the envelope and uneven distribution of teichoic acids is discussed.


Assuntos
Proteínas de Bactérias/metabolismo , Membrana Celular/metabolismo , Proteínas de Membrana/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Animais , Proteínas de Bactérias/genética , Membrana Celular/genética , Parede Celular/genética , Parede Celular/metabolismo , Humanos , Proteínas de Membrana/genética , Peptidoglicano/metabolismo , Transporte Proteico , Staphylococcus aureus/genética
4.
Arch Microbiol ; 200(7): 1075-1086, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29737367

RESUMO

The ESAT-6-like secretion system (ESS) of Staphylococcus aureus plays a significant role in persistent infections. EssB is a highly conserved bitopic ESS protein comprising a cytosolic N-terminus, single transmembrane helix and a C-terminus located on the trans-side of the membrane. Six systematic truncations covering various domains of EssB were constructed, followed by bacterial two-hybrid screening of their interaction with EsaA, another conserved integral membrane component of the ESS pathway. Results show that the transmembrane domain of EssB is critical for heterodimerization with EsaA. In vivo crosslinking followed by Western blot analysis revealed high molecular weight species when wild-type EssB and EsaA were crosslinked, but this band was not detected in the absence of the transmembrane domain of EssB. Heterologous overproduction of EssB, EsaA and five other components of the ESS pathway in Escherichia coli BL21(DE3), followed by fractionation experiments led to a remarkable increase in the periplasmic protein content, suggesting the assembly of partially regulated secretion mechanism. These data identify the transmembrane domain of EssB as indispensable for interaction with EsaA, thereby facilitating protein secretion across bacterial membranes in a fashion that requires other components of the ESS pathway.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Membrana/metabolismo , Staphylococcus aureus/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Ligação Proteica , Domínios Proteicos , Transporte Proteico , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/química , Staphylococcus aureus/genética
5.
J Bacteriol ; 200(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29229702

RESUMO

Bacillus anthracis, the causative agent of anthrax disease, elaborates a secondary cell wall polysaccharide (SCWP) that is essential for bacterial growth and cell division. B. anthracis SCWP is comprised of trisaccharide repeats with the structure, [→4)-ß-ManNAc-(1→4)-ß-GlcNAc(O3-α-Gal)-(1→6)-α-GlcNAc(O3-α-Gal, O4-ß-Gal)-(1→]6-12 The genes whose products promote the galactosylation of B. anthracis SCWP are not yet known. We show here that the expression of galE1, encoding a UDP-glucose 4-epimerase necessary for the synthesis of UDP-galactose, is required for B. anthracis SCWP galactosylation. The galE1 mutant assembles surface (S) layer and S layer-associated proteins that associate with ketal-pyruvylated SCWP via their S layer homology domains similarly to wild-type B. anthracis, but the mutant displays a defect in γ-phage murein hydrolase binding to SCWP. Furthermore, deletion of galE1 diminishes the capsulation of B. anthracis with poly-d-γ-glutamic acid (PDGA) and causes a reduction in bacterial virulence. These data suggest that SCWP galactosylation is required for the physiologic assembly of the B. anthracis cell wall envelope and for the pathogenesis of anthrax disease.IMPORTANCE Unlike virulent Bacillus anthracis isolates, B. anthracis strain CDC684 synthesizes secondary cell wall polysaccharide (SCWP) trisaccharide repeats without galactosyl modification, exhibits diminished growth in vitro in broth cultures, and is severely attenuated in an animal model of anthrax. To examine whether SCWP galactosylation is a requirement for anthrax disease, we generated variants of B. anthracis strains Sterne 34F2 and Ames lacking UDP-glucose 4-epimerase by mutating the genes galE1 and galE2 We identified galE1 as necessary for SCWP galactosylation. Deletion of galE1 decreased the poly-d-γ-glutamic acid (PDGA) capsulation of the vegetative form of B. anthracis and increased the bacterial inoculum required to produce lethal disease in mice, indicating that SCWP galactosylation is indeed a determinant of anthrax disease.


Assuntos
Antraz/microbiologia , Bacillus anthracis/metabolismo , Bacillus anthracis/patogenicidade , Proteínas de Bactérias/genética , Galactose/metabolismo , Polissacarídeos Bacterianos/metabolismo , Animais , Bacillus anthracis/genética , Bacillus anthracis/crescimento & desenvolvimento , Proteínas de Bactérias/metabolismo , Divisão Celular , Parede Celular/química , Parede Celular/genética , Parede Celular/fisiologia , Feminino , Galactose/genética , Galactosidases/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Trissacarídeos/química , Trissacarídeos/metabolismo , UDPglucose 4-Epimerase/genética , Uridina Difosfato Galactose/biossíntese , Uridina Difosfato Galactose/metabolismo
6.
PLoS One ; 11(6): e0158293, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27336691

RESUMO

Staphylococcus aureus produces numerous factors that facilitate survival in the human host. S. aureus coagulase (Coa) and von Willebrand factor-binding protein (vWbp) are known to clot plasma through activation of prothrombin and conversion of fibrinogen to fibrin. In addition, S. aureus clumping factor A (ClfA) binds fibrinogen and contributes to platelet aggregation via a fibrinogen- or complement-dependent mechanism. Here, we evaluated the contribution of Coa, vWbp and ClfA to S. aureus pathogenesis in a rabbit model of skin and soft tissue infection. Compared to skin abscesses caused by the Newman wild-type strain, those caused by isogenic coa, vwb, or clfA deletion strains, or a strain deficient in coa and vwb, were significantly smaller following subcutaneous inoculation in rabbits. Unexpectedly, we found that fibrin deposition and abscess capsule formation appear to be independent of S. aureus coagulase activity in the rabbit infection model. Similarities notwithstanding, S. aureus strains deficient in coa and vwb elicited reduced levels of several proinflammatory molecules in human blood in vitro. Although a specific mechanism remains to be determined, we conclude that S. aureus Coa, vWbp and ClfA contribute to abscess formation in rabbits.


Assuntos
Abscesso/microbiologia , Coagulase/metabolismo , Infecções dos Tecidos Moles/microbiologia , Infecções Cutâneas Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidade , Abscesso/patologia , Animais , Proteínas de Bactérias/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Coelhos , Infecções dos Tecidos Moles/patologia , Infecções Cutâneas Estafilocócicas/patologia
7.
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
8.
J Bacteriol ; 197(11): 1854-61, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25777674

RESUMO

UNLABELLED: D-Glutamate is an essential component of bacterial peptidoglycan and a building block of the poly-γ-D-glutamic acid (PDGA) capsule of Bacillus anthracis, the causative agent of anthrax. Earlier work suggested that two glutamate racemases, encoded by racE1 and racE2, are each essential for growth of B. anthracis, supplying D-glutamic acid for the synthesis of peptidoglycan and PDGA capsule. Earlier work could not explain, however, why two enzymes that catalyze the same reaction may be needed for bacterial growth. Here, we report that deletion of racE1 or racE2 did not prevent growth of B. anthracis Sterne (pXO1(+) pXO2(-)), the noncapsulating vaccine strain, or of B. anthracis Ames (pXO1(+) pXO2(+)), a fully virulent, capsulating isolate. While mutants with deletions in racE1 and racE2 were not viable, racE2 deletion delayed vegetative growth of B. anthracis following spore germination and caused aberrant cell shapes, phenotypes that were partially restored by exogenous D-glutamate. Deletion of racE1 or racE2 from B. anthracis Ames did not affect the production or stereochemical composition of the PDGA capsule. A model is presented whereby B. anthracis, similar to Bacillus subtilis, utilizes two functionally redundant racemase enzymes to synthesize D-glutamic acid for peptidoglycan synthesis. IMPORTANCE: Glutamate racemases, enzymes that convert L-glutamate to D-glutamate, are targeted for antibiotic development. Glutamate racemase inhibitors may be useful for the treatment of bacterial infections such as anthrax, where the causative agent, B. anthracis, requires d-glutamate for the synthesis of peptidoglycan and poly-γ-D-glutamic acid (PDGA) capsule. Here we show that B. anthracis possesses two glutamate racemase genes that can be deleted without abolishing either bacterial growth or PDGA synthesis. These data indicate that drug candidates must inhibit both glutamate racemases, RacE1 and RacE2, in order to block B. anthracis growth and achieve therapeutic efficacy.


Assuntos
Isomerases de Aminoácido/genética , Isomerases de Aminoácido/metabolismo , Bacillus anthracis/enzimologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Deleção de Sequência , Bacillus anthracis/genética , Bacillus anthracis/crescimento & desenvolvimento , Deleção de Genes , Ácido Glutâmico/metabolismo , Ácido Poliglutâmico/análogos & derivados , Ácido Poliglutâmico/metabolismo
9.
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
10.
Vaccine ; 33(4): 523-6, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25488332

RESUMO

Staphylococcus aureus is a cause of sepsis and meningitis in very-low-birth-weight (VLBW) infants. Clinical trials with S. aureus specific antibodies failed to protect VLBW neonates, which may be due to the immune evasive attributes of staphylococcal protein A (SpA). Here we show that mouse monoclonal antibody SpAKKAA-mAb 3F6, which neutralizes the immunoglobulin Fcγ-binding and B cell receptor crosslinking attributes of SpA, protects neonatal mice against S. aureus sepsis and raises protective immunity against subsequent staphylococcal infection. We developed a humanized SpAKKAA-mAb that protects neonatal mice against S. aureus sepsis and may therefore be subjected to clinical testing in VLBW neonates.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Infecções Estafilocócicas/prevenção & controle , Proteína Estafilocócica A/imunologia , Staphylococcus aureus/imunologia , Fatores de Virulência/imunologia , Animais , Animais Recém-Nascidos , Anticorpos Antibacterianos/uso terapêutico , Bacteriemia/prevenção & controle , Modelos Animais de Doenças , Meningites Bacterianas/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sobrevida , Resultado do Tratamento
11.
Science ; 342(6160): 863-6, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24233725

RESUMO

Bacterial invasion of host tissues triggers polymorphonuclear leukocytes to release DNA [neutrophil extracellular traps (NETs)], thereby immobilizing microbes for subsequent clearance by innate defenses including macrophage phagocytosis. We report here that Staphylococcus aureus escapes these defenses by converting NETs to deoxyadenosine, which triggers the caspase-3-mediated death of immune cells. Conversion of NETs to deoxyadenosine requires two enzymes, nuclease and adenosine synthase, that are secreted by S. aureus and are necessary for the exclusion of macrophages from staphylococcal abscesses. Thus, the pathogenesis of S. aureus infections has evolved to anticipate host defenses and to repurpose them for the destruction of the immune system.


Assuntos
Apoptose/imunologia , Desoxiadenosinas/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/patogenicidade , Abscesso/imunologia , Abscesso/microbiologia , Animais , Células Cultivadas , Citotoxicidade Imunológica , Desoxirribonucleases/metabolismo , Humanos , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos Endogâmicos BALB C , Staphylococcus aureus/enzimologia
12.
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
13.
J Infect Dis ; 208(1): 92-100, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23532100

RESUMO

Staphylococcus aureus (S. aureus) is a frequent cause of catheter-related infections. S. aureus secretes the coagulases staphylocoagulase and von Willebrand factor-binding protein, both of which form a staphylothrombin complex upon binding to prothrombin. Although fibrinogen and fibrin facilitate the adhesion of S. aureus to catheters, the contribution of staphylothrombin-mediated fibrin has not been examined. In this study, we use a S. aureus mutant lacking both coagulases (Δcoa/vwb) and dabigatran, a pharmacological inhibitor of both staphylothrombin and thrombin, to address this question. Genetic absence or chemical inhibition of pathogen-driven coagulation reduced both fibrin deposition and the retention of S. aureus on catheters in vitro. In a mouse model of jugular vein catheter infection, dabigatran reduced bacterial load on jugular vein catheters, as well as metastatic kidney infection. Importantly, inhibition of staphylothrombin improved the efficacy of vancomycin treatment both in vitro and in the mouse model.


Assuntos
Infecções Relacionadas a Cateter/microbiologia , Fibrina/metabolismo , Infecções Estafilocócicas/microbiologia , Trombina/metabolismo , Animais , Aderência Bacteriana , Carga Bacteriana , Benzimidazóis/farmacologia , Infecções Relacionadas a Cateter/etiologia , Cateteres Venosos Centrais/microbiologia , Coagulase/metabolismo , Coagulase/fisiologia , Dabigatrana , Modelos Animais de Doenças , Veias Jugulares , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Infecções Estafilocócicas/etiologia , Trombina/antagonistas & inibidores , beta-Alanina/análogos & derivados , beta-Alanina/farmacologia
14.
Curr Protoc Microbiol ; Chapter 9: Unit 9C.1, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23408134

RESUMO

Staphylococcus aureus is a facultative anaerobic Gram-positive coccus and a member of the normal skin flora as well as the nasal passages of humans. S. aureus is also the etiological agent of suppurative abscesses, as first described by Sir Alexander Ogston in 1880. Ever since, studies on S. aureus have focused on the complex battery of virulence factors and regulators that allow for its swift transition between commensalism and pathogenic states and escape from host immune defenses. The success of this pathogen is further evidenced by its ability to acquire antibiotic resistance traits through mechanisms that often remain poorly understood.


Assuntos
Técnicas Bacteriológicas/métodos , Staphylococcus aureus/crescimento & desenvolvimento , Contenção de Riscos Biológicos/métodos , Meios de Cultura/química , Preservação Biológica/métodos
15.
Infect Immun ; 81(3): 1008-17, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23319564

RESUMO

Bacillus cereus strains harboring a pXO1-like virulence plasmid cause respiratory anthrax-like disease in humans, particularly in welders. We developed mouse models for intraperitoneal as well as aerosol challenge with spores of B. cereus G9241, harboring pBCXO1 and pBC218 virulence plasmids. Compared to wild-type B. cereus G9241, spores with a deletion of the pBCXO1-carried protective antigen gene (pagA1) were severely attenuated, whereas spores with a deletion of the pBC218-carried protective antigen homologue (pagA2) were not. Anthrax vaccine adsorbed (AVA) immunization raised antibodies that bound and neutralized the pagA1-encoded protective antigen (PA1) but not the PA2 orthologue encoded by pagA2. AVA immunization protected mice against a lethal challenge with spores from B. cereus G9241 or B. cereus Elc4, a strain that had been isolated from a fatal case of anthrax-like disease. As the pathogenesis of B. cereus anthrax-like disease in mice is dependent on pagA1 and PA-neutralizing antibodies provide protection, AVA immunization may also protect humans from respiratory anthrax-like death.


Assuntos
Bacillus cereus/imunologia , Vacinas Bacterianas/imunologia , Infecções por Bactérias Gram-Positivas/prevenção & controle , Infecções Respiratórias/prevenção & controle , Animais , Feminino , Infecções por Bactérias Gram-Positivas/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Infecções Respiratórias/microbiologia
16.
J Bacteriol ; 195(5): 977-89, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23243307

RESUMO

The envelope of Bacillus anthracis encompasses a proteinaceous S-layer with two S-layer proteins (Sap and EA1). Protein assembly in the envelope of B. anthracis requires S-layer homology domains (SLH) within S-layer proteins and S-layer-associated proteins (BSLs), which associate with the secondary cell wall polysaccharide (SCWP), an acetylated carbohydrate that is tethered to peptidoglycan. Here, we investigated the contributions of two putative acetyltransferases, PatA1 and PatA2, on SCWP acetylation and S-layer assembly. We show that mutations in patA1 and patA2 affect the chain lengths of B. anthracis vegetative forms and perturb the deposition of the BslO murein hydrolase at cell division septa. The patA1 and patA2 mutants are defective for the assembly of EA1 in the envelope but retain the ability of S-layer formation with Sap. SCWP isolated from the patA1 patA2 mutant lacked acetyl moieties identified in wild-type polysaccharide and failed to associate with the SLH domains of EA1. A model is discussed whereby patA1- and patA2-mediated acetylation of SCWP enables the deposition of EA1 as well as BslO near the septal region of the B. anthracis envelope.


Assuntos
Acetiltransferases/metabolismo , Bacillus anthracis/enzimologia , Parede Celular/metabolismo , Glicoproteínas de Membrana/biossíntese , Polissacarídeos Bacterianos/metabolismo , Acetilação , Acetiltransferases/genética , Bacillus anthracis/genética , Bacillus anthracis/metabolismo , Bacillus subtilis/enzimologia , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Mutação , N-Acetil-Muramil-L-Alanina Amidase/metabolismo
17.
Infect Immun ; 80(10): 3721-32, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22802349

RESUMO

Staphylococcus aureus is a frequent cause of skin infection and sepsis in humans. Preclinical vaccine studies with S. aureus have used a mouse model with intraperitoneal challenge and survival determination as a measure for efficacy. To appreciate the selection of protective antigens in this model, we sought to characterize the pathological attributes of S. aureus infection in the peritoneal cavity. Testing C57BL/6J and BALB/c mice, >10(9) CFU of S. aureus Newman were needed to produce a lethal outcome in 90% of animals infected via intraperitoneal injection. Both necropsy and histopathology revealed the presence of intraperitoneal abscesses in the vicinity of inoculation sites. Abscesses were comprised of fibrin as well as collagen deposits and immune cells with staphylococci replicating at the center of these lesions. Animals that succumbed to challenge harbored staphylococci in abscess lesions and in blood. The establishment of lethal infections, but not the development of intraperitoneal abscesses, was dependent on S. aureus expression of alpha-hemolysin (Hla). Active immunization with nontoxigenic Hla(H35L) or passive immunization with neutralizing monoclonal antibodies protected mice against early lethal events associated with intraperitoneal S. aureus infection but did not affect the establishment of abscess lesions. These results characterize a mouse model for the study of intraperitoneal abscess formation by S. aureus, a disease that occurs frequently in humans undergoing continuous ambulatory peritoneal dialysis for end-stage renal disease.


Assuntos
Abscesso/microbiologia , Toxinas Bacterianas/toxicidade , Proteínas Hemolisinas/toxicidade , Peritonite/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Animais , Anticorpos Neutralizantes , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Colágeno/metabolismo , Modelos Animais de Doenças , Feminino , Fibrina/metabolismo , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Nefropatias/microbiologia , Nefropatias/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções Estafilocócicas/imunologia
18.
Infect Immun ; 80(10): 3389-98, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22825443

RESUMO

During infection, Staphylococcus aureus secretes two coagulases (Coa and von Willebrand factor binding protein [vWbp]), which, following an association with host prothrombin and fibrinogen, form fibrin clots and enable the establishment of staphylococcal disease. Within the genomes of different S. aureus isolates, coagulase gene sequences are variable, and this has been exploited for a classification of types. We show here that antibodies directed against the variable prothrombin binding portion of coagulases confer type-specific immunity through the neutralization of S. aureus clotting activity and protection from staphylococcal disease in mice. By combining variable portions of coagulases from North American isolates into hybrid Coa and vWbp proteins, a subunit vaccine that provided protection against challenge with different coagulase-type S. aureus strains in mice was derived.


Assuntos
Anticorpos Antibacterianos/imunologia , Coagulase/imunologia , Regulação Bacteriana da Expressão Gênica/imunologia , Regulação Enzimológica da Expressão Gênica/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Animais , Sítios de Ligação , Coagulase/genética , Coagulase/metabolismo , Feminino , Genoma Bacteriano , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Ligação Proteica , Coelhos , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas/imunologia , Staphylococcus aureus/classificação , Staphylococcus aureus/enzimologia , Staphylococcus aureus/genética , Fatores de Tempo
19.
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
20.
J Bacteriol ; 194(15): 3841-50, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22609926

RESUMO

Bacillus anthracis grows in chains of rod-shaped cells, a trait that contributes to its escape from phagocytic clearance in host tissues. Using a genetic approach to search for determinants of B. anthracis chain length, we identified mutants with insertional lesions in secA2. All isolated secA2 mutants exhibited an exaggerated chain length, whereas the dimensions of individual cells were not changed. Complementation studies revealed that slaP (S-layer assembly protein), a gene immediately downstream of secA2 on the B. anthracis chromosome, is also a determinant of chain length. Both secA2 and slaP are required for the efficient secretion of Sap and EA1 (Eag), the two S-layer proteins of B. anthracis, but not for the secretion of S-layer-associated proteins or of other secreted products. S-layer assembly via secA2 and slaP contributes to the proper positioning of BslO, the S-layer-associated protein, and murein hydrolase, which cleaves septal peptidoglycan to separate chains of bacilli. SlaP was found to be both soluble in the bacterial cytoplasm and associated with the membrane. The purification of soluble SlaP from B. anthracis-cleared lysates did not reveal a specific ligand, and the membrane association of SlaP was not dependent on SecA2, Sap, or EA1. We propose that SecA2 and SlaP promote the efficient secretion of S-layer proteins by modifying the general secretory pathway of B. anthracis to transport large amounts of Sap and EA1.


Assuntos
Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Bacillus anthracis/citologia , Bacillus anthracis/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Bacillus anthracis/crescimento & desenvolvimento , Deleção de Genes , Genes Bacterianos , Teste de Complementação Genética , Mutagênese Insercional , Canais de Translocação SEC , Proteínas SecA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...