Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 41(9): 111721, 2022 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-36450248

RESUMO

Influenza infection is substantially worsened by the onset of secondary pneumonia caused by bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA). The bidirectional interaction between the influenza-injured lung microenvironment and MRSA is poorly understood. By conditioning MRSA ex vivo in bronchoalveolar lavage fluid collected from mice at various time points of influenza infection, we found that the influenza-injured lung microenvironment dynamically induces MRSA to increase cytotoxin expression while decreasing metabolic pathways. LukAB, a SaeRS two-component system-dependent cytotoxin, is particularly important to the severity of post-influenza MRSA pneumonia. LukAB's activity is likely shaped by the post-influenza lung microenvironment, as LukAB binds to (and is activated by) heparan sulfate (HS) oligosaccharide sequences shed from the epithelial glycocalyx after influenza. Our findings indicate that post-influenza MRSA pneumonia is shaped by bidirectional host-pathogen interactions: host injury triggers changes in bacterial expression of toxins, the activity of which may be shaped by host-derived HS fragments.


Assuntos
Coinfecção , Influenza Humana , Staphylococcus aureus Resistente à Meticilina , Pneumonia Bacteriana , Animais , Camundongos , Humanos , Influenza Humana/complicações , Virulência , Pneumonia Bacteriana/complicações , Citotoxinas , Heparitina Sulfato , Pulmão
2.
Nat Microbiol ; 6(6): 731-745, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33875847

RESUMO

Staphylococcus aureus has evolved into diverse lineages, known as clonal complexes (CCs), which exhibit differences in the coding sequences of core virulence factors. Whether these alterations affect functionality is poorly understood. Here, we studied the highly polymorphic pore-forming toxin LukAB. We discovered that the LukAB toxin variants produced by S. aureus CC30 and CC45 kill human phagocytes regardless of whether CD11b, the previously established LukAB receptor, is present, and instead target the human hydrogen voltage-gated channel 1 (HVCN1). Biochemical studies identified the domain within human HVCN1 that drives LukAB species specificity, enabling the generation of humanized HVCN1 mice with enhanced susceptibility to CC30 LukAB and to bloodstream infection caused by CC30 S. aureus strains. Together, this work advances our understanding of an important S. aureus toxin and underscores the importance of considering genetic variation in characterizing virulence factors and understanding the tug of war between pathogens and the host.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Canais Iônicos/metabolismo , Leucocidinas/genética , Leucocidinas/metabolismo , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/metabolismo , Animais , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Variação Genética , Humanos , Canais Iônicos/genética , Camundongos Endogâmicos C57BL , Fagócitos/metabolismo , Fagócitos/microbiologia , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética
3.
Sci Adv ; 6(21): eaaz4926, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32494740

RESUMO

Cholesterol-dependent cytolysins (CDCs) form pores in cholesterol-rich membranes, but cholesterol alone is insufficient to explain their cell and host tropism. Here, we show that all eight major CDCs have high-affinity lectin activity that identifies glycans as candidate cellular receptors. Streptolysin O, vaginolysin, and perfringolysin O bind multiple glycans, while pneumolysin, lectinolysin, and listeriolysin O recognize a single glycan class. Addition of exogenous carbohydrate receptors for each CDC inhibits toxin activity. We present a structure for suilysin domain 4 in complex with two distinct glycan receptors, P1 antigen and αGal/Galili. We report a wide range of binding affinities for cholesterol and for the cholesterol analog pregnenolone sulfate and show that CDCs bind glycans and cholesterol independently. Intermedilysin binds to the sialyl-TF O-glycan on its erythrocyte receptor, CD59. Removing sialyl-TF from CD59 reduces intermedilysin binding. Glycan-lectin interactions underpin the cellular tropism of CDCs and provide molecular targets to block their cytotoxic activity.


Assuntos
Colesterol , Citotoxinas , Colesterol/metabolismo , Citotoxinas/química , Citotoxinas/farmacologia , Lectinas , Polissacarídeos , Receptores de Superfície Celular
4.
Sci Transl Med ; 11(475)2019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30651319

RESUMO

A key aspect underlying the severity of infections caused by Staphylococcus aureus is the abundance of virulence factors that the pathogen uses to thwart critical components of the human immune response. One such mechanism involves the destruction of host immune cells by cytolytic toxins secreted by S. aureus, including five bicomponent leukocidins: PVL, HlgAB, HlgCB, LukED, and LukAB. Purified leukocidins can lyse immune cells ex vivo, and systemic injections of purified LukED or HlgAB can acutely kill mice. Here, we describe the generation and characterization of centyrins that bind S. aureus leukocidins with high affinity and protect primary human immune cells from toxin-mediated cytolysis. Centyrins are small protein scaffolds derived from the fibronectin type III-binding domain of the human protein tenascin-C. Although centyrins are potent in tissue culture assays, their short serum half-lives limit their efficacies in vivo. By extending the serum half-lives of centyrins through their fusion to an albumin-binding consensus domain, we demonstrate the in vivo efficacy of these biologics in a murine intoxication model and in models of both prophylactic and therapeutic treatment of live S. aureus systemic infections. These biologics that target S. aureus virulence factors have potential for treating and preventing serious staphylococcal infections.


Assuntos
Fatores Biológicos/farmacologia , Leucocidinas/metabolismo , Testes de Neutralização , Staphylococcus aureus/metabolismo , Sequência de Aminoácidos , Animais , Citoproteção/efeitos dos fármacos , Citotoxicidade Imunológica , Hemólise/efeitos dos fármacos , Humanos , Leucocidinas/química , Camundongos , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Fagócitos/efeitos dos fármacos , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/efeitos dos fármacos
5.
J Infect Dis ; 215(7): 1124-1131, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28186295

RESUMO

The 2-component leukotoxin LukAB is critical for Staphylococcus aureus targeting and killing of human neutrophils ex vivo and is produced in the setting of human infection. We report 3 LukAB-specific human monoclonal antibodies (mAbs) with distinct mechanisms of toxin neutralization and in vivo efficacy. Three hybridomas secreting mAbs with anti-LukAB activity (designated SA-13, -15, and -17) were generated from B cells obtained from a 12-year-old boy with S. aureus osteomyelitis. Each of the 3 mAbs neutralized LukAB-mediated neutrophil toxicity, exhibited differing levels of potency, recognized different antigenic sites on the toxin, and displayed at least 2 distinct mechanisms for cytotoxic inhibition. SA-15 bound exclusively to the dimeric form of the toxin, suggesting that human B cells recognize epitopes on the dimerized form of LukAB during natural infection. Both SA-13 and SA-17 bound the LukA monomer and the LukAB dimer. Although all 3 mAbs potently neutralized cytotoxicity, only SA-15 and SA-17 significantly inhibited toxin association with the cell surface. Treatment with a 1:1 mixture of mAbs SA-15 and SA-17 resulted in significantly lower bacterial colony counts in heart, liver, and kidneys in a murine model of S. aureus sepsis. These data describe the isolation of diverse and efficacious antitoxin mAbs.


Assuntos
Anticorpos Antibacterianos/sangue , Anticorpos Monoclonais/sangue , Proteínas de Bactérias/imunologia , Leucocidinas/imunologia , Neutrófilos/imunologia , Infecções Estafilocócicas/microbiologia , Animais , Linfócitos B/imunologia , Criança , Feminino , Humanos , Hibridomas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Análise de Regressão , Staphylococcus aureus
6.
Cell Host Microbe ; 18(3): 363-70, 2015 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-26320997

RESUMO

In order for Staphylococcus aureus to thrive inside the mammalian host, the bacterium has to overcome iron scarcity. S. aureus is thought to produce toxins that lyse erythrocytes, releasing hemoglobin, the most abundant iron source in mammals. Here we identify the Duffy antigen receptor for chemokines (DARC) as the receptor for the S. aureus hemolytic leukocidins LukED and HlgAB. By assessing human erythrocytes with DARC polymorphisms, we determined that HlgAB- and LukED-mediated lysis directly relates to DARC expression. DARC is required for S. aureus-mediated lysis of human erythrocytes, and DARC overexpression is sufficient to render cells susceptible to toxin-mediated lysis. HlgA and LukE bind directly to DARC through different regions, and by targeting DARC, HlgAB and LukED support S. aureus growth in a hemoglobin-acquisition-dependent manner. These findings elucidate how S. aureus targets and lyses erythrocytes to release one of the scarcest nutrients within the mammalian host.


Assuntos
Sistema do Grupo Sanguíneo Duffy/metabolismo , Eritrócitos/efeitos dos fármacos , Hemólise , Interações Hospedeiro-Patógeno , Leucocidinas/metabolismo , Receptores de Superfície Celular/metabolismo , Staphylococcus aureus/metabolismo , Humanos , Ligação Proteica
7.
Mol Microbiol ; 96(2): 325-48, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25598361

RESUMO

Bacillus subtilis and its closest relatives have multiple rap-phr quorum sensing gene pairs that coordinate a variety of physiological processes with population density. Extra-chromosomal rap-phr genes are also present on mobile genetic elements, yet relatively little is known about their function. In this work, we demonstrate that Rap60-Phr60 from plasmid pTA1060 coordinates a variety of biological processes with population density including sporulation, cannibalism, biofilm formation and genetic competence. Similar to other Rap proteins that control sporulation, Rap60 modulates phosphorylation of the transcription factor Spo0A by acting as a phosphatase of Spo0F∼P, an intermediate of the sporulation phosphorelay system. Additionally, Rap60 plays a noncanonical role in regulating the autophosphorylation of the sporulation-specific kinase KinA, a novel activity for Rap proteins. In contrast, Rap proteins that modulate genetic competence interfere with DNA binding by the transcription factor ComA. Rap60 regulates the activity of ComA in a unique manner by forming a Rap60-ComA-DNA ternary complex that inhibits transcription of target genes. Taken together, this work provides new insight into two novel mechanisms of regulating Spo0A and ComA by Rap60 and expands our general understanding of how plasmid-encoded quorum sensing pairs regulate important biological processes.


Assuntos
Bacillus subtilis/fisiologia , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Bacteriana da Expressão Gênica , Plasmídeos/genética , Percepção de Quorum , Fatores de Transcrição/metabolismo , Bacillus subtilis/genética , Proteínas de Bactérias/genética , Proteínas de Ligação a DNA/genética , Plasmídeos/metabolismo , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...