Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Sex Med ; 19(6): 1024-1031, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35414488

RESUMO

BACKGROUND: Delayed infection, thought to be due to gradual biofilm formation, remains a feared complication after inflatable penile prosthesis (IPP) insertion. Understanding and preventing biofilm formation is necessary to prevent infections. AIM: To develop an in vitro model and compare growth of biofilm by different bacteria on IPPs and evaluate the anti-infective efficacy of the Coloplast Titan and AMS 700 InhibiZone. METHODS: Sterile IPPs (Coloplast) were cut into rings and incubated with S. epidermidis, S. aureus, P. aeruginosa, A. baumannii, or K. pneumoniae cultures in tryptic soy broth (TSB) (4 hour) to ensure adequate bacteria attachment, and then in only TSB (120 hours) to allow for biofilm formation. Rings were fixed with ethanol and biofilm measured by spectrophotometer (OD570) after crystal violet staining. This methodology was repeated for S. epidermidis and P. aeruginosa with Coloplast rings dipped in 10 ml of a 10 mg/ml Rifampin, 1 mg/ml Gentamicin, and deionized water solution and undipped AMS InhibiZone rings. Crystal violet assay (OD570) was repeated after incubation within bacteria (2 hour), and then only TSB (120 hours). OUTCOMES: The primary outcome of the study was OD570 readings, indirectly measuring biofilm mass on implant rings. RESULTS: S. epidermidis, S. aureus, A. baumannii, P. aeruginosa, and K. pneumoniae all formed significant biofilm. P. aeruginosa showed the strongest predilection to grow biofilm on IPPs. P. aeruginosa also formed significant biofilm on antibiotic-treated Coloplast and AMS rings, while S. epidermidis was inhibited. No significant difference was found in biofilm inhibition between the implants. CLINICAL TRANSLATION: Our findings suggest gram-negative bacteria may form biofilm more proficiently and quickly on IPPs than gram-positive organisms. Commonly used antibiotic treatments on IPPs may be effective against S. epidermidis but not against P. aeruginosa biofilm formation. STRENGTHS & LIMITATIONS: This is the first study comparing biofilm formation by different bacteria organisms on IPPs and the inhibitive ability of Coloplast and AMS implants against biofilm formation. Clinical data on organisms responsible for infected IPPs is needed to determine the clinical relevance of our findings. CONCLUSION: Our novel in vitro model of biofilm formation of IPPs evaluated the effect of a gentamicin/rifampin antibiotic dip on Coloplast Titan implants and the anti-infective capacity of the minocycline/rifampin precoated AMS 700 InhibiZone against S. epidermidis and P. aeruginosa. P. aeruginosa was able to grow on both antibiotic-treated implants, with no significant difference, and should continue to be a specific target of investigation to reduce delayed post-operative IPP infections. Narasimman M, Ory J, Bartra SS, et al. Evaluation of Bacteria in a Novel In Vitro Biofilm Model of Penile Prosthesis. J Sex Med 2022;19:1024-1031.


Assuntos
Prótese de Pênis , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Biofilmes , Gentamicinas/farmacologia , Violeta Genciana , Humanos , Rifampina/uso terapêutico , Staphylococcus aureus
2.
J Immunol ; 204(8): 2242-2256, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32161097

RESUMO

Type I IFNs play a complex role in determining the fate of microbial pathogens and may also be deleterious to the host during bacterial and viral infections. Upon ligand binding, a receptor proximal complex consisting of IFN-α and -ß receptors 1 and 2 (IFNAR1, IFNAR2, respectively), tyrosine kinase 2 (Tyk2), Jak1, and STAT2 are assembled and promote the phosphorylation of STAT1 and STAT2. However, how the IFNARs proximal complex is assembled upon binding to IFN is poorly understood. In this study, we show that the membrane-associated pore-forming protein Perforin-2 (P2) is critical for LPS-induced endotoxic shock in wild-type mice. Type I IFN-mediated JAK-STAT signaling is severely impaired, and activation of MAPKs and PI3K signaling pathways are delayed in P2-deficient mouse bone marrow-derived macrophages, mouse embryonic fibroblasts (MEFs), and human HeLa cells upon IFN stimulation. The P2 N-glycosylated extracellular membrane attack complex/perforin domain and the P2 domain independently associate with the extracellular regions of IFNAR1 and IFNAR2, respectively, in resting MEFs. In addition, the P2 cytoplasmic tail domain mediated the constitutive interaction between STAT2 and IFNAR2 in resting MEFs, an interaction that is dependent on the association of the extracellular regions of P2 and IFNAR2. Finally, the constitutive association of P2 with both receptors and STAT2 is critical for the receptor proximal complex assembly and reciprocal transphosphorylation of Jak1 and Tyk2 as well as the phosphorylation and activation of STAT1 and STAT2 upon IFN-ß stimulation.


Assuntos
Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Transdução de Sinais/imunologia , Animais , Células Cultivadas , Células HeLa , Humanos , Lipopolissacarídeos , Camundongos , Camundongos Knockout , Choque Séptico/induzido quimicamente , Choque Séptico/imunologia
3.
Biophys J ; 120(3): 453-462, 2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33359463

RESUMO

Understanding microbe-host interactions at the molecular level is a major goal of fundamental biology and therapeutic drug development. Structural biology strives to capture biomolecular structures in action, but the samples are often highly simplified versions of the complex native environment. Here, we present an Escherichia coli model system that allows us to probe the structure and function of Ail, the major surface protein of the deadly pathogen Yersinia pestis. We show that cell surface expression of Ail produces Y. pestis virulence phenotypes in E. coli, including resistance to human serum, cosedimentation of human vitronectin, and pellicle formation. Moreover, isolated bacterial cell envelopes, encompassing inner and outer membranes, yield high-resolution solid-state NMR spectra that reflect the structure of Ail and reveal Ail sites that are sensitive to the bacterial membrane environment and involved in the interactions with human serum components. The data capture the structure and function of Ail in a bacterial outer membrane and set the stage for probing its interactions with the complex milieu of immune response proteins present in human serum.


Assuntos
Yersinia pestis , Proteínas da Membrana Bacteriana Externa , Escherichia coli , Humanos , Virulência , Fatores de Virulência
4.
Mol Microbiol ; 114(3): 510-520, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32462782

RESUMO

The outer membrane is a key virulence determinant of gram-negative bacteria. In Yersinia pestis, the deadly agent that causes plague, the protein Ail and lipopolysaccharide (LPS)6 enhance lethality by promoting resistance to human innate immunity and antibiotics, enabling bacteria to proliferate in the human host. Their functions are highly coordinated. Here we describe how they cooperate to promote pathogenesis. Using a multidisciplinary approach, we identify mutually constructive interactions between Ail and LPS that produce an extended conformation of Ail at the membrane surface, cause thickening and rigidification of the LPS membrane, and collectively promote Y. pestis survival in human serum, antibiotic resistance, and cell envelope integrity. The results highlight the importance of the Ail-LPS assembly as an organized whole, rather than its individual components, and provide a handle for targeting Y. pestis pathogenesis.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/metabolismo , Fatores de Virulência/imunologia , Fatores de Virulência/metabolismo , Yersinia pestis/imunologia , Yersinia pestis/metabolismo , Motivos de Aminoácidos , Antibacterianos/farmacologia , Farmacorresistência Bacteriana , Humanos , Testes de Sensibilidade Microbiana , Simulação de Dinâmica Molecular , Mutação , Peste/imunologia , Peste/microbiologia , Ligação Proteica , Conformação Proteica , Yersinia pestis/efeitos dos fármacos
5.
PLoS Pathog ; 15(12): e1008001, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31869388

RESUMO

The enteropathogen Yersinia pseudotuberculosis and the related plague agent Y. pestis require the Ysc type III secretion system (T3SS) to subvert phagocyte defense mechanisms and cause disease. Yet type III secretion (T3S) in Yersinia induces growth arrest and innate immune recognition, necessitating tight regulation of the T3SS. Here we show that Y. pseudotuberculosis T3SS expression is kept low under anaerobic, iron-rich conditions, such as those found in the intestinal lumen where the Yersinia T3SS is not required for growth. In contrast, the Yersinia T3SS is expressed under aerobic or anaerobic, iron-poor conditions, such as those encountered by Yersinia once they cross the epithelial barrier and encounter phagocytic cells. We further show that the [2Fe-2S] containing transcription factor, IscR, mediates this oxygen and iron regulation of the T3SS by controlling transcription of the T3SS master regulator LcrF. IscR binds directly to the lcrF promoter and, importantly, a mutation that prevents this binding leads to decreased disseminated infection of Y. pseudotuberculosis but does not perturb intestinal colonization. Similar to E. coli, Y. pseudotuberculosis uses the Fe-S cluster occupancy of IscR as a readout of oxygen and iron conditions that impact cellular Fe-S cluster homeostasis. We propose that Y. pseudotuberculosis has coopted this system to sense entry into deeper tissues and induce T3S where it is required for virulence. The IscR binding site in the lcrF promoter is completely conserved between Y. pseudotuberculosis and Y. pestis. Deletion of iscR in Y. pestis leads to drastic disruption of T3S, suggesting that IscR control of the T3SS evolved before Y. pestis split from Y. pseudotuberculosis.


Assuntos
Ferro/metabolismo , Oxigênio/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Infecções por Yersinia pseudotuberculosis/imunologia , Animais , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica/genética , Regiões Promotoras Genéticas/genética , Transcrição Gênica , Yersinia/metabolismo , Yersinia pseudotuberculosis/patogenicidade , Infecções por Yersinia pseudotuberculosis/metabolismo
6.
Microb Pathog ; 154: 104852, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33762201

RESUMO

Cellular Perforin-2 (MPEG1) is a pore-forming MACPF family protein that plays a critical role in the defense against bacterial pathogens. Macrophages, neutrophils, and several other cell types that are part of the front line of innate defenses constitutively express high levels of Perforin-2; whereas, most other cell types must be induced to express Perforin-2 by interferons (α, ß and γ) and/or PAMPs such as LPS. In this study, we demonstrate that many bacterial pathogens can limit the expression of Perforin-2 in cells normally inducible for Perforin-2 expression, while ordinarily commensal or non-pathogenic bacteria triggered high levels of Perforin-2 expression in these same cell types. The mechanisms by which pathogens suppress Perforin-2 expression was explored further using Salmonella enterica serovar Typhimurium and cultured MEFs as well as intestinal epithelial cell lines. These studies identified multiple factors required to minimize the expression of Perforin-2 in cell types inducible for Perforin-2 expression. These included the PmrAB and PhoPQ two-component systems, select LPS modification enzymes and the Type III secretion effector protein AvrA.


Assuntos
Lipopolissacarídeos , Salmonella typhimurium , Proteínas de Bactérias/genética , Células Epiteliais , Fibroblastos , Perforina/genética , Sorogrupo
7.
J Bacteriol ; 202(15)2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32424009

RESUMO

Chlamydia trachomatis Scc4 (formerly CT663) engages the transcription machinery and the pathogenic type III secretion system (T3SS). Both machines are required for Chlamydia infection. These requirements and the limited ability for genetic manipulation in Chlamydia have hampered dissection of Scc4's contributions. Here, by developing bacterial systems that permit the controlled expression and stable maintenance of Scc4, we assess Scc4's effects on chlamydial growth phenotype, secretion, and the patterns of T3SS gene expression. Expressing Scc4 in Escherichia coli lacking a T3SS injectisome causes a growth defect. This deficiency is rescued by overexpressing the ß-subunit of RNA polymerase (RNAP) or by exploiting sigma 70 (σ70) (homologous to chlamydial σ66) mutants that strengthen the interaction between σ70 region 4 and the ß-flap, confirming Scc4's distinction as a module of RNAP holoenzyme capable of modulating transcription. Yersinia pestis expressing Scc4 sustains a functional T3SS, through which CopN secretion is boosted by cooption of Scc4 and Scc1. Finally, conditional expression of Scc4 in C. trachomatis results in fast expansion of the Chlamydia-containing vacuole and accelerated chlamydial development, coupled to selective up- or downregulation of gene expression from different T3SS genes. This work reveals, for the first time, the context-dependent action of Scc4 linking it to diverse protein networks in bacteria. It establishes that Scc4, when overexpressed, exerts incredible effects on chlamydial development by reinforcing control of the T3SS.IMPORTANCE The T3SS is a key virulence factor required for C. trachomatis infection. The control of the T3SS has not been well studied in this obligate intracellular pathogen. Here, we show that Scc4 plays a major role for precise control of the pathogenic T3SS at the levels of gene expression and effector secretion through genetically separable protein networks, allowing a fast adaptive mode of C. trachomatis development during infection in human epithelial cells.


Assuntos
Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Proteínas de Bactérias/genética , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/genética , Chlamydia trachomatis/crescimento & desenvolvimento , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Regulação Bacteriana da Expressão Gênica , Humanos , Transporte Proteico , Fator sigma/genética , Fator sigma/metabolismo , Sistemas de Secreção Tipo III/genética
8.
Microb Pathog ; 141: 103993, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31988008

RESUMO

Yersinia pestis, a Gram-negative bacterium, is the etiologic agent of plague. A hallmark of Y. pestis infection is the organism's ability to rapidly disseminate through an animal host. Y. pestis expresses the outer membrane protein, Ail (Attachment invasion locus), which is associated with host invasion and serum resistance. However, whether Ail plays a role in host dissemination remains unclear. In this study, C57BL/6J mice were challenged with a defined Y. pestis strain, KimD27, or an isogenic ail-deleted mutant derived from KimD27 via metacarpal paw pad inoculation, nasal drops, orogastric infection, or tail vein injection to mimic bubonic, pneumonic, oral, or septicemic plague, respectively. Our results showed that ail-deleted Y. pestis KimD27 lost the ability to invade host cells, leading to failed host dissemination in the pneumonic and oral plague models but not in the bubonic or septicemic plague models, which do not require invasiveness. Therefore, this study demonstrated that whether Ail plays a role in Y. pestis pathogenesis depends on the infection route.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Peste/microbiologia , Fatores de Virulência/metabolismo , Virulência , Yersinia pestis , Animais , Proteínas de Bactérias/metabolismo , Modelos Animais de Doenças , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Boca/microbiologia , Yersinia pestis/metabolismo , Yersinia pestis/patogenicidade
9.
J Immunol ; 201(9): 2710-2720, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30249808

RESUMO

Perforin-2, the product of the MPEG1 gene, limits the spread and dissemination of bacterial pathogens in vivo. It is highly expressed in murine and human phagocytes, and macrophages lacking Perforin-2 are compromised in their ability to kill phagocytosed bacteria. In this study, we used Salmonella enterica serovar Typhimurium as a model intracellular pathogen to elucidate the mechanism of Perforin-2's bactericidal activity. In vitro Perforin-2 was found to facilitate the degradation of Ags contained within the envelope of phagocytosed bacteria. In contrast, degradation of a representative surface Ag was found to be independent of Perforin-2. Consistent with our in vitro results, a protease-sensitive, periplasmic superoxide dismutase (SodCII) contributed to the virulence of S. Typhimurium in Perforin-2 knockout but not wild-type mice. In aggregate, our studies indicate that Perforin-2 breaches the envelope of phagocytosed bacteria, facilitating the delivery of proteases and other antimicrobial effectors to sites within the bacterial cell.


Assuntos
Proteínas Citotóxicas Formadoras de Poros/imunologia , Salmonelose Animal/imunologia , Animais , Parede Celular , Camundongos , Camundongos Knockout , Fagocitose/imunologia , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Salmonelose Animal/metabolismo , Salmonella typhimurium
10.
Microbiology (Reading) ; 164(3): 338-348, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29458689

RESUMO

The needle structures of type III secretion (T3S) systems are formed by the secretion and polymerization of a needle subunit protein, YscF in Yersinia pestis. A subset of T3S systems employ unique heterodimeric chaperones, YscE and YscG in Y. pestis, to prevent the polymerization of needle subunits within the bacterial cell. We demonstrate that the YscE/YscG chaperone is also required for stable YscF expression and for secretion of YscF. Overexpression of a functional maltose-binding protein (MBP)-YscG hybrid protein stabilized cytoplasmic YscF but YscF was not secreted in the absence of YscE. Furthermore, a YscE mutant protein was identified that functioned with YscG to stabilize cytosolic YscF; however, YscF was not secreted. These findings confirm a role for the YscE/YscG chaperone in YscF secretion and suggest that YscE may have a specific role in this process. Recent studies have shown that YscF deleted of its N-terminal 15 residues is still secreted and functional, suggesting that YscF may not require an N-terminal secretion signal. However, we demonstrate that YscF contains an N-terminal secretion signal and that a functional N-terminal signal is required for YscF secretion.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Sinais Direcionadores de Proteínas/genética , Sistemas de Secreção Tipo III/metabolismo , Yersinia pestis/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Citoplasma/metabolismo , Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Proteínas de Membrana/genética , Chaperonas Moleculares/genética , Mutação , Ligação Proteica , Multimerização Proteica , Yersinia pestis/genética
11.
Biochim Biophys Acta ; 1848(2): 712-20, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25433311

RESUMO

The surrounding environment has significant consequences for the structural and functional properties of membrane proteins. While native structure and function can be reconstituted in lipid bilayer membranes, the detergents used for protein solubilization are not always compatible with biological activity and, hence, not always appropriate for direct detection of ligand binding by NMR spectroscopy. Here we describe how the sample environment affects the activity of the outer membrane protein Ail (attachment invasion locus) from Yersinia pestis. Although Ail adopts the correct ß-barrel fold in micelles, the high detergent concentrations required for NMR structural studies are not compatible with the ligand binding functionality of the protein. We also describe preparations of Ail embedded in phospholipid bilayer nanodiscs, optimized for NMR studies and ligand binding activity assays. Ail in nanodiscs is capable of binding its human ligand fibronectin and also yields high quality NMR spectra that reflect the proper fold. Binding activity assays, developed to be performed directly with the NMR samples, show that ligand binding involves the extracellular loops of Ail. The data show that even when detergent micelles support the protein fold, detergents can interfere with activity in subtle ways.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Fibronectinas/química , Lipídeos de Membrana/química , Fatores de Virulência/química , Yersinia pestis/química , Sequência de Aminoácidos , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Dimiristoilfosfatidilcolina/química , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Glicolipídeos/química , Humanos , Fosfatos de Inositol/química , Ligantes , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Nanoestruturas/química , Fosfatidilgliceróis/química , Éteres Fosfolipídicos/química , Fosforilcolina/análogos & derivados , Fosforilcolina/química , Dobramento de Proteína , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
12.
PLoS Pathog ; 10(2): e1003954, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24586162

RESUMO

Chlamydia trachomatis, the causative agent of trachoma and sexually transmitted infections, employs a type III secretion (T3S) system to deliver effector proteins into host epithelial cells to establish a replicative vacuole. Aside from the phosphoprotein TARP, a Chlamydia effector that promotes actin re-arrangements, very few factors mediating bacterial entry and early inclusion establishment have been characterized. Like many T3S effectors, TARP requires a chaperone (Slc1) for efficient translocation into host cells. In this study, we defined proteins that associate with Slc1 in invasive C. trachomatis elementary bodies (EB) by immunoprecipitation coupled with mass spectrometry. We identified Ct875, a new Slc1 client protein and T3S effector, which we renamed TepP (Translocated early phosphoprotein). We provide evidence that T3S effectors form large molecular weight complexes with Scl1 in vitro and that Slc1 enhances their T3S-dependent secretion in a heterologous Yersinia T3S system. We demonstrate that TepP is translocated early during bacterial entry into epithelial cells and is phosphorylated at tyrosine residues by host kinases. However, TepP phosphorylation occurs later than TARP, which together with the finding that Slc1 preferentially engages TARP in EBs leads us to postulate that these effectors are translocated into the host cell at different stages during C. trachomatis invasion. TepP co-immunoprecipitated with the scaffolding proteins CrkI-II during infection and Crk was recruited to EBs at entry sites where it remained associated with nascent inclusions. Importantly, C. trachomatis mutants lacking TepP failed to recruit CrkI-II to inclusions, providing genetic confirmation of a direct role for this effector in the recruitment of a host factor. Finally, endocervical epithelial cells infected with a tepP mutant showed altered expression of a subset of genes associated with innate immune responses. We propose a model wherein TepP acts downstream of TARP to recruit scaffolding proteins at entry sites to initiate and amplify signaling cascades important for the regulation of innate immune responses to Chlamydia.


Assuntos
Chlamydia trachomatis/genética , Imunidade Inata/genética , Chaperonas Moleculares/genética , Proteínas Proto-Oncogênicas c-crk/metabolismo , Sequência de Aminoácidos , Chlamydia trachomatis/imunologia , Chlamydia trachomatis/metabolismo , Cromatografia Líquida , Imunofluorescência , Células HeLa , Humanos , Imunidade Inata/imunologia , Imunoprecipitação , Chaperonas Moleculares/imunologia , Chaperonas Moleculares/metabolismo , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Espectrometria de Massas em Tandem
13.
Microbiology (Reading) ; 161(11): 2174-2183, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26377177

RESUMO

Yersinia pestis, the agent of plague, requires the Ail (attachment invasion locus) outer membrane protein to survive in the blood and tissues of its mammalian hosts. Ail is important for both attachment to host cells and for resistance to complement-dependent bacteriolysis. Previous studies have shown that Ail interacts with components of the extracellular matrix, including fibronectin, laminin and heparan sulfate proteoglycans, and with the complement inhibitor C4b-binding protein. Here, we demonstrate that Ail-expressing Y. pestis strains bind vitronectin - a host protein with functions in cell attachment, fibrinolysis and inhibition of the complement system. The Ail-dependent recruitment of vitronectin resulted in efficient cleavage of vitronectin by the outer membrane Pla (plasminogen activator protease). Escherichia coli DH5α expressing Y. pestis Ail bound vitronectin, but not heat-treated vitronectin. The ability of Ail to directly bind vitronectin was demonstrated by ELISA using purified refolded Ail in nanodiscs.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Interações Hospedeiro-Patógeno , Fatores de Virulência/metabolismo , Vitronectina/metabolismo , Yersinia pestis/metabolismo , Proteínas de Bactérias/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Hidrólise , Ativadores de Plasminogênio/metabolismo , Ligação Proteica
14.
Immunol Cell Biol ; 93(9): 815-24, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25829141

RESUMO

Yersinia pestis is a Gram-negative bacterium that causes plague. After Y. pestis overcomes the skin barrier, it encounters antigen-presenting cells (APCs), such as Langerhans and dendritic cells. They transport the bacteria from the skin to the lymph nodes. However, the molecular mechanisms involved in bacterial transmission are unclear. Langerhans cells (LCs) express Langerin (CD207), a calcium-dependent (C-type) lectin. Furthermore, Y. pestis possesses exposed core oligosaccharides. In this study, we show that Y. pestis invades LCs and Langerin-expressing transfectants. However, when the bacterial core oligosaccharides are shielded or truncated, Y. pestis propensity to invade Langerhans and Langerin-expressing cells decreases. Moreover, the interaction of Y. pestis with Langerin-expressing transfectants is inhibited by purified Langerin, a DC-SIGN (DC-specific intercellular adhesion molecule 3 grabbing nonintegrin)-like molecule, an anti-CD207 antibody, purified core oligosaccharides and several oligosaccharides. Furthermore, covering core oligosaccharides reduces the mortality associated with murine infection by adversely affecting the transmission of Y. pestis to lymph nodes. These results demonstrate that direct interaction of core oligosaccharides with Langerin facilitates the invasion of LCs by Y. pestis. Therefore, Langerin-mediated binding of Y. pestis to APCs may promote its dissemination and infection.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Antígenos CD/imunologia , Lectinas Tipo C/imunologia , Lectinas de Ligação a Manose/imunologia , Fagocitose/imunologia , Yersinia pestis/imunologia , Animais , Células Apresentadoras de Antígenos/microbiologia , Antígenos CD/metabolismo , Aderência Bacteriana/imunologia , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Citometria de Fluxo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Células de Langerhans/imunologia , Células de Langerhans/metabolismo , Lectinas Tipo C/metabolismo , Lectinas de Ligação a Manose/metabolismo , Camundongos , Antígenos O/imunologia , Antígenos O/metabolismo , Peste/imunologia , Peste/microbiologia , Ligação Proteica/imunologia , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Análise de Sobrevida , Yersinia pestis/metabolismo , Yersinia pestis/fisiologia
15.
J Bacteriol ; 195(4): 777-87, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23222719

RESUMO

LcrV, the type III needle cap protein of pathogenic Yersinia, has been proposed to function as a tether between YscF, the needle protein, and YopB-YopD to constitute the injectisome, a conduit for the translocation of effector proteins into host cells. Further, insertion of LcrV-capped needles from a calcium-rich environment into host cells may trigger the low-calcium signal for effector translocation. Here, we used a genetic approach to test the hypothesis that the needle cap responds to the low-calcium signal by promoting injectisome assembly. Growth restriction of Yersinia pestis in the absence of calcium (low-calcium response [LCR(+)] phenotype) was exploited to isolate dominant negative lcrV alleles with missense mutations in its amber stop codon (lcrV(*327)). The addition of at least four amino acids or the eight-residue Strep tag to the C terminus was sufficient to generate an LCR(-) phenotype, with variant LcrV capping type III needles that cannot assemble the YopD injectisome component. The C-terminal Strep tag appears buried within the cap structure, blocking effector transport even in Y. pestis yscF variants that are otherwise calcium blind, a constitutive type III secretion phenotype. Thus, LcrV(*327) mutants arrest the needle cap in a state in which it cannot respond to the low-calcium signal with either injectisome assembly or the activation of type III secretion. Insertion of the Strep tag at other positions of LcrV produced variants with wild-type LCR(+), LCR(-), or dominant negative LCR(-) phenotypes, thereby allowing us to identify discrete sites within LcrV as essential for its attributes as a secretion substrate, needle cap, and injectisome assembly factor.


Assuntos
Antígenos de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Yersinia enterocolitica/metabolismo , Yersinia pestis/metabolismo , Sequência de Aminoácidos , Antígenos de Bactérias/genética , Técnicas Bacteriológicas , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Insercional , Mutação , Proteínas Citotóxicas Formadoras de Poros/genética , Conformação Proteica , Fatores de Tempo , Yersinia enterocolitica/genética , Yersinia enterocolitica/crescimento & desenvolvimento , Yersinia pestis/genética , Yersinia pestis/crescimento & desenvolvimento
16.
Infect Immun ; 81(9): 3163-72, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23774602

RESUMO

Na(+)/H(+) antiporters are ubiquitous membrane proteins that play a central role in the ion homeostasis of cells. In this study, we examined the possible role of Na(+)/H(+) antiport in Yersinia pestis virulence and found that Y. pestis strains lacking the major Na(+)/H(+) antiporters, NhaA and NhaB, are completely attenuated in an in vivo model of plague. The Y. pestis derivative strain lacking the nhaA and nhaB genes showed markedly decreased survival in blood and blood serum ex vivo. Complementation of either nhaA or nhaB in trans restored the survival of the Y. pestis nhaA nhaB double deletion mutant in blood. The nhaA nhaB double deletion mutant also showed inhibited growth in an artificial serum medium, Opti-MEM, and a rich LB-based medium with Na(+) levels and pH values similar to those for blood. Taken together, these data strongly suggest that intact Na(+)/H(+) antiport is indispensable for the survival of Y. pestis in the bloodstreams of infected animals and thus might be regarded as a promising noncanonical drug target for infections caused by Y. pestis and possibly for those caused by other blood-borne bacterial pathogens.


Assuntos
Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , Virulência/genética , Yersinia pestis/genética , Yersinia pestis/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Feminino , Camundongos , Peste/genética , Peste/metabolismo , Peste/microbiologia , Deleção de Sequência/genética , Ovinos/sangue , Ovinos/microbiologia
17.
Artigo em Inglês | MEDLINE | ID: mdl-23695558

RESUMO

The human pathogen Yersinia pestis requires the assembly of the type III secretion system (T3SS) for virulence. The structural component of the T3SS contains an external needle and a tip complex, which is formed by LcrV in Y. pestis. The structure of an LcrV triple mutant (K40A/D41A/K42A) in a C273S background has previously been reported to 2.2 Šresolution. Here, the crystal structure of LcrV without the triple mutation in a C273S background is reported at a higher resolution of 1.65 Å. Overall the two structures are similar, but there are also notable differences, particularly near the site of the triple mutation. The refined structure revealed a slight shift in the backbone positions of residues Gly28-Asn43 and displayed electron density in the loop region consisting of residues Ile46-Val63, which was disordered in the original structure. In addition, the helical turn region spanning residues Tyr77-Gln95 adopts a different orientation.


Assuntos
Antígenos de Bactérias/química , Proteínas Citotóxicas Formadoras de Poros/química , Yersinia pestis , Antígenos de Bactérias/genética , Cristalografia por Raios X , Mutação , Proteínas Citotóxicas Formadoras de Poros/genética , Estrutura Secundária de Proteína , Yersinia pestis/genética
18.
Microb Pathog ; 52(1): 41-6, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22023991

RESUMO

The plague bacterium Yersinia pestis has a number of well-described strategies to protect itself from both host cells and soluble factors. In an effort to identify additional anti-host factors, we employed a transposon site hybridization (TraSH)-based approach to screen 10(5)Y. pestis mutants in an in vitro infection system. In addition to loci encoding various components of the well-characterized type III secretion system (T3SS), our screen unambiguously identified ompA as a pro-survival gene. We go on to show that an engineered Y. pestis ΔompA strain, as well as a ΔompA strain of the closely related pathogen Yersinia pseudotuberculosis, have fully functioning T3SSs but are specifically defective in surviving within macrophages. Additionally, the Y. pestis ΔompA strain was out competed by the wild-type strain in a mouse co-infection assay. Unlike in other bacterial pathogens in which OmpA can promote adherence, invasion, or serum resistance, the OmpA of Y. pestis is restricted to enhancing intracellular survival. Our data show that OmpA of the pathogenic Yersinia is a virulence factor on par with the T3SS.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Viabilidade Microbiana , Peste/microbiologia , Yersinia pestis/metabolismo , Yersinia pestis/patogenicidade , Animais , Proteínas da Membrana Bacteriana Externa/genética , Feminino , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Virulência , Yersinia pestis/genética , Yersinia pestis/crescimento & desenvolvimento
19.
J Bacteriol ; 193(9): 2276-89, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21357482

RESUMO

YscD is an essential component of the plasmid pCD1-encoded type III secretion system (T3SS) of Yersinia pestis. YscD has a single transmembrane (TM) domain that connects a small N-terminal cytoplasmic region (residues 1 to 121) to a larger periplasmic region (residues 143 to 419). Deletion analyses established that both the N-terminal cytoplasmic region and the C-terminal periplasmic region are required for YscD function. Smaller targeted deletions demonstrated that a predicted cytoplasmic forkhead-associated (FHA) domain is also required to assemble a functional T3SS; in contrast, a predicted periplasmic phospholipid binding (BON) domain and a putative periplasmic "ring-building motif" domain of YscD could be deleted with no significant effect on the T3S process. Although deletion of the putative "ring-building motif" domain did not disrupt T3S activity per se, the calcium-dependent regulation of the T3S apparatus was affected. The extreme C-terminal region of YscD (residues 354 to 419) was essential for secretion activity and had a strong dominant-negative effect on the T3S process when exported to the periplasm of the wild-type parent strain. Coimmunoprecipitation studies demonstrated that this region of YscD mediates the interaction of YscD with the outer membrane YscC secretin complex. Finally, replacement of the YscD TM domain with a TM domain of dissimilar sequence had no effect on the T3S process, indicating that the TM domain has no sequence-specific function in the assembly or function of the T3SS.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Membrana Celular/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Proteínas de Membrana Transportadoras/metabolismo , Yersinia pestis/metabolismo , Sequência de Aminoácidos , Proteínas da Membrana Bacteriana Externa/genética , Sequência Conservada , Proteínas de Membrana Transportadoras/genética , Modelos Moleculares , Dados de Sequência Molecular , Família Multigênica , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Yersinia pestis/genética
20.
J Bacteriol ; 193(14): 3490-6, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21571996

RESUMO

The Chlamydia pneumoniae CopN protein is a member of the YopN/TyeA/InvE/MxiC family of secreted proteins that function to regulate the secretion of type III secretion system (T3SS) translocator and effector proteins. In this study, the Scc1 (CP0432) and Scc4 (CP0033) proteins of C. pneumoniae AR-39 were demonstrated to function together as a type III secretion chaperone that binds to an N-terminal region of CopN. The Scc1/Scc4 chaperone promoted the efficient secretion of CopN via a heterologous T3SS, whereas, the Scc3 chaperone, which binds to a C-terminal region of CopN, reduced CopN secretion.


Assuntos
Proteínas de Bactérias/metabolismo , Chlamydophila pneumoniae/metabolismo , Chaperonas Moleculares/metabolismo , Motivos de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Chlamydophila pneumoniae/química , Chlamydophila pneumoniae/genética , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Ligação Proteica , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA