Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Biol Chem ; 291(3): 1251-66, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26553869

RESUMO

HIV-1 infection results in a chronic illness because long-term highly active antiretroviral therapy can lower viral titers to an undetectable level. However, discontinuation of therapy rapidly increases virus burden. Moreover, patients under highly active antiretroviral therapy frequently develop various metabolic disorders, neurocognitive abnormalities, and cardiovascular diseases. We have previously shown that exosomes containing trans-activating response (TAR) element RNA enhance susceptibility of undifferentiated naive cells to HIV-1 infection. This study indicates that exosomes from HIV-1-infected primary cells are highly abundant with TAR RNA as detected by RT-real time PCR. Interestingly, up to a million copies of TAR RNA/µl were also detected in the serum from HIV-1-infected humanized mice suggesting that TAR RNA may be stable in vivo. Incubation of exosomes from HIV-1-infected cells with primary macrophages resulted in a dramatic increase of proinflammatory cytokines, IL-6 and TNF-ß, indicating that exosomes containing TAR RNA could play a direct role in control of cytokine gene expression. The intact TAR molecule was able to bind to PKR and TLR3 effectively, whereas the 5' and 3' stems (TAR microRNAs) bound best to TLR7 and -8 and none to PKR. Binding of TAR to PKR did not result in its phosphorylation, and therefore, TAR may be a dominant negative decoy molecule in cells. The TLR binding through either TAR RNA or TAR microRNA potentially can activate the NF-κB pathway and regulate cytokine expression. Collectively, these results imply that exosomes containing TAR RNA could directly affect the proinflammatory cytokine gene expression and may explain a possible mechanism of inflammation observed in HIV-1-infected patients under cART.


Assuntos
Fatores Ativadores da Transcrição/metabolismo , Citocinas/metabolismo , Exossomos/metabolismo , HIV-1/imunologia , Leucócitos/metabolismo , MicroRNAs/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Linhagem Celular Transformada , Transformação Celular Viral , Células Cultivadas , Exossomos/imunologia , Exossomos/virologia , Infecções por HIV/sangue , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Interleucina-6/metabolismo , Leucócitos/imunologia , Leucócitos/virologia , Linfotoxina-alfa/metabolismo , Camundongos Endogâmicos NOD , Camundongos Transgênicos , MicroRNAs/sangue , Receptor 3 Toll-Like/antagonistas & inibidores , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/metabolismo , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
2.
Biochim Biophys Acta Gen Subj ; 1861(1 Pt A): 3019-3029, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27612662

RESUMO

BACKGROUND: Using Bacillus anthracis as a model gram-positive bacterium, we investigated the effects of host protein S-nitrosylation during bacterial infection. B. anthracis possesses a bacterial nitric oxide synthase (bNOS) that is important for its virulence and survival. However, the role of S-nitrosylation of host cell proteins during B. anthracis infection has not been determined. METHODS: Nitrosoproteomic analysis of human small airway epithelial cells (HSAECs) infected with toxigenic B. anthracis Sterne was performed, identifying peroxiredoxin 1 (Prx1) as one predominant target. Peroxidase activity of Prx during infection was measured using 2-Cys-Peroxiredoxin activity assay. Chaperone activity of S-nitrosylated Prx1 was measured by insulin aggregation assay, and analysis of formation of multimeric species using Native PAGE. Griess assay and DAF-2DA fluorescence assay were used to measure NO production. Cell viability was measured using the Alamar Blue assay and the ATPlite assay (Perkin Elmer). RESULTS: S-nitrosylation of Prx1 in Sterne-infected HSAECs leads to a decrease in its peroxidase activity while enhancing its chaperone function. Treatment with bNOS inhibitor, or infection with bNOS deletion strain, reduces S-nitrosylation of Prx1 and decreases host cell survival. Consistent with this, siRNA knockdown of Prx1 lowers bNOS-dependent protection of HSAEC viability. CONCLUSIONS: Anthrax infection results in S-nitrosylation of multiple host proteins, including Prx1. The nitrosylation-dependent decrease in peroxidase activity of Prx1 and increase in its chaperone activity is one factor contributing to enhancing infected cell viability. GENERAL SIGNIFICANCE: These results provide a new venue of mechanistic investigation for inhalational anthrax that could lead to novel and potentially effective countermeasures.


Assuntos
Antraz/microbiologia , Antraz/patologia , Bacillus anthracis/patogenicidade , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Pulmão/patologia , Peroxirredoxinas/metabolismo , Bacillus anthracis/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Deleção de Genes , Humanos , Espectrometria de Massas , Modelos Biológicos , Chaperonas Moleculares/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Nitrosação , Peroxidase/metabolismo , Reprodutibilidade dos Testes
3.
Biochem J ; 470(2): 243-53, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26188040

RESUMO

In addition to membrane disruption, the cathelicidin antimicrobial peptide (AMP) LL-37 translocates through the bacterial inner membrane to target intracellular molecules. The present study aims to identify an alternate mechanism and a cytoplasmic target of LL-37 in Francisella. LL-37 binding proteins from Francisella novicida U112 bacterial lysates were precipitated by using biotinylated LL-37 (B-LL-37) and NeutrAvidin-agarose beads. Bound proteins were identified by LC-MS/MS, validated and characterized by bead pull-down assays and differential scanning fluorimetry (DSF). The cationic AMP (CAMP) LL-37 was able to interact with Francisella cytoplasmic acyl carrier protein (AcpP; FTN1340/FTT1376). Further study confirmed that LL-37 peptide could bind to AcpP and that the sheep cathelicidin SMAP-29 (Sheep Myeloid Antimicrobial Peptide 29) further increased LL-37 binding to AcpP, suggesting a synergistic effect of SMAP-29 on the binding. LL-37 could also bind to both AcpP of Escherichia coli and Bacillus anthracis, implying a mechanism of broad action of LL-37-AcpP binding. Overexpression of the acpP gene in F. novicida led to an increase in LL-37 susceptibility. LL-37 binding to AcpP changed the fatty acid composition profiles. Taken together, we identified a novel cytoplasmic target of LL-37 in Francisella, suggesting a mechanism of action of this peptide beyond membrane permeabilization. Our findings highlight a novel mechanism of antimicrobial activity of this peptide and document a previously unexplored target of α-helical CAMPs.


Assuntos
Proteína de Transporte de Acila/metabolismo , Antibacterianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas de Bactérias/metabolismo , Francisella/metabolismo , Sequência de Aminoácidos , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Citoplasma/metabolismo , Ácidos Graxos/metabolismo , Francisella/efeitos dos fármacos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Catelicidinas
4.
Appl Environ Microbiol ; 81(20): 7057-66, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26231649

RESUMO

In many bacteria, the ability to modulate biofilm production relies on specific signaling molecules that are either self-produced or made by neighboring microbes within the ecological niche. We analyzed the potential interspecies signaling effect of the Burkholderia diffusible signal factor (BDSF) on Francisella novicida, a model organism for Francisella tularensis, and demonstrated that BDSF both inhibits the formation and causes the dispersion of Francisella biofilm. Specificity was demonstrated for the cis versus the trans form of BDSF. Using transcriptome sequencing, quantitative reverse transcription-PCR, and activity assays, we found that BDSF altered the expression of many F. novicida genes, including genes involved in biofilm formation, such as chitinases. Using a chitinase inhibitor, the antibiofilm activity of BDSF was also shown to be chitinase dependent. In addition, BDSF caused an increase in RelA expression and increased levels of (p)ppGpp, leading to decreased biofilm production. These results support our observation that exposure of F. novicida to BDSF causes biofilm dispersal. Furthermore, BDSF upregulated the genes involved in iron acquisition (figABCD), increasing siderophore production. Thus, this study provides evidence for a potential role and mechanism of diffusible signal factor (DSF) signaling in the genus Francisella and suggests the possibility of interspecies signaling between Francisella and other bacteria. Overall, this study suggests that in response to the interspecies DSF signal, F. novicida can alter its gene expression and regulate its biofilm formation.


Assuntos
Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Burkholderia/metabolismo , Ácidos Graxos Monoinsaturados/metabolismo , Francisella tularensis/efeitos dos fármacos , Interações Microbianas , Sideróforos/metabolismo , Francisella tularensis/fisiologia , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de DNA
5.
Biochem Biophys Res Commun ; 430(1): 125-30, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-23178574

RESUMO

Bacillus anthracis, a causative agent of anthrax, is able to germinate and survive within macrophages. A recent study suggested that B. anthracis-derived nitric oxide (bNO) is a key aspect of bacterial defense that protects bacterial DNA from oxidative burst in the macrophages. However, the virulent effect of bNO in host cells has not been investigated. Here, we report that bNO contributes macrophage killing by S-nitrosylation of bioenergetic-relating proteins within mitochondria. Toxigenic Sterne induces expression of the bnos gene and produces bNO during early stage of infection. Nitroso-proteomic analysis coupled with a biotin-switch technique demonstrated that toxigenic infection induces protein S-nitrosylation in B. anthracis-susceptible RAW264.7. For each target enzyme tested (complex I, complex III and complex IV), infection by B. anthracis Sterne caused enzyme inhibition. Nω-nitro-L-arginine methyl ester, a NO synthase inhibitor, reduced S-nitrosylation and partially restored cell viability evaluated by intracellular ATP levels in macrophages. Our data suggest that bNO leads to energy depletion driven by impaired mitochondrial bioenergetic machinery that ultimately contributes to macrophage death. This novel mechanism of anthrax pathogenesis may offer specific approach to the development of therapeutics.


Assuntos
Antraz/imunologia , Apoptose/imunologia , Bacillus anthracis/patogenicidade , Macrófagos/microbiologia , Mitocôndrias/microbiologia , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico/metabolismo , Animais , Antraz/enzimologia , Antraz/microbiologia , Bacillus anthracis/metabolismo , Linhagem Celular , Complexo I de Transporte de Elétrons/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Expressão Gênica , Macrófagos/enzimologia , Camundongos , Mitocôndrias/enzimologia , Óxido Nítrico Sintase Tipo I/genética
6.
Cell Microbiol ; 14(8): 1219-30, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22452315

RESUMO

To achieve widespread dissemination in the host, Bacillus anthracis cells regulate their attachment to host endothelium during infection. Previous studies identified BslA (Bacillus anthracis S-layer Protein A), a virulence factor of B. anthracis, as necessary and sufficient for adhesion of vegetative cells to human endothelial cells. While some factors have been identified, bacteria-specific contributions to BslA mediated adhesion remain unclear. Using the attenuated vaccine Sterne 7702 strain of B. anthracis, we tested the hypothesis that InhA (immune inhibitor A), a B. anthracis protease, regulates BslA levels affecting the bacteria's ability to bind to endothelium. To test this, a combination of inhA mutant and complementation analysis in adhesion and invasion assays, Western blot and InhA inhibitor assays were employed. Results show InhA downregulates BslA activity reducing B. anthracis adhesion and invasion in human brain endothelial cells. BslA protein levels in ΔinhA bacteria were significantly higher than wild-type and complemented strains showing InhA levels and BslA expression are inversely related. BslA was sensitive to purified InhA degradation in a concentration- and time-dependent manner. Taken together these data support the role of InhA regulation of BslA-mediated vegetative cell adhesion and invasion.


Assuntos
Adesinas Bacterianas/metabolismo , Bacillus anthracis/enzimologia , Aderência Bacteriana , Células Endoteliais/microbiologia , Metaloproteases/metabolismo , Animais , Bacillus anthracis/genética , Bacillus anthracis/fisiologia , Bicarbonatos/química , Encéfalo/irrigação sanguínea , Meios de Cultura/química , Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Técnicas de Inativação de Genes , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/microbiologia , Metaloproteases/genética , Camundongos , Viabilidade Microbiana , Microvasos/citologia , Fenantrolinas/farmacologia , Inibidores de Proteases/farmacologia , Proteólise , Fatores de Virulência/metabolismo
7.
J Med Microbiol ; 58(Pt 6): 737-744, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19429749

RESUMO

Anthrax is a zoonotic disease caused by Bacillus anthracis. The infection is associated with inflammation and sepsis, but little is known about the acute-phase response during disease and the nature of the bacterial factors causing it. In this study, we examined the levels of the acute-phase proteins (APPs) in comparative experiments using mice challenged with spores and a purified B. anthracis protease InhA as a possible factor mediating the response. A strong increase in the plasma levels of APPs such as haptoglobin and serum amyloid A was observed during infection. Protein and mRNA levels of plasminogen activator inhibitor (PAI)-1 in the liver were also increased concurrently with bacterial dissemination at 72 h post-infection. Similar effects were observed at 6 h post injection with InhA. Induction of hepatic transforming growth factor-beta1, a PAI-1 inducer, was also found in the liver of InhA-injected mice. PAI-1 elevation by InhA resulted in an increased level of urokinase-type plasminogen activator complex with PAI-1 and a decreased level of D-dimers indicating inhibition of blood fibrinolysis. These results reveal an acute liver response to anthrax infection and provide a plausible pathophysiological link between the host inflammatory response and the pro-thrombotic haemostatic imbalance in the course of disease through PAI-1 induction in the liver.


Assuntos
Proteínas de Fase Aguda/metabolismo , Antraz/fisiopatologia , Bacillus anthracis/enzimologia , Bacillus anthracis/patogenicidade , Metaloproteases/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Regulação para Cima , Animais , Antraz/imunologia , Antraz/microbiologia , Bacillus anthracis/fisiologia , Proteínas de Bactérias/metabolismo , Feminino , Haptoglobinas/metabolismo , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Esporos Bacterianos/patogenicidade , Trombose/imunologia , Trombose/microbiologia
8.
BMC Immunol ; 9: 67, 2008 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-19014542

RESUMO

BACKGROUND: Bacillus anthracis secretes several virulence factors targeting different host organs and cell types during inhalational anthrax infection. The bacterial expression of a key virulence factor, lethal toxin (LeTx) is closely tied to another factor, edema toxin (EdTx). Both are transcribed on the same virulence plasmid (pXO1) and both have been the subject of much individual study. Their combined effect during virulent anthrax likely modulates both the global transcriptional and the phenotypic response of macrophages and phagocytes. In fact, responses brought about by the toxins may be different than each of their individual effects. RESULTS: Here we report the transcriptional and apoptotic responses of the macrophage-like phagocytic cell line THP-1 exposed to B. anthracis Sterne (pXO1+) spores, and B. anthracis Delta Sterne (pXO1-) spores. These cells are resistant to LeTx-induced cytolysis, a phenotype seen in macrophages from several mouse strains which are sensitive to toxigenic anthrax infection. Our results indicate that the pXO1-containing strain induces higher pro-inflammatory transcriptional responses during the first 4 hours of interaction with bacterium, evident in the upregulation of several genes relevant to Nf-kappaB, phosphatases, prostaglandins, and TNF-alpha, along with decreases in expression levels of genes for mitochondrial components. Both bacterial strains induce apoptosis, but in the toxigenic strain-challenged cells, apoptosis is delayed. CONCLUSION: This delay in apoptosis occurs despite the much higher level of TNF-alpha secretion induced by the toxigenic-strain challenge. Interestingly, CFLAR, an important apoptotic inhibitor which blocks apoptosis induced by large amounts of extracellular TNF-alpha, is upregulated significantly during toxigenic-strain infection, but not at all during non-toxigenic-strain infection, indicating that it may play a role in blocking or delaying TNF-alpha-mediated apoptosis. The suppression of apoptosis by the toxigenic anthrax strain is consistent with the notion that apoptosis itself may represent a protective host cell response.


Assuntos
Antraz/imunologia , Antígenos de Bactérias/imunologia , Apoptose , Toxinas Bacterianas/imunologia , Macrófagos/imunologia , Transcrição Gênica , Fatores de Virulência/imunologia , Animais , Antraz/microbiologia , Antígenos de Bactérias/metabolismo , Bacillus anthracis/imunologia , Toxinas Bacterianas/metabolismo , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Linhagem Celular , Expressão Gênica , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Virulência/metabolismo
9.
FEMS Immunol Med Microbiol ; 54(3): 309-18, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19049643

RESUMO

Bacillus anthracis infection is associated with severe hemostatic disturbances but their roles and contribution to fatality remain incompletely characterized. We undertook analyses of circulating antithrombin levels during the course of infection using a comparison of lethal and nonlethal murine anthrax models. Plasma samples were obtained from DBA/2 mice challenged intraperitoneally with the spores of either toxigenic B. anthracis Sterne strain or nontoxigenic, avirulent delta Sterne strain. We found that plasma antithrombin levels were rapidly depleted in Sterne spore-challenged mice, concomitant with elevation of neutrophil elastase (NE) and massive syndecan shedding from the liver into circulation. The shed syndecan bound with antithrombin accelerated NE-mediated antithrombin proteolysis. The liver response to infection demonstrated strain-specific compensatory increases of antithrombin and syndecan gene transcription. Both bacterial strains induced changes in blood coagulation parameters consistent with the onset of disseminated intravascular coagulation. We propose that antithrombin depletion proceeding through activation of neutrophils and massive shedding of heparin-like syndecan from the liver into circulation contribute to anthrax coagulopathy.


Assuntos
Antraz , Antitrombinas/metabolismo , Bacillus anthracis/patogenicidade , Coagulação Intravascular Disseminada/fisiopatologia , Elastase de Leucócito/sangue , Sindecanas/metabolismo , Animais , Antraz/complicações , Antraz/microbiologia , Antraz/mortalidade , Bacillus anthracis/fisiologia , Modelos Animais de Doenças , Coagulação Intravascular Disseminada/etiologia , Feminino , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos DBA , Ativação de Neutrófilo , Esporos Bacterianos/fisiologia , Sindecana-1/metabolismo , Sindecana-4/metabolismo , Virulência
10.
PLoS One ; 9(3): e93119, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24664176

RESUMO

Biofilms, multicellular communities of bacteria, may be an environmental survival and transmission mechanism of Francisella tularensis. Chitinases of F. tularensis ssp. novicida (Fn) have been suggested to regulate biofilm formation on chitin surfaces. However, the underlying mechanisms of how chitinases may regulate biofilm formation are not fully determined. We hypothesized that Fn chitinase modulates bacterial surface properties resulting in the alteration of biofilm formation. We analyzed biofilm formation under diverse conditions using chitinase mutants and their counterpart parental strain. Substratum surface charges affected biofilm formation and initial attachments. Biophysical analysis of bacterial surfaces confirmed that the chi mutants had a net negative-charge. Lectin binding assays suggest that chitinase cleavage of its substrates could have exposed the concanavalin A-binding epitope. Fn biofilm was sensitive to chitinase, proteinase and DNase, suggesting that Fn biofilm contains exopolysaccharides, proteins and extracellular DNA. Exogenous chitinase increased the drug susceptibility of Fn biofilms to gentamicin while decreasing the amount of biofilm. In addition, chitinase modulated bacterial adhesion and invasion of A549 and J774A.1 cells as well as intracellular bacterial replication. Our results support a key role of the chitinase(s) in biofilm formation through modulation of the bacterial surface properties. Our findings position chitinase as a potential anti-biofilm enzyme in Francisella species.


Assuntos
Proteínas de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Quitinases/metabolismo , Francisella/fisiologia , Mutação , Antibacterianos/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Aderência Bacteriana/fisiologia , Proteínas de Bactérias/genética , Biofilmes/efeitos dos fármacos , Linhagem Celular , Quitinases/genética , Gentamicinas/farmacologia , Humanos
11.
Pathog Dis ; 71(2): 109-20, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24449527

RESUMO

Exosomes have recently been classified as the newest family members of 'bioactive vesicles' that function to promote intercellular communication. Long ignored and thought to be only a mechanism by which cellular waste is removed, exosomes have garnered a huge amount of interest in recent years as their critical functions in maintaining homeostasis through intercellular communication and also in different types of diseases have been demonstrated. Many groundbreaking studies of exosome functions have been performed in the cancer field and the infectious disease areas of study, revealing the importance and also the fascinating complexity of exosomal packaging, targeting, and functions. Selective packaging of exosomes in response to the type of infection, exosomal modulation of the immune response and host signaling pathways, exosomal regulation of pathogen spread, and effects of exosomes on the degree of pathogenesis have all been well documented. In this review, we provide a synthesis of the current understanding of the role of exosomes during infections caused by human pathogens and discuss the implications of these findings for a better understanding of pathogenic mechanisms and future therapeutic and diagnostic applications.


Assuntos
Comunicação Celular , Fenômenos Fisiológicos Celulares , Doenças Transmissíveis/patologia , Doenças Transmissíveis/fisiopatologia , Exossomos/metabolismo , Humanos , Modelos Biológicos
12.
Microbes Infect ; 15(1): 37-44, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23108317

RESUMO

Bacillus anthracis is the causative agent of anthrax and is acquired by three routes of infection: inhalational, gastrointestinal and cutaneous. Gastrointestinal (GI) anthrax is rare, but can rapidly result in severe, systemic disease that is fatal in 25%-60% of cases. Disease mechanisms of GI anthrax remain unclear due to limited numbers of clinical cases and the lack of experimental animal models. Here, we developed an in vivo murine model of GI anthrax where spore survival was maximized through the neutralization of stomach acid followed by an intragastric administration of a thiabendazole paste spore formulation. Infected mice showed a dose-dependent mortality rate and pathological features closely mimicking human GI anthrax. Since Peyer's patches in the murine intestine are the primary sites of B. anthracis growth, we developed a human M (microfold)-like-cell model using a Caco-2/Raji B-cell co-culturing system to study invasive mechanisms of GI anthrax across the intestinal epithelium. Translocation of B. anthracis spores was higher in M-like cells than Caco-2 monolayers, suggesting that M-like cells may serve as an initial entry site for spores. Here, we developed an in vivo murine model of GI anthrax and an in vitro M-like cell model that could be used to further our knowledge of GI anthrax pathogenesis.


Assuntos
Antraz/microbiologia , Bacillus anthracis/fisiologia , Modelos Animais de Doenças , Gastroenteropatias/microbiologia , Animais , Antraz/patologia , Bacillus anthracis/patogenicidade , Aderência Bacteriana/fisiologia , Carga Bacteriana , Translocação Bacteriana , Células CACO-2 , Feminino , Gastroenteropatias/patologia , Interações Hospedeiro-Patógeno , Humanos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos DBA , Esporos Bacterianos
13.
Artigo em Inglês | MEDLINE | ID: mdl-23730627

RESUMO

Bacillus anthracis is a dangerous pathogen of humans and many animal species. Its virulence has been mainly attributed to the production of Lethal and Edema toxins as well as the antiphagocytic capsule. Recent data indicate that the nitric oxide (NO) synthase (baNOS) plays an important pathogenic role at the early stage of disease by protecting bacteria from the host reactive species and S-nytrosylating the mitochondrial proteins in macrophages. In this study we for the first time present evidence that bacteria-derived NO participates in the generation of highly reactive oxidizing species which could be abolished by the NOS inhibitor L - NAME, free thiols, and superoxide dismutase but not catalase. The formation of toxicants is likely a result of the simultaneous formation of NO and superoxide leading to a labile peroxynitrite and its stable decomposition product, nitrogen dioxide. The toxicity of bacteria could be potentiated in the presence of bovine serum albumin. This effect is consistent with the property of serum albumin to serves as a trap of a volatile NO accelerating its reactions. Our data suggest that during infection in the hypoxic environment of pre-mortal host the accumulated NO is expected to have a broad toxic impact on host cell functions.


Assuntos
Bacillus anthracis/metabolismo , Bacillus anthracis/patogenicidade , Óxido Nítrico/metabolismo , Soroalbumina Bovina/metabolismo , Animais , Bovinos , Humanos , Hipóxia , Óxido Nítrico/toxicidade , Superóxidos/metabolismo , Superóxidos/toxicidade
14.
FEMS Immunol Med Microbiol ; 61(1): 15-27, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20946354

RESUMO

Bacillus anthracis generates virulence factors such as lethal and edema toxins, capsule, and hemolytic proteins under conditions of reduced oxygenation. Here, we report on the acute cytotoxicity of culture supernatants (Sups) of six nonencapsulated B. anthracis strains grown till the stationary phase under static microaerobic conditions. Human small airway epithelial, umbilical vein endothelial, Caco-2, and Hep-G2 cells were found to be susceptible. Sups displayed a reduction of pH to 5.3-5.5, indicating the onset of acid anaerobic fermentation; however, low pH itself was not a major factor of toxicity. The pore-forming hemolysin, anthrolysin O (ALO), contributed to the toxicity in a concentration-dependent manner. Its effect was found to be synergistic with a metabolic product of B. anthracis, succinic acid. Cells exposed to Sups demonstrated cytoplasmic membrane blebbing, increased permeability, loss of ATP, mitochondrial membrane potential collapse, and arrest of cell respiration. The toxicity was reduced by inhibition of ALO by cholesterol, decomposition of reactive oxygen species, and inhibition of mitochondrial succinate dehydrogenase. Cell death appears to be caused by an acute primary membrane permeabilization by ALO, followed by a burst of reactive radicals from the mitochondria fuelled by the succinate, which is generated by bacteria in the hypoxic environment. This mechanism of metabolic toxicity is relevant to the late-stage conditions of hypoxia and acidosis found in anthrax patients and might operate at anatomical locations of the host deprived from oxygen supply.


Assuntos
Bacillus anthracis/metabolismo , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Células Epiteliais/efeitos dos fármacos , Fermentação , Glicoproteínas de Membrana/toxicidade , Aerobiose , Bacillus anthracis/genética , Bacillus anthracis/crescimento & desenvolvimento , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Células CACO-2 , Sobrevivência Celular/efeitos dos fármacos , Colesterol/metabolismo , Meios de Cultivo Condicionados/química , Células Epiteliais/metabolismo , Deleção de Genes , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Pulmão/citologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mitocôndrias/efeitos dos fármacos , Espécies Reativas de Oxigênio/toxicidade , Proteínas Recombinantes/toxicidade , Ácido Succínico/toxicidade , Fatores de Virulência/metabolismo , Fatores de Virulência/farmacologia , Fatores de Virulência/toxicidade
15.
FEMS Immunol Med Microbiol ; 62(2): 173-81, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21395696

RESUMO

The fibrinolytic system is often the target for pathogenic bacteria, resulting in increased fibrinolysis, bacterial dissemination, and inflammation. The purpose of this study was to explore whether proteases NprB and InhA secreted by Bacillus anthracis could activate the host's fibrinolytic system. NprB efficiently activated human pro-urokinase plasminogen activator (pro-uPA), a key protein in the fibrinolytic cascade. Conversely, InhA had little effect on pro-uPA. Plasminogen activator inhibitors (PAI)-1, 2 and the uPA receptor were also targets for NprB in vitro. InhA efficiently degraded the thrombin-activatable fibrinolysis inhibitor (TAFI) in vitro. Mice infected with B. anthracis showed a significant decrease in blood TAFI levels. In another mouse experiment, animals infected with isogenic inhA deletion mutants restored TAFI levels, while the levels in the parent strain decreased. We propose that NprB and InhA may contribute to the activation of the fibrinolytic system in anthrax infection.


Assuntos
Bacillus anthracis/enzimologia , Fibrinólise , Peptídeo Hidrolases/metabolismo , Animais , Antraz/microbiologia , Antraz/patologia , Carboxipeptidase B2/sangue , Carboxipeptidase B2/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos DBA , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Inibidor 2 de Ativador de Plasminogênio/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteínas Recombinantes/metabolismo , Doenças dos Roedores/microbiologia , Doenças dos Roedores/patologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
16.
PLoS One ; 6(3): e17921, 2011 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-21437287

RESUMO

Hemorrhagic meningitis is a fatal complication of anthrax, but its pathogenesis remains poorly understood. The present study examined the role of B. anthracis-secreted metalloprotease InhA on monolayer integrity and permeability of human brain microvasculature endothelial cells (HBMECs) which constitute the blood-brain barrier (BBB). Treatment of HBMECs with purified InhA resulted in a time-dependent decrease in trans-endothelial electrical resistance (TEER) accompanied by zonula occluden-1 (ZO-1) degradation. An InhA-expressing B. subtilis exhibited increased permeability of HBMECs, which did not occur with the isogenic inhA deletion mutant (ΔinhA) of B. anthracis, compared with the corresponding wild-type strain. Mice intravenously administered with purified InhA or nanoparticles-conjugated to InhA demonstrated a time-dependent Evans Blue dye extravasation, leptomeningeal thickening, leukocyte infiltration, and brain parenchymal distribution of InhA indicating BBB leakage and cerebral hemorrhage. Mice challenged with vegetative bacteria of the ΔinhA strain of B. anthracis exhibited a significant decrease in leptomeningeal thickening compared to the wildtype strain. Cumulatively, these findings indicate that InhA contributes to BBB disruption associated with anthrax meningitis through proteolytic attack on the endothelial tight junctional protein zonula occluden (ZO)-1.


Assuntos
Bacillus anthracis/enzimologia , Proteínas de Bactérias/metabolismo , Barreira Hematoencefálica/microbiologia , Barreira Hematoencefálica/patologia , Hemorragia Cerebral/microbiologia , Hemorragia Cerebral/patologia , Metaloproteases/metabolismo , Animais , Antraz/microbiologia , Antraz/patologia , Western Blotting , Citoplasma/metabolismo , Impedância Elétrica , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Células Endoteliais/patologia , Feminino , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Mutantes/metabolismo , Nanosferas , Permeabilidade , Fosfoproteínas/metabolismo , Proteína da Zônula de Oclusão-1
17.
PLoS One ; 6(3): e18119, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21464960

RESUMO

The causative agent of anthrax, Bacillus anthracis, is capable of circumventing the humoral and innate immune defense of the host and modulating the blood chemistry in circulation to initiate a productive infection. It has been shown that the pathogen employs a number of strategies against immune cells using secreted pathogenic factors such as toxins. However, interference of B. anthracis with the innate immune system through specific interaction of the spore surface with host proteins such as the complement system has heretofore attracted little attention. In order to assess the mechanisms by which B. anthracis evades the defense system, we employed a proteomic analysis to identify human serum proteins interacting with B. anthracis spores, and found that plasminogen (PLG) is a major surface-bound protein. PLG efficiently bound to spores in a lysine- and exosporium-dependent manner. We identified α-enolase and elongation factor tu as PLG receptors. PLG-bound spores were capable of exhibiting anti-opsonic properties by cleaving C3b molecules in vitro and in rabbit bronchoalveolar lavage fluid, resulting in a decrease in macrophage phagocytosis. Our findings represent a step forward in understanding the mechanisms involved in the evasion of innate immunity by B. anthracis through recruitment of PLG resulting in the enhancement of anti-complement and anti-opsonization properties of the pathogen.


Assuntos
Bacillus anthracis/imunologia , Complemento C3b/imunologia , Fibrinolisina/metabolismo , Imunidade Inata/imunologia , Plasminogênio/metabolismo , Animais , Bacillus anthracis/efeitos dos fármacos , Líquido da Lavagem Broncoalveolar/imunologia , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Eletroforese em Gel Bidimensional , Humanos , Imunidade Inata/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Proteínas Opsonizantes/imunologia , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Ligação Proteica/efeitos dos fármacos , Coelhos , Proteínas Recombinantes/metabolismo , Esporos Bacterianos/efeitos dos fármacos , Esporos Bacterianos/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/farmacologia
18.
J Biol Chem ; 283(15): 9531-42, 2008 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-18263586

RESUMO

Pathology data from the anthrax animal models show evidence of significant increases in vascular permeability coincident with hemostatic imbalances manifested by thrombocytopenia, transient leucopenia, and aggressive disseminated intravascular coagulation. In this study we hypothesized that anthrax infection modulates the activity of von Willebrand factor (VWF) and its endogenous regulator ADAMTS13, which play important roles in hemostasis and thrombosis, including interaction of endothelial cells with platelets. We previously demonstrated that purified anthrax neutral metalloproteases Npr599 and InhA are capable of cleaving a variety of host structural and regulatory proteins. Incubation of human plasma with these proteases at 37 degrees C in the presence of urea as a mild denaturant results in proteolysis of VWF. Also in these conditions, InhA directly cleaves plasma ADAMTS13 protein. Npr599 and InhA digest synthetic VWF substrate FRETS-VWF73. Amino acid sequencing of VWF fragments produced by InhA suggests that one of the cleavage sites of VWF is located at domain A2, the target domain of ADAMTS13. Proteolysis of VWF by InhA impairs its collagen binding activity (VWF:CBA) and ristocetin-induced platelet aggregation activity. In plasma from anthrax spore-challenged DBA/2 mice, VWF antigen levels increase up to 2-fold at day 3 post-infection with toxigenic Sterne 34F(2) strain, whereas VWF:CBA levels drop in a time-dependent manner, suggesting dysfunction of VWF instead of its quantitative deficiency. This conclusion is further supported by significant reduction in the amount of VWF circulating in blood in the ultra-large forms. In addition, Western blot analysis shows proteolytic depletion of ADAMTS13 from plasma of spore-challenged mice despite its increased expression in the liver. Our results suggest a new mechanism of anthrax coagulopathy affecting the levels and functional activities of both VWF and its natural regulator ADAMTS13. This mechanism may contribute to hemorrhage and thrombosis typical in anthrax.


Assuntos
Proteínas ADAM/metabolismo , Antraz/enzimologia , Proteínas de Bactérias/metabolismo , Coagulação Intravascular Disseminada/enzimologia , Metaloproteases/metabolismo , Fator de von Willebrand/metabolismo , Proteína ADAMTS13 , Animais , Antraz/patologia , Antibacterianos/farmacologia , Plaquetas/metabolismo , Plaquetas/microbiologia , Plaquetas/patologia , Comunicação Celular/efeitos dos fármacos , Colágeno/metabolismo , Modelos Animais de Doenças , Coagulação Intravascular Disseminada/microbiologia , Coagulação Intravascular Disseminada/patologia , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Células Endoteliais/patologia , Hemostasia/efeitos dos fármacos , Humanos , Leucopenia/enzimologia , Leucopenia/microbiologia , Leucopenia/patologia , Metaloendopeptidases/metabolismo , Camundongos , Plasma/enzimologia , Plasma/microbiologia , Agregação Plaquetária/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Ristocetina/farmacologia , Esporos Bacterianos/enzimologia , Especificidade por Substrato/efeitos dos fármacos , Trombocitopenia/enzimologia , Trombocitopenia/microbiologia , Trombocitopenia/patologia , Trombose/enzimologia , Trombose/microbiologia , Trombose/patologia , Fatores de Tempo , Ureia/farmacologia
19.
J Biol Chem ; 281(42): 31408-18, 2006 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-16926147

RESUMO

To evaluate the pathogenic potential of Bacillus anthracis-secreted proteases distinct from lethal toxin, two neutral zinc metalloproteases were purified to apparent homogeneity from the culture supernatant of a non-virulent delta Ames strain (pXO1-, pXO2-). The first (designated Npr599) is a thermolysin-like enzyme highly homologous to bacillolysins from other Bacillus species. The second (designated InhA) is a homolog of the Bacillus thuringiensis immune inhibitor A. These proteases belong to the M4 and M6 families, respectively. Both enzymes digested various substrates, including extracellular matrix proteins, endogenous inhibitors, and coagulation proteins, with some differences in specificity. In addition, InhA accelerated urokinase-mediated plasminogen activation, suggesting that InhA acts as a modulator of plasmin in the host inflammatory system. Relevant to epithelial barrier function, Npr599 and InhA significantly enhanced syndecan-1 shedding from cultured normal murine mammary gland cells without affecting their viability through stimulation of the host cell ectodomain shedding mechanism. In addition, Npr599 and InhA directly cleaved recombinant syndecan-1 fused to glutathione S-transferase. Mass spectrometric analysis suggested that the cleavage sites of Npr599 and InhA are the Asp(39)-Asp(40) and Gly(48)-Thr(49) bonds, respectively. We propose that Npr599 and InhA from B. anthracis are multifunctional pathogenic factors that may contribute to anthrax pathology through direct degradation of host tissues, increases in barrier permeability, and/or modulation of host defenses.


Assuntos
Bacillus anthracis/enzimologia , Bacillus anthracis/patogenicidade , Metaloproteases/química , Animais , Células Cultivadas , Colagenases/química , Relação Dose-Resposta a Droga , Regulação Bacteriana da Expressão Gênica , Glutationa Transferase/metabolismo , Inflamação , Cinética , Espectrometria de Massas , Camundongos , Estrutura Terciária de Proteína , Proteínas Recombinantes/química
20.
J Biol Chem ; 279(53): 56042-52, 2004 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-15496418

RESUMO

Transcription of the nonmuscle myosin heavy chain II-A (NMHC-A) gene is regulated by various factors, including cell type, proliferation and differentiation stage, and extracellular stimuli. We have identified an intronic region (designated 32kb-150), which is located 32 kb downstream of the transcription start sites in the human NMHC-A gene, as a transcriptional regulatory region. 32kb-150 contains an interferon-stimulated response element (ISRE). By using HeLa and NIH3T3 cells, in which NMHC-A is constitutively expressed, interferon regulatory factor (IRF)-2 was found to be the only major protein, among the IRF family proteins, that bound to the ISRE in 32kb-150 both in vitro and in intact cells. IRF-2, which is known to either repress or activate target gene expression, acts as a transcriptional activator in the context of the 32kb-150 reporter gene. The carboxyl-terminal basic region of IRF-2 serves as an activation domain in this context. This is in contrast to its acting as a repressor domain in the context of the synthetic core ISRE. Furthermore, after treatment of promyelocytic HL-60 cells with 12-O-tetradecanoylphorbol-13-acetate (TPA), which triggers differentiation into macrophages, both NMHC-A expression and IRF-2 expression were found to be up-regulated with a similar time course. TPA treatment leads to recruitment of IRF-2 to 32kb-150 of the endogenous NMHC-A gene and acetylation of the core histones surrounding this region. In addition, the ISRE in the 32kb-150 reporter gene recruits IRF-2 and mediates TPA-induced activation of a reporter gene in HL-60 cells. Together, these results indicate that IRF-2 contributes to transcriptional activation of the NMHC-A gene via 32kb-150 during TPA-induced differentiation of HL-60 cells.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica , Miosina não Muscular Tipo IIA/biossíntese , Proteínas Repressoras/fisiologia , Fatores de Transcrição/fisiologia , Regulação para Cima , Animais , Sequência de Bases , Northern Blotting , Western Blotting , Adesão Celular , Diferenciação Celular , Linhagem Celular , Imunoprecipitação da Cromatina , Sequência Conservada , Proteínas de Ligação a DNA/metabolismo , Deleção de Genes , Genes Reporter , Vetores Genéticos , Células HL-60 , Células HeLa , Humanos , Fator Regulador 2 de Interferon , Íntrons , Luciferases/metabolismo , Macrófagos/metabolismo , Camundongos , Modelos Genéticos , Dados de Sequência Molecular , Células NIH 3T3 , Plasmídeos/metabolismo , Ligação Proteica , Proteínas Repressoras/metabolismo , Homologia de Sequência do Ácido Nucleico , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Transcrição/metabolismo , Transcrição Gênica , Ativação Transcricional , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA