Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
PLoS Pathog ; 10(10): e1004439, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25340543

RESUMO

Francisella tularensis causes the disease tularemia. Human pulmonary exposure to the most virulent form, F. tularensis subsp. tularensis (Ftt), leads to high morbidity and mortality, resulting in this bacterium being classified as a potential biothreat agent. However, a closely-related species, F. novicida, is avirulent in healthy humans. No tularemia vaccine is currently approved for human use. We demonstrate that a single dose vaccine of a live attenuated F. novicida strain (Fn iglD) protects against subsequent pulmonary challenge with Ftt using two different animal models, Fischer 344 rats and cynomolgus macaques (NHP). The Fn iglD vaccine showed protective efficacy in rats, as did a Ftt iglD vaccine, suggesting no disadvantage to utilizing the low human virulent Francisella species to induce protective immunity. Comparison of specific antibody profiles in vaccinated rat and NHP sera by proteome array identified a core set of immunodominant antigens in vaccinated animals. This is the first report of a defined live attenuated vaccine that demonstrates efficacy against pulmonary tularemia in a NHP, and indicates that the low human virulence F. novicida functions as an effective tularemia vaccine platform.


Assuntos
Vacinas Bacterianas/imunologia , Francisella tularensis , Epitopos Imunodominantes/imunologia , Tularemia/imunologia , Animais , Macaca fascicularis , Camundongos , Modelos Animais , Ratos Endogâmicos F344 , Tularemia/mortalidade , Tularemia/prevenção & controle , Vacinação , Vacinas Atenuadas/imunologia
2.
J Bacteriol ; 197(14): 2400-11, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25962917

RESUMO

UNLABELLED: Bacteria sustain an infection by acquiring nutrients from the host to support replication. The host sequesters these nutrients as a growth-restricting strategy, a concept termed "nutritional immunity." Historically, the study of nutritional immunity has centered on iron uptake because many bacteria target hemoglobin, an abundant circulating protein, as an iron source. Left unresolved are the mechanisms that bacteria use to attain other nutrients from host sources, including amino acids. We employed a novel medium designed to mimic the chemical composition of human serum, and we show here that Bacillus anthracis, the causative agent of anthrax disease, proteolyzes human hemoglobin to liberate essential amino acids which enhance its growth. This property can be traced to the actions of InhA1, a secreted metalloprotease, and extends to at least three other serum proteins, including serum albumin. The results suggest that we must also consider proteolysis of key host proteins to be a way for bacterial pathogens to attain essential nutrients, and we provide an experimental framework to determine the host and bacterial factors involved in this process. IMPORTANCE: The mechanisms by which bacterial pathogens acquire nutrients during infection are poorly understood. Here we used a novel defined medium that approximates the chemical composition of human blood serum, blood serum mimic (BSM), to better model the nutritional environment that pathogens encounter during bacteremia. Removing essential amino acids from BSM revealed that two of the most abundant proteins in blood-hemoglobin and serum albumin-can satiate the amino acid requirement for Bacillus anthracis, the causative agent of anthrax. We further demonstrate that hemoglobin is proteolyzed by the secreted protease InhA1. These studies highlight that common blood proteins can be a nutrient source for bacteria. They also challenge the historical view that hemoglobin is solely an iron source for bacterial pathogens.


Assuntos
Aminoácidos/metabolismo , Bacillus anthracis/metabolismo , Proteínas Sanguíneas/metabolismo , Soro/química , Sequência de Aminoácidos , Aminoácidos/química , Bacillus anthracis/genética , Proteínas de Bactérias/metabolismo , Proteínas Sanguíneas/química , Meios de Cultura/química , Heme/metabolismo , Hemoglobinas , Humanos , Ferro/metabolismo , Dados de Sequência Molecular
3.
PLoS Pathog ; 9(4): e1003306, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23637599

RESUMO

Bacterial capsules are common targets for antibody-mediated immunity. The capsule of Bacillus anthracis is unusual among capsules because it is composed of a polymer of poly-γ-d-glutamic acid (γdPGA). We previously generated murine IgG3 monoclonal antibodies (mAbs) to γdPGA that were protective in a murine model of pulmonary anthrax. IgG3 antibodies are characteristic of the murine response to polysaccharide antigens. The goal of the present study was to produce subclass switch variants of the γdPGA mAbs (IgG3 → IgG1 → IgG2b → IgG2a) and assess the contribution of subclass to antibody affinity and protection. Subclass switch antibodies had identical variable regions but differed in their heavy chains. The results showed that a switch from the protective IgG3 to IgG1, IgG2b or IgG2a was accompanied by i) a loss of protective activity ii) a change in mAb binding to the capsular matrix, and iii) a loss of affinity. These results identify a role for the heavy chain constant region in mAb binding. Hybrid mAbs were constructed in which the CH1, CH2 or CH3 heavy chain constant domains from a non-protective, low binding IgG2b mAb were swapped into the protective IgG3 mAb. The IgG3 mAb that contained the CH1 domain from IgG2b showed no loss of affinity or protection. In contrast, swapping the CH2 or CH3 domains from IgG2b into IgG3 produced a reduction in affinity and a loss of protection. These studies identify a role for the constant region of IgG heavy chains in affinity and protection against an encapsulated bacterial pathogen.


Assuntos
Antraz/imunologia , Bacillus anthracis/imunologia , Regiões Constantes de Imunoglobulina/imunologia , Imunoglobulina G/imunologia , Cadeias Pesadas de Imunoglobulinas/imunologia , Animais , Antraz/microbiologia , Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos , Reações Antígeno-Anticorpo , Cápsulas Bacterianas/imunologia , Ácido Glutâmico/imunologia , Switching de Imunoglobulina , Imunoglobulina G/química , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Terciária de Proteína
4.
Am J Pathol ; 184(12): 3205-16, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25285720

RESUMO

Inhalational anthrax is caused by inhalation of Bacillus anthracis spores. The ability of B. anthracis to cause anthrax is attributed to the plasmid-encoded A/B-type toxins, edema toxin (edema factor and protective antigen) and lethal toxin (lethal factor and protective antigen), and a poly-d-glutamic acid capsule. To better understand the contribution of these toxins to the disease pathophysiology in vivo, we used B. anthracis Ames strain and isogenic toxin deletion mutants derived from the Ames strain to examine the role of lethal toxin and edema toxin after pulmonary spore challenge of cynomolgus macaques. Lethal toxin, but not edema toxin, was required to induce sustained bacteremia and death after pulmonary challenge with spores delivered via bronchoscopy. After intravenous challenge with bacilli to model the systemic phase of infection, lethal toxin contributed to bacterial proliferation and subsequent host death to a greater extent than edema toxin. Deletion of protective antigen resulted in greater loss of virulence after intravenous challenge with bacilli than deletion of lethal toxin or edema toxin alone. These findings are consistent with the ability of anti-protective antigen antibodies to prevent anthrax and suggest that lethal factor is the dominant toxin that contributes to the escape of significant numbers of bacilli from the thoracic cavity to cause anthrax after inhalation challenge with spores.


Assuntos
Antraz/microbiologia , Antígenos de Bactérias/metabolismo , Bacillus anthracis/patogenicidade , Toxinas Bacterianas/metabolismo , Pulmão/microbiologia , Infecções Respiratórias/microbiologia , Animais , Anticorpos Antibacterianos/sangue , Feminino , Macaca , Masculino , Esporos Bacterianos/patogenicidade , Virulência , Fatores de Virulência/metabolismo
5.
Proc Natl Acad Sci U S A ; 109(46): E3168-76, 2012 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-23093667

RESUMO

Autophagy is a cell biological pathway affecting immune responses. In vitro, autophagy acts as a cell-autonomous defense against Mycobacterium tuberculosis, but its role in vivo is unknown. Here we show that autophagy plays a dual role against tuberculosis: antibacterial and anti-inflammatory. M. tuberculosis infection of Atg5(fl/fl) LysM-Cre(+) mice relative to autophagy-proficient littermates resulted in increased bacillary burden and excessive pulmonary inflammation characterized by neutrophil infiltration and IL-17 response with increased IL-1α levels. Macrophages from uninfected Atg5(fl/fl) LysM-Cre(+) mice displayed a cell-autonomous IL-1α hypersecretion phenotype, whereas T cells showed propensity toward IL-17 polarization during nonspecific activation or upon restimulation with mycobacterial antigens. Thus, autophagy acts in vivo by suppressing both M. tuberculosis growth and damaging inflammation.


Assuntos
Autofagia/imunologia , Proteínas Associadas aos Microtúbulos/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Animais , Autofagia/genética , Proteína 5 Relacionada à Autofagia , Interleucina-17/imunologia , Interleucina-1alfa/genética , Interleucina-1alfa/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/genética , Infiltração de Neutrófilos/genética , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Linfócitos T/imunologia , Linfócitos T/microbiologia , Tuberculose/genética , Tuberculose/microbiologia
6.
Proc Natl Acad Sci U S A ; 108(2): 739-44, 2011 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-21187383

RESUMO

One of the two essential virulence factors of Bacillus anthracis is the poly-γ-D-glutamic acid (γDPGA) capsule. Five γDPGA-specific antibody antigen-binding fragments (Fabs) were generated from immunized chimpanzees. The two selected for further study, Fabs 11D and 4C, were both converted into full-length IgG1 and IgG3 mAbs having human IgG1 or IgG3 constant regions. These two mAbs had similar binding affinities, in vitro opsonophagocytic activities, and in vivo efficacies, with the IgG1 and IgG3 subclasses reacting similarly. The mAbs bound to γDPGA specifically with estimated binding affinities (K(d)) of 35-70 nM and effective affinities (effective K(d)) of 0.1-0.3 nM. The LD(50) in an opsonophagocytic bactericidal assay was ≈10 ng/mL of 11D or 4C. A single 30-µg dose of either mAb given to BALB/c mice 18 h before challenge conferred about 50% protection against a lethal intratracheal spore challenge by the virulent B. anthracis Ames strain. More importantly, either mAb given 8 h or 20 h after challenge provided significant protection against lethal infection. Thus, these anti-γDPGA mAbs should be useful, alone or in combination with antitoxin mAbs, for achieving a safe and efficacious postexposure therapy for anthrax.


Assuntos
Antraz/prevenção & controle , Antraz/terapia , Anticorpos Monoclonais/química , Bacillus anthracis/metabolismo , Sequência de Aminoácidos , Animais , Antraz/imunologia , Anti-Infecciosos/farmacologia , Humanos , Imunoglobulina G/química , Cinética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Pan troglodytes , Fagocitose , Ligação Proteica , Homologia de Sequência de Aminoácidos , Ressonância de Plasmônio de Superfície
7.
Infect Immun ; 80(7): 2414-25, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22526673

RESUMO

The development of therapeutics against biothreats requires that we understand the pathogenesis of the disease in relevant animal models. The rabbit model of inhalational anthrax is an important tool in the assessment of potential therapeutics against Bacillus anthracis. We investigated the roles of B. anthracis capsule and toxins in the pathogenesis of inhalational anthrax in rabbits by comparing infection with the Ames strain versus isogenic mutants with deletions of the genes for the capsule operon (capBCADE), lethal factor (lef), edema factor (cya), or protective antigen (pagA). The absence of capsule or protective antigen (PA) resulted in complete avirulence, while the presence of either edema toxin or lethal toxin plus capsule resulted in lethality. The absence of toxin did not influence the ability of B. anthracis to traffic to draining lymph nodes, but systemic dissemination required the presence of at least one of the toxins. Histopathology studies demonstrated minimal differences among lethal wild-type and single toxin mutant strains. When rabbits were coinfected with the Ames strain and the PA- mutant strain, the toxin produced by the Ames strain was not able to promote dissemination of the PA- mutant, suggesting that toxigenic action occurs in close proximity to secreting bacteria. Taken together, these findings suggest that a major role for toxins in the pathogenesis of anthrax is to enable the organism to overcome innate host effector mechanisms locally and that much of the damage during the later stages of infection is due to the interactions of the host with the massive bacterial burden.


Assuntos
Antraz/microbiologia , Antraz/patologia , Antígenos de Bactérias/biossíntese , Bacillus anthracis/patogenicidade , Toxinas Bacterianas/biossíntese , Fatores de Virulência/biossíntese , Animais , Antraz/mortalidade , Antígenos de Bactérias/genética , Cápsulas Bacterianas/genética , Toxinas Bacterianas/genética , Modelos Animais de Doenças , Feminino , Deleção de Genes , Histocitoquímica , Coelhos , Análise de Sobrevida , Virulência
8.
Infect Immun ; 79(4): 1770-8, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21282410

RESUMO

Pneumonic tularemia is a life-threatening disease caused by inhalation of the highly infectious intracellular bacterium Francisella tularensis. The most serious form of the disease associated with the type A strains can be prevented in experimental animals through vaccination with the attenuated live vaccine strain (LVS). The protection is largely cell mediated, but the contribution of antibodies remains controversial. We addressed this issue in a series of passive immunization studies in Fischer 344 (F344) rats. Subcutaneous LVS vaccination induced a robust serum antibody response dominated by IgM, IgG2a, and IgG2b antibodies. Prophylactic administration of LVS immune serum or purified immune IgG reduced the severity and duration of disease in naïve rats challenged intratracheally with a lethal dose of the virulent type A strain SCHU S4. The level of resistance increased with the volume of immune serum given, but the maximum survivable SCHU S4 challenge dose was at least 100-fold lower than that shown for LVS-vaccinated rats. Protection correlated with reduced systemic bacterial growth, less severe histopathology in the liver and spleen during the early phase of infection, and bacterial clearance by a T cell-dependent mechanism. Our results suggest that treatment with immune serum limited the sequelae associated with infection, thereby enabling a sterilizing T cell response to develop and resolve the infection. Thus, antibodies induced by LVS vaccination may contribute to the defense of F344 rats against respiratory infection by type A strains of F. tularensis.


Assuntos
Anticorpos Antibacterianos/imunologia , Vacinas Bacterianas/imunologia , Francisella tularensis/imunologia , Imunização Passiva , Infecções Respiratórias/imunologia , Tularemia/imunologia , Tularemia/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/uso terapêutico , Separação Celular , Feminino , Citometria de Fluxo , Ratos , Ratos Endogâmicos F344 , Ratos Nus , Infecções Respiratórias/prevenção & controle , Vacinação , Vacinas Atenuadas/imunologia
9.
J Bacteriol ; 192(2): 400-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19915022

RESUMO

In the current study, we examined the regulatory interactions of a serine/threonine phosphatase (BA-Stp1), serine/threonine kinase (BA-Stk1) pair in Bacillus anthracis. B. anthracis STPK101, a null mutant lacking BA-Stp1 and BA-Stk1, was impaired in its ability to survive within macrophages, and this correlated with an observed reduction in virulence in a mouse model of pulmonary anthrax. Biochemical analyses confirmed that BA-Stp1 is a PP2C phosphatase and dephosphorylates phosphoserine and phosphothreonine residues. Treatment of BA-Stk1 with BA-Stp1 altered BA-Stk1 kinase activity, indicating that the enzymatic function of BA-Stk1 can be influenced by BA-Stp1 dephosphorylation. Using a combination of mass spectrometry and mutagenesis approaches, three phosphorylated residues, T165, S173, and S214, in BA-Stk1 were identified as putative regulatory targets of BA-Stp1. Further analysis found that T165 and S173 were necessary for optimal substrate phosphorylation, while S214 was necessary for complete ATP hydrolysis, autophosphorylation, and substrate phosphorylation. These findings provide insight into a previously undescribed Stp/Stk pair in B. anthracis.


Assuntos
Bacillus anthracis/enzimologia , Bacillus anthracis/patogenicidade , Proteínas de Bactérias/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Virulência/fisiologia , Animais , Bacillus anthracis/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Western Blotting , Linhagem Celular , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Mutagênese Sítio-Dirigida , Proteína Básica da Mielina/metabolismo , Fosfopeptídeos/metabolismo , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Virulência/genética
10.
Infect Immun ; 77(1): 429-35, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18981254

RESUMO

Bacillus anthracis strains harboring virulence plasmid pXO1 that encodes the toxin protein protective antigen (PA), lethal factor, and edema factor and virulence plasmid pXO2 that encodes capsule biosynthetic enzymes exhibit different levels of virulence in certain animal models. In the murine model of pulmonary infection, B. anthracis virulence was capsule dependent but toxin independent. We examined the role of toxins in subcutaneous (s.c.) infections using two different genetically complete (pXO1(+) pXO2(+)) strains of B. anthracis, strains Ames and UT500. Similar to findings for the pulmonary model, toxin was not required for infection by the Ames strain, because the 50% lethal dose (LD(50)) of a PA-deficient (PA(-)) Ames mutant was identical to that of the parent Ames strain. However, PA was required for efficient s.c. infection by the UT500 strain, because the s.c. LD(50) of a UT500 PA(-) mutant was 10,000-fold higher than the LD(50) of the parent UT500 strain. This difference between the Ames strain and the UT500 strain could not be attributed to differences in spore coat properties or the rate of germination, because s.c. inoculation with the capsulated bacillus forms also required toxin synthesis by the UT500 strain to cause lethal infection. The toxin-dependent phenotype of the UT500 strain was host phagocyte dependent, because eliminating Gr-1(+) phagocytes restored virulence to the UT500 PA(-) mutant. These experiments demonstrate that the dominant virulence factors used to establish infection by B. anthracis depend on the route of inoculation and the bacterial strain.


Assuntos
Antraz/microbiologia , Antraz/patologia , Bacillus anthracis/patogenicidade , Fatores de Virulência/fisiologia , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/fisiologia , Toxinas Bacterianas/genética , Feminino , Dose Letal Mediana , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fagócitos/imunologia , Fagócitos/microbiologia , Análise de Sobrevida , Virulência , Fatores de Virulência/genética
11.
Infect Immun ; 77(5): 2010-21, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19237526

RESUMO

Parenteral and respiratory vaccinations with the intracellular bacterium Francisella tularensis have been studied using the live vaccine strain (LVS) in a mouse model, and spleen cells from immune mice are often used for immunological studies. However, mechanisms of host immunological responses may be different in nonlymphoid organs that are important sites of infection, such as lung and liver. Using parenteral (intradermal) or respiratory (cloud aerosol) vaccination, here we examine the functions of resulting LVS-immune liver or lung cells, respectively. Surprisingly, LVS was considerably more virulent when administered by cloud aerosol than by intranasal instillation, suggesting method-dependent differences in initial localization and/or dissemination patterns. Only low doses were sublethal, and resolution of sublethal cloud aerosol infection was dependent on gamma interferon (IFN-gamma), tumor necrosis factor alpha, and inducible nitric oxide synthase. Nonetheless, survival of cloud aerosol or parenteral infection resulted in the development of a protective immune response against lethal LVS intraperitoneal or aerosol challenge, reflecting development of systemic secondary immunity in both cases. Such immunity was further detected by directly examining the functions of LVS-immune lung or liver lymphocytes in vitro. Lung lymphocytes primed by respiratory infection, as well as liver lymphocytes primed by parenteral infection, clearly controlled in vitro intracellular bacterial growth primarily via mechanisms that were not dependent on IFN-gamma activity. Thus, our results indicate functional similarities between immune T cells residing in spleens, livers, and lungs of LVS-immune mice.


Assuntos
Vacinas Bacterianas/imunologia , Francisella tularensis/imunologia , Fígado/imunologia , Pulmão/imunologia , Linfócitos T/imunologia , Tularemia/prevenção & controle , Animais , Contagem de Colônia Microbiana , Feminino , Francisella tularensis/crescimento & desenvolvimento , Interferon gama/deficiência , Interferon gama/imunologia , Fígado/microbiologia , Pulmão/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Baço/microbiologia , Análise de Sobrevida
12.
Infect Immun ; 77(4): 1649-63, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19168734

RESUMO

Bacillus anthracis is the causative agent of anthrax. We have developed a novel whole-bacterial-cell anthrax vaccine utilizing B. anthracis that is killed but metabolically active (KBMA). Vaccine strains that are asporogenic and nucleotide excision repair deficient were engineered by deleting the spoIIE and uvrAB genes, rendering B. anthracis extremely sensitive to photochemical inactivation with S-59 psoralen and UV light. We also introduced point mutations into the lef and cya genes, which allowed inactive but immunogenic toxins to be produced. Photochemically inactivated vaccine strains maintained a high degree of metabolic activity and secreted protective antigen (PA), lethal factor, and edema factor. KBMA B. anthracis vaccines were avirulent in mice and induced less injection site inflammation than recombinant PA adsorbed to aluminum hydroxide gel. KBMA B. anthracis-vaccinated animals produced antibodies against numerous anthrax antigens, including high levels of anti-PA and toxin-neutralizing antibodies. Vaccination with KBMA B. anthracis fully protected mice against challenge with lethal doses of toxinogenic unencapsulated Sterne 7702 spores and rabbits against challenge with lethal pneumonic doses of fully virulent Ames strain spores. Guinea pigs vaccinated with KBMA B. anthracis were partially protected against lethal Ames spore challenge, which was comparable to vaccination with the licensed vaccine anthrax vaccine adsorbed. These data demonstrate that KBMA anthrax vaccines are well tolerated and elicit potent protective immune responses. The use of KBMA vaccines may be broadly applicable to bacterial pathogens, especially those for which the correlates of protective immunity are unknown.


Assuntos
Vacinas contra Antraz/imunologia , Antraz/imunologia , Anticorpos Antibacterianos/sangue , Bacillus anthracis , Vacinas de Produtos Inativados/imunologia , Animais , Antraz/microbiologia , Antraz/prevenção & controle , Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/genética , Antígenos de Bactérias/imunologia , Bacillus anthracis/genética , Bacillus anthracis/imunologia , Bacillus anthracis/patogenicidade , Bacillus anthracis/efeitos da radiação , Feminino , Furocumarinas , Cobaias , Imunidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Mutação , Coelhos , Esporos Bacterianos/genética , Raios Ultravioleta , Vacinação , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/genética , Virulência
13.
Hum Immunol ; 69(9): 552-61, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18662733

RESUMO

Bacillus anthracis possesses three primary virulence factors: capsule, lethal toxin (LT), and edema toxin (ET). Dendritic cells (DCs) are critical to innate and acquired immunity and represent potential targets for these factors. We examined the ability of B. anthracis spores and bacilli to stimulate human monocyte-derived DC (MDDC), primary myeloid DC (mDC), and plasmacytoid DC (pDC) cytokine secretion. Exposure of MDDCs and mDCs to spores or vegetative bacilli of the genetically complete strain UT500 induced significantly increased cytokine secretion. Spores lacking genes required for capsule biosynthesis stimulated significantly higher cytokine secretion than UT500 spores from mDCs, but not MDDCs. In contrast, bacilli lacking capsule stimulated significantly higher cytokine secretion than UT500 bacilli in both MDDCs and mDCs. Spores or bacilli lacking both LT and ET stimulated significantly higher cytokine secretion than UT500 spores or bacilli, respectively, in both mDCs and MDDCs. pDCs exposed to spores or bacilli did not produce significant amounts of cytokines even when virulence factors were absent. In conclusion, B. anthracis employs toxins as well as capsule to inhibit human MDDC and mDC cytokine secretion, whereas human pDCs respond poorly even when capsule or both toxins are absent.


Assuntos
Bacillus anthracis/patogenicidade , Citocinas/metabolismo , Células Dendríticas/imunologia , Esporos Bacterianos/patogenicidade , Fatores de Virulência/imunologia , Bacillus anthracis/metabolismo , Citocinas/biossíntese , Citocinas/imunologia , Células Dendríticas/metabolismo , Humanos , Esporos Bacterianos/metabolismo , Fatores de Virulência/metabolismo
14.
Protein J ; 27(5): 292-302, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18649128

RESUMO

This article reports the design of a bivalent protein ligand with dual use in therapy and diagnosis of anthrax caused by Bacillus anthracis. The ligand specifically binds to PA and thereby blocks the intracellular delivery of LF and EF toxins that, respectively, cause cell lysis and edema. The ligand is a chimeric scaffold with two PA-binding domains (called VWA) linked to an IgG-Fc frame. Molecular modeling and binding measurements reveal that the VWA-Fc dimer binds to PA with high affinity (K(D)=0.2 nM). An in vitro bio-luminescence assay shows that VWA-Fc (at nanomolar concentration) protects mouse macrophages from lysis by PA/LF. In vivo studies demonstrate that VWA-Fc at low doses (approximately 50 microg/animal) are able to rescue animals from lethal doses of PA/LF and B. anthracis spores. Finally, VWA-Fc is utilized as the capture molecule in the sensitive (down to 30 picomolar) detection of PA using surface plasmon resonance.


Assuntos
Antraz/diagnóstico , Antraz/terapia , Antígenos de Bactérias/química , Antígenos de Bactérias/metabolismo , Animais , Antígenos de Bactérias/genética , Linhagem Celular , Humanos , Ligantes , Camundongos , Modelos Moleculares , Estrutura Quaternária de Proteína , Sensibilidade e Especificidade
15.
Infect Immun ; 75(12): 5726-34, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17923523

RESUMO

Bacillus anthracis transitions from a dormant spore to a vegetative bacillus through a series of structural and biochemical changes collectively referred to as germination. The timing of germination is important during early steps in infection and may determine if B. anthracis survives or succumbs to responsive macrophages. In the current study experiments determined the contribution of endogenous D-alanine production to the efficiency and timing of B. anthracis spore germination under in vitro and in vivo conditions. Racemase-mediated production of endogenous D-alanine by B. anthracis altered the kinetics for initiation of germination over a range of spore densities and exhibited a threshold effect wherein small changes in spore number resulted in major changes in germination efficiency. This threshold effect correlated with D-alanine production, was prevented by an alanine racemase inhibitor, and required L-alanine. Interestingly, endogenous production of inhibitory levels of D-alanine was detected under experimental conditions that did not support germination and in a germination-deficient mutant of B. anthracis. Racemase-dependent production of D-alanine enhanced survival of B. anthracis during interaction with murine macrophages, suggesting a role for inhibition of germination during interaction with these cells. Finally, in vivo experiments revealed an approximately twofold decrease in the 50% lethal dose of B. anthracis spores administered in the presence of D-alanine, indicating that rates of germination may be directly influenced by the levels of this amino acid during early stages of disease.


Assuntos
Alanina/biossíntese , Antraz/metabolismo , Antraz/microbiologia , Bacillus anthracis/fisiologia , Alanina/antagonistas & inibidores , Alanina Racemase/química , Animais , Bacillus anthracis/química , Bacillus anthracis/metabolismo , Feminino , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Esporos Bacterianos/química , Esporos Bacterianos/crescimento & desenvolvimento , Esporos Bacterianos/metabolismo , Estereoisomerismo , Virulência
16.
Respir Res ; 8: 24, 2007 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-17352829

RESUMO

BACKGROUND: The resolution of inflammatory responses in the lung has not been described in detail and the role of specific cytokines influencing the resolution process is largely unknown. METHODS: The present study was designed to describe the resolution of inflammation from 3 h through 90 d following an acute injury by a single intratracheal instillation of F344/N rats with LPS. We documented the inflammatory cell types and cytokines found in the bronchoalveolar lavage fluid (BALF), and epithelial changes in the axial airway and investigated whether IL-18 may play a role in the resolution process by reducing its levels with anti-IL-18 antibodies. RESULTS: Three major stages of inflammation and resolution were observed in the BALF during the resolution. The first stage was characterized by PMNs that increased over 3 h to 1 d and decreased to background levels by d 6-8. The second stage of inflammation was characterized by macrophage influx reaching maximum numbers at d 6 and decreasing to background levels by d 40. A third stage of inflammation was observed for lymphocytes which were elevated over d 3-6. Interestingly, IL-18 and IL-9 levels in the BALF showed a cyclic pattern with peak levels at d 4, 8, and 16 while decreasing to background levels at d 1-2, 6, and 12. Depletion of IL-18 caused decreased PMN numbers at d 2, but no changes in inflammatory cell number or type at later time points. CONCLUSION: These data suggest that IL-18 plays a role in enhancing the LPS-induced neutrophilic inflammation of the lung, but does not affect the resolution of inflammation.


Assuntos
Células Caliciformes/patologia , Interleucina-18/fisiologia , Lipopolissacarídeos/toxicidade , Pulmão/patologia , Infiltração de Neutrófilos/imunologia , Mucosa Respiratória/patologia , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Células Caliciformes/efeitos dos fármacos , Células Caliciformes/imunologia , Hiperplasia , Inflamação/etiologia , Inflamação/imunologia , Inflamação/patologia , Pulmão/imunologia , Masculino , Ratos , Ratos Endogâmicos F344 , Mucosa Respiratória/imunologia
17.
Comput Biol Med ; 37(11): 1539-52, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17433283

RESUMO

Ideally biosignatures can be detected at the early infection phase and used both for developing diagnostic patterns and for prognostic triage. Such biosignatures are important for vaccine validation and to provide risk stratification to a population such as for the identification of individuals who are exposed to biological or chemical agents and who are at high risk for developing an infection. The research goal is to detect broad based biosignature models and is initially focused on developing effective computer-augmented pathology tied to animal models developed at the University of New Mexico (UNM). Using lung tissue from infected and nai ve mice, feature extraction from images of the tissue under a specialized microscope, and Bayesian networks to analyze the data sets of features, we were able to differentiate normal from diseased samples and viral from bacterial samples in mid to late stages of infection. This effort has shown the potential effectiveness of computer-augmented pathology in this application. The extended research intends to couple analysis of serum, microarray analysis of organs, proteomic data and the pathology. The rational for the current invasive procedure on animal models is to facilitate the development of data analysis and machine learning techniques that can eventually be generalized to the task of discovering non-invasive and early stage biosignatures for human models.


Assuntos
Inteligência Artificial , Simulação por Computador , Diagnóstico por Computador , Infecções/diagnóstico , Algoritmos , Animais , Teorema de Bayes , Humanos , Interpretação de Imagem Assistida por Computador , Infecções/classificação , Pulmão/anatomia & histologia , Pneumopatias/diagnóstico , Camundongos
18.
DNA Cell Biol ; 25(11): 608-16, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17132091

RESUMO

Yersinia pestis, the causative agent of plague, can be transmitted by infected flea bite or inhaled aerosol. Both routes of infection have a high mortality rate, and pneumonic infections of Y. pestis represent a significant concern as a tool of bioterrorism. Understanding the transcriptional program of this pathogen during pulmonary infection should be valuable in understanding plague pathogenesis, as well as potentially offering insights into new vaccines and therapeutics. Toward this goal we developed a long oligonucleotide microarray to the plague bacillus and evaluated the expression profiles of Y. pestis in vitro and in the mouse pulmonary infection model in vivo. The in vitro analysis compared expression patterns at 27 versus 37 degrees C, as a surrogate of the transition from the flea to the mammalian host. The in vivo analysis used intranasal challenge to the mouse lung. By amplifying the Y. pestis RNA from individual mouse lungs we were able to map the transcriptional profile of plague at postinfection days 1 to 3. Our data present a very different transcriptional profile between in vivo and in vitro expression, suggesting Y. pestis responds to a variety of host signals during infection. Of note was the number of genes found in genomic regions with altered %GC content that are upregulated within the mouse lung environment. These data suggest these regions may provide particularly promising targets for both vaccines and therapeutics.


Assuntos
Perfilação da Expressão Gênica/métodos , Pulmão/microbiologia , Peste/genética , Peste/microbiologia , Yersinia pestis/genética , Administração Intranasal , Animais , Composição de Bases , Bioterrorismo , Gatos , Contagem de Colônia Microbiana , Ilhas Genômicas/genética , Fígado/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos , Vacina contra a Peste , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/microbiologia , Temperatura , Transcrição Gênica , Yersinia pestis/crescimento & desenvolvimento
19.
Biotechniques ; 37(4): 654-8, 660, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15517977

RESUMO

There has been increasing interest and efforts devoted to developing biosensor technologies for identifying pathogens, particularly in the biothreat area. In this study, a universal set of short 12- and 13-mer oligonucleotide probes was derived independently of a priori genomic sequence information and used to generate unique species-dependent genomic hybridization signatures. The probe set sequences were algorithmically generated to be maximally distant in sequence space and not dependent on the sequence of any particular genome. The probe set is universally applicable because it is unbiased and independent of hybridization predictions based upon simplified assumptions regarding probe-target duplex formation from linear sequence analysis. Tests were conducted on microarrays containing 14,283 unique probes synthesized using an in situ light-directed synthesis methodology. The genomic DNA hybridization intensity patterns reproducibly differentiated various organisms (Bacillus subtilis, Yersinia pestis, Streptococcus pneumonia, Bacillus anthracis, and Homo sapiens), including the correct identification of a blinded "unknown" sample. Applications of this method include not only pathological and forensic genome identification in medicine and basic science, but also potentially a novel method for the discovery of unknown targets and associations inherent in dynamic nucleic acid populations such as represented by differential gene expression.


Assuntos
Sondas de DNA , DNA Bacteriano/genética , Perfilação da Expressão Gênica/métodos , Análise de Sequência com Séries de Oligonucleotídeos/instrumentação , Sondas de Oligonucleotídeos , Bacillus anthracis/genética , Bacillus subtilis/genética , Bioterrorismo , Perfilação da Expressão Gênica/instrumentação , Dados de Sequência Molecular , Hibridização de Ácido Nucleico/métodos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Streptococcus pneumoniae/genética , Yersinia pestis/genética
20.
Virology ; 412(2): 411-25, 2011 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-21334039

RESUMO

Orthopoxviruses encode multiple proteins that modulate host immune responses. We determined whether cowpox virus (CPXV), a representative orthopoxvirus, modulated innate and acquired immune functions of human primary myeloid DCs and plasmacytoid DCs and monocyte-derived DCs (MDDCs). A CPXV infection of DCs at a multiplicity of infection of 10 was nonproductive, altered cellular morphology, and failed to reduce cell viability. A CPXV infection of DCs did not stimulate cytokine or chemokine secretion directly, but suppressed toll-like receptor (TLR) agonist-induced cytokine secretion and a DC-stimulated mixed leukocyte reaction (MLR). LPS-stimulated NF-κB nuclear translocation and host cytokine gene transcription were suppressed in CPXV-infected MDDCs. Early viral immunomodulatory genes were upregulated in MDDCs, consistent with early DC immunosuppression via synthesis of intracellular viral proteins. We conclude that a nonproductive CPXV infection suppressed DC immune function by synthesizing early intracellular viral proteins that suppressed DC signaling pathways.


Assuntos
Vírus da Varíola Bovina/imunologia , Vírus da Varíola Bovina/patogenicidade , Células Dendríticas/imunologia , Células Dendríticas/virologia , Evasão da Resposta Imune , Sobrevivência Celular , Células Cultivadas , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Humanos , NF-kappa B/antagonistas & inibidores , NF-kappa B/imunologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa