Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Clin Vaccine Immunol ; 24(1)2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27760780

RESUMO

Adenylate cyclase toxin (ACT) is an essential virulence factor of Bordetella pertussis, and antibodies to ACT protect against B. pertussis infection in mice. The toxin is therefore a strong candidate antigen for addition to future acellular pertussis vaccines. In order to characterize the functionality of the immunologic response to ACT after infection, we developed an assay for testing the ability of serum samples from subjects infected with B. pertussis to neutralize ACT-induced cytotoxicity in J774 macrophage cells. Baboons develop neutralizing anti-ACT antibodies following infection with B. pertussis, and all sera from baboons with positive anti-ACT IgG enzyme-linked immunosorbent assay (ELISA) results neutralized ACT cytotoxicity. The toxin neutralization assay (TNA) was positive in some baboon sera in which ELISA remained negative. Of serum samples obtained from humans diagnosed with pertussis by PCR, anti-ACT IgG ELISA was positive in 72%, and TNA was positive in 83%. All samples positive for anti-ACT IgG ELISA were positive by TNA, and none of the samples from humans without pertussis neutralized toxin activity. These findings indicate that antibodies to ACT generated following infection with B. pertussis consistently neutralize toxin-induced cytotoxicity and that TNA can be used to improve understanding of the immunologic response to ACT after infection or vaccination.


Assuntos
Toxina Adenilato Ciclase/imunologia , Formação de Anticorpos , Bordetella pertussis/imunologia , Testes de Neutralização/métodos , Coqueluche/imunologia , Adolescente , Adulto , Animais , Linhagem Celular , Sobrevivência Celular , Criança , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Macrófagos/fisiologia , Masculino , Camundongos , Pessoa de Meia-Idade , Papio , Adulto Jovem
2.
Infect Immun ; 81(4): 1306-15, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23381997

RESUMO

Staphylococcus aureus is one of the most common etiological agents of community-acquired skin and soft tissue infection (SSTI). Although the majority of S. aureus community-acquired SSTIs are uncomplicated and self-clearing in nature, some percentage of these cases progress into life-threatening invasive infections. Current animal models of S. aureus SSTI suffer from two drawbacks: these models are a better representation of hospital-acquired SSTI than community-acquired SSTI, and they involve methods that are difficult to replicate. For these reasons, we sought to develop a murine model of community-acquired methicillin-resistant S. aureus SSTI (CA-MRSA SSTI) that can be consistently reproduced with a high degree of precision. We utilized this model to begin to characterize the host immune response to this type of infection. We infected mice via epicutaneous challenge of the skin on the outer ear pinna using Morrow-Brown allergy test needles coated in S. aureus USA300. When mice were challenged in this model, they developed small, purulent, self-clearing lesions with predictable areas of inflammation that mimicked a human infection. CFU in the ear pinna peaked at day 7 before dropping by day 14. The T(h)1 and T(h)17 cytokines gamma interferon (IFN-γ), interleukin-12 (IL-12) p70, tumor necrosis factor alpha (TNF-α), IL-17A, IL-6, and IL-21 were all significantly increased in the draining lymph node of infected mice, and there was neutrophil recruitment to the infection site. In vivo neutrophil depletion demonstrated that neutrophils play a protective role in preventing bacterial dissemination and fatal invasive infection.


Assuntos
Infecções Comunitárias Adquiridas/microbiologia , Infecções Comunitárias Adquiridas/patologia , Modelos Animais de Doenças , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Infecções Cutâneas Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/patologia , Animais , Carga Bacteriana , Citocinas/análise , Citocinas/imunologia , Orelha Externa/microbiologia , Orelha Externa/patologia , Feminino , Linfonodos/química , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/imunologia , Pele/microbiologia , Pele/patologia , Fatores de Tempo
3.
Infect Immun ; 80(9): 3189-93, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22753373

RESUMO

Inhalational anthrax is caused by the sporulating bacterium Bacillus anthracis. A current model for progression in mammalian hosts includes inhalation of bacterial spores, phagocytosis of spores in the nasal mucosa-associated lymphoid tissue (NALT) and lungs by macrophages and dendritic cells, trafficking of phagocytes to draining lymph nodes, germination of spores and multiplication of vegetative bacteria in the NALT and lymph nodes, and dissemination of bacteria via the bloodstream to multiple organs. In previous studies, the kinetics of infection varied greatly among mice, leading us to hypothesize the existence of a bottleneck past which very few spores (perhaps only one) progress to allow the infection to proceed. To test this hypothesis, we engineered three strains of B. anthracis Sterne, each marked with a different fluorescent protein, enabling visual differentiation of strains grown on plates. Mice were infected with a mixture of the three strains, the infection was allowed to proceed, and the strains colonizing the organs were identified. Although the inoculum consisted of approximately equal numbers of each of the three strains, the distal organs were consistently colonized by a majority of only one of the three strains, with the dominant strain varying among animals. Such dominance of one strain over the other two was also found at early time points in the cervical lymph nodes but not in the mediastinal lymph nodes. These results support the existence of a bottleneck in the infectious process.


Assuntos
Antraz/patologia , Bacillus anthracis/patogenicidade , Animais , Bacillus anthracis/classificação , Bacillus anthracis/isolamento & purificação , Sangue/microbiologia , Células Dendríticas/microbiologia , Modelos Animais de Doenças , Humanos , Inalação , Linfonodos/microbiologia , Macrófagos/microbiologia , Camundongos , Esporos Bacterianos/patogenicidade , Coloração e Rotulagem
4.
PLoS One ; 7(5): e37610, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22662179

RESUMO

Moraxella catarrhalis is a gram negative bacterium and a leading causative agent of otitis media (OM) in children. Several recent reports have provided strong evidence for an association between toll like receptors and OM. It has been found that both Streptococcus pneumoniae and nontypeable Haemophilus influenzae activate host protective immune responses through toll like receptors (TLRs), however, the precise mechanism by which Moraxella catarrhalis initiates the host immune response is currently unknown. In this report, using murine macrophages generated from a series of knock-out mice, we have demonstrated that M. catarrhalis lipooligosaccharide (LOS) and either heat killed or live bacteria are recognized by one or more TLRs. LOS activates the host immune response through a membrane bound CD14-TLR4 complex, while both heat killed and live M.cat require recognition by multiple toll like receptors such as TLR2, TLR4 and TLR9 without the requirement of CD14. We have also shown that M.cat stimuli are capable of triggering the host innate immune response by both MyD88- and TRIF- dependent signaling pathways. We further showed that M.cat induced activation of mitogen activated protein kinase (MAPK) is essential in order to achieve optimal secretion of pro-inflammatory cytokine TNF-α. We finally showed that TLR4 mutant C3H/HeJ mice produce significantly lower levels of pro-inflammatory cytokines TNF-α and IL-6 in vivo, An increased bacterial loads at 12 and 24 hours (P<0.001) in their lungs upon challenge with live M.cat in an aerosol chamber compared to wild-type (WT) control mice. These data suggest that TLRs are crucial for an effective innate immune response induced by M.cat. The results of these studies contribute to an increased understanding of molecular mechanism and possible novel treatment strategies for diseases caused by M.cat by specifically targeting TLRs and their signaling pathways.


Assuntos
Pulmão/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Moraxella catarrhalis/imunologia , Receptor 4 Toll-Like/genética , Receptores Toll-Like/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Citocinas/metabolismo , Feminino , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/imunologia , Lipopolissacarídeos/imunologia , Pulmão/metabolismo , Pulmão/microbiologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Mutação , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/genética
5.
J Immunol ; 188(3): 1469-78, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22205026

RESUMO

The complement system, especially the alternative pathway, plays essential roles in the induction of injury in collagen Ab-induced arthritis (CAIA) in mice. The goal of the current study was to directly compare the roles of receptors for C3a and C5a, as well as the membrane attack complex, as effector mechanisms in the pathogenesis of CAIA. Clinical disease activity in C3aR(-/-), C5aR(-/-), and C6-deficient (C6-def) mice was decreased by 52, 94, and 65%, respectively, as compared with wild-type mice. Decreases in histopathologic injury as well as in IgG and C3 deposition paralleled the clinical disease activity. A decrease in the percentage of synovial neutrophils was observed in C3aR(-/-), C5aR(-/-), and C6-def mice, and a decrease in macrophages was observed in C3aR(-/-) and C5aR(-/-), but not in C6-def, mice. Synovial mRNA obtained by laser capture microdissection exhibited a decrease in TNF-α in C5aR(-/-) mice and in IL-1ß in both C5aR(-/-) and C6-def mice, whereas C3aR(-/-) mice demonstrated no change in either cytokine. Our findings show that absent C3aR-, C5aR-, or membrane attack complex-initiated effector mechanisms each decrease susceptibility to CAIA, with clinical effects most pronounced in C5aR-deficient mice. Although the absence of C3aR, C5aR, or C6 led to differential deficiencies in effector mechanisms, decreased proximal joint IgG and C3 deposition was common to all three genotypes in comparison with wild-type mice. These data suggest the existence of positive-feedback amplification pathways downstream of all three effectors that promote additional IgG deposition and C3 activation in the joint.


Assuntos
Artrite Experimental/etiologia , Complemento C6/deficiência , Receptor da Anafilatoxina C5a/fisiologia , Receptores de Complemento/fisiologia , Animais , Ativação do Complemento , Complemento C3a/imunologia , Complemento C6/fisiologia , Citocinas , Suscetibilidade a Doenças , Imunoglobulina G , Macrófagos/patologia , Camundongos , Camundongos Knockout , Neutrófilos/patologia , Líquido Sinovial/imunologia
6.
Infect Immun ; 78(6): 2418-28, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20308305

RESUMO

During advanced stages of inhalation anthrax, Bacillus anthracis accumulates at high levels in the bloodstream of the infected host. This bacteremia leads to sepsis during late-stage anthrax; however, the mechanisms through which B. anthracis-derived factors contribute to the pathology of infected hosts are poorly defined. Peptidoglycan, a major component of the cell wall of Gram-positive bacteria, can provoke symptoms of sepsis in animal models. We have previously shown that peptidoglycan of B. anthracis can induce the production of proinflammatory cytokines by cells in human blood. Here, we show that biologically active peptidoglycan is shed from an active culture of encapsulated B. anthracis strain Ames in blood. Peptidoglycan is able to bind to surfaces of responding cells, and internalization of peptidoglycan is required for the production of inflammatory cytokines. We also show that the peptidoglycan traffics to lysosomes, and lysosomal function is required for cytokine production. We conclude that peptidoglycan of B. anthracis is initially bound by an unknown extracellular receptor, is phagocytosed, and traffics to lysosomes, where it is degraded to a product recognized by an intracellular receptor. Binding of the peptidoglycan product to the intracellular receptor causes a proinflammatory response. These findings provide new insight into the mechanism by which B. anthracis triggers sepsis during a critical stage of anthrax disease.


Assuntos
Bacillus anthracis/imunologia , Citocinas/imunologia , Lisossomos/metabolismo , Peptidoglicano/imunologia , Fagocitose , Animais , Sangue/microbiologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovinos
7.
Infect Immun ; 77(10): 4529-37, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19620350

RESUMO

Toll-like receptors and Nod-like receptors (NLR) play an important role in sensing invading microorganisms for pathogen clearance and eliciting adaptive immunity for protection against rechallenge. Nod1 and Nod2, members of the NLR family, are capable of detecting bacterial peptidoglycan motifs in the host cytosol for triggering proinflammatory cytokine production. In the current study, we sought to determine if Nod1/Nod2 are involved in sensing Bacillus anthracis infection and eliciting protective immune responses. Using mice deficient in both Nod1 and Nod2 proteins, we showed that Nod1/Nod2 are involved in detecting B. anthracis for production of tumor necrosis factor alpha, interleukin-1 alpha (IL-1 alpha), IL-1 beta, CCL5, IL-6, and KC. Proinflammatory responses were higher when cells were exposed to viable spores than when they were exposed to irradiated spores, indicating that recognition of vegetative bacilli through Nod1/Nod2 is significant. We also identify a critical role for Nod1/Nod2 in priming responses after B. anthracis aerosol exposure, as mice deficient in Nod1/Nod2 were impaired in their ability to mount an anamnestic antibody response and were more susceptible to secondary lethal challenge than wild-type mice.


Assuntos
Aerossóis , Antraz/imunologia , Bacillus anthracis/imunologia , Proteína Adaptadora de Sinalização NOD1/imunologia , Proteína Adaptadora de Sinalização NOD2/imunologia , Animais , Anticorpos Antibacterianos/sangue , Citocinas/metabolismo , Camundongos , Camundongos Knockout , Proteína Adaptadora de Sinalização NOD1/deficiência , Proteína Adaptadora de Sinalização NOD2/deficiência , Esporos Bacterianos/imunologia , Análise de Sobrevida
8.
Infect Immun ; 76(5): 2177-82, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18316379

RESUMO

The threat of bioterrorist use of Bacillus anthracis has focused urgent attention on the efficacy and mechanisms of protective immunity induced by available vaccines. However, the mechanisms of infection-induced immunity have been less well studied and defined. We used a combination of complement depletion along with immunodeficient mice and adoptive transfer approaches to determine the mechanisms of infection-induced protective immunity to B. anthracis. B- or T-cell-deficient mice lacked the complete anamnestic protection observed in immunocompetent mice. In addition, T-cell-deficient mice generated poor antibody titers but were protected by the adoptive transfer of serum from B. anthracis-challenged mice. Adoptively transferred sera were protective in mice lacking complement, Fc receptors, or both, suggesting that they operate independent of these effectors. Together, these results indicate that antibody-mediated neutralization provides significant protection in B. anthracis infection-induced immunity.


Assuntos
Anticorpos Antibacterianos/imunologia , Bacillus anthracis/imunologia , Proteínas do Sistema Complemento/imunologia , Receptores Fc/imunologia , Transferência Adotiva , Animais , Anticorpos Antibacterianos/sangue , Linfócitos B/imunologia , Imunização Passiva , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Fc/deficiência , Análise de Sobrevida , Linfócitos T/imunologia
9.
Free Radic Res ; 42(1): 49-56, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18324523

RESUMO

Anthrax is caused by the gram-negative bacterium, Bacillus anthracis. Infection by this microbe results from delivery of the endospore form of the bacillus through direct contact, either topical or inhalation. With regard to the latter route of administration, it is proposed that endospores of B. anthracis enter the lungs and are phagocytized by host alveolar macrophages. Thereafter, it is unclear as to how endospores travel to distal loci and what tissues are the targets. Herein, this study describes the spin labelling of endospores through two different approaches with various aminoxyls. Indeed, after exposure to RAW 264.7 cells, these aminoxyl-containing endospores were phagocytized, as demonstrated by EPR spectroscopy of the infected macrophage, thus providing a potential tool for EPR imaging in animals.


Assuntos
Bacillus anthracis/metabolismo , Óxidos N-Cíclicos , Espectroscopia de Ressonância de Spin Eletrônica , Macrófagos/microbiologia , Maleimidas , Fagocitose , Marcadores de Spin , Coloração e Rotulagem/métodos , Animais , Bacillus anthracis/patogenicidade , Linhagem Celular , Camundongos , Espectroscopia de Infravermelho com Transformada de Fourier , Esporos Bacterianos/metabolismo
10.
Infect Immun ; 73(11): 7535-40, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16239556

RESUMO

Bacillus anthracis is a spore-forming, gram-positive organism that is the causative agent of the disease anthrax. Recognition of Bacillus anthracis by the host innate immune system likely plays a key protective role following infection. In the present study, we examined the role of TLR2, TLR4, and MyD88 in the response to B. anthracis. Heat-killed Bacillus anthracis stimulated TLR2, but not TLR4, signaling in HEK293 cells and stimulated tumor necrosis factor alpha (TNF-alpha) production in C3H/HeN, C3H/HeJ, and C57BL/6J bone marrow-derived macrophages. The ability of heat-killed B. anthracis to induce a TNF-alpha response was preserved in TLR2-/- but not in MyD88-/- macrophages. In vivo studies revealed that TLR2-/- mice and TLR4-deficient mice were resistant to challenge with aerosolized Sterne strain spores but MyD88-/- mice were as susceptible as A/J mice. We conclude that, although recognition of B. anthracis occurs via TLR2, additional MyD88-dependent pathways contribute to the host innate immune response to anthrax infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antígenos de Diferenciação/metabolismo , Bacillus anthracis/imunologia , Receptores Imunológicos/metabolismo , Transdução de Sinais , Esporos Bacterianos/imunologia , Receptor 2 Toll-Like/fisiologia , Receptor 4 Toll-Like/fisiologia , Aerossóis , Animais , Linhagem Celular , Humanos , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética , Fator de Necrose Tumoral alfa/metabolismo
11.
Infect Immun ; 72(11): 6382-9, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15501768

RESUMO

Bacillus anthracis, the etiological agent of anthrax, is a gram-positive, spore-forming bacterium. The inhalational form of anthrax is the most severe and is associated with rapid progression of the disease and the outcome is frequently fatal. Transfer from the respiratory epithelium to regional lymph nodes appears to be an essential early step in the establishment of infection. This transfer is believed to occur by means of carriage within alveolar macrophages following phagocytosis. Therefore, the ability of B. anthracis to transit through the host macrophage or dendritic cell appears to be an early and critical step in B. anthracis pathogenesis. In this work, we examined the cytokine responses to spore infection in mouse primary peritoneal macrophages, in primary human dendritic cells, and during a spore aerosol infection model utilizing the susceptible A/J mouse strain. We demonstrated that both mouse peritoneal macrophages and human dendritic cells exhibited significant intracellular bactericidal activity during the first hours following uptake, providing the necessary time to mount a cytokine response prior to cell lysis. Strong tumor necrosis factor (TNF-alpha) and interleukin-6 (IL-6) responses were seen in mouse peritoneal macrophages. In addition to TNF-alpha and IL-6, human dendritic cells produced the cytokines IL-1beta, IL-8, and IL-12. A mixture of Th1 and Th2 cytokines were detected in sera obtained from infected animals. In this study, we provide further evidence of an acute cytokine response when cells in culture and mice are infected with B. anthracis spores.


Assuntos
Antraz/imunologia , Bacillus anthracis/fisiologia , Bacillus anthracis/patogenicidade , Citocinas/metabolismo , Células Dendríticas/imunologia , Macrófagos Peritoneais/imunologia , Animais , Antraz/microbiologia , Bacillus anthracis/imunologia , Células Cultivadas , Citocinas/sangue , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Interleucina-6/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Esporos Bacterianos/imunologia , Esporos Bacterianos/patogenicidade , Fator de Necrose Tumoral alfa/metabolismo
12.
Infect Immun ; 72(5): 3069-72, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15102824

RESUMO

Herein we report that infection of a murine macrophage cell line with Bacillus anthracis results in the production of tumor necrosis factor alpha and interleukin-12 (IL-12). When infected with B. anthracis spores in combination with lipopolysaccharide, macrophages release increased amounts of IL-12. We found no evidence of inhibition of cytokine responses in macrophages infected with B. anthracis spores.


Assuntos
Bacillus anthracis/imunologia , Bacillus anthracis/patogenicidade , Interleucina-12/biossíntese , Macrófagos/imunologia , Macrófagos/microbiologia , Fator de Necrose Tumoral alfa/biossíntese , Animais , Antraz/etiologia , Antraz/imunologia , Bacillus anthracis/crescimento & desenvolvimento , Linhagem Celular , Lipopolissacarídeos/toxicidade , Camundongos , Esporos Bacterianos/imunologia , Esporos Bacterianos/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA