Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
2.
PLoS Pathog ; 8(7): e1002817, 2012.
Article in English | MEDLINE | ID: mdl-22911267

ABSTRACT

Type I interferons (IFN-I) broadly control innate immunity and are typically transcriptionally induced by Interferon Regulatory Factors (IRFs) following stimulation of pattern recognition receptors within the cytosol of host cells. For bacterial infection, IFN-I signaling can result in widely variant responses, in some cases contributing to the pathogenesis of disease while in others contributing to host defense. In this work, we addressed the role of type I IFN during Yersinia pestis infection in a murine model of septicemic plague. Transcription of IFN-ß was induced in vitro and in vivo and contributed to pathogenesis. Mice lacking the IFN-I receptor, Ifnar, were less sensitive to disease and harbored more neutrophils in the later stage of infection which correlated with protection from lethality. In contrast, IRF-3, a transcription factor commonly involved in inducing IFN-ß following bacterial infection, was not necessary for IFN production but instead contributed to host defense. In vitro, phagocytosis of Y. pestis by macrophages and neutrophils was more effective in the presence of IRF-3 and was not affected by IFN-ß signaling. This activity correlated with limited bacterial growth in vivo in the presence of IRF-3. Together the data demonstrate that IRF-3 is able to activate pathways of innate immunity against bacterial infection that extend beyond regulation of IFN-ß production.


Subject(s)
Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Interferon-beta/metabolism , Plague/immunology , Plague/metabolism , Yersinia pestis/immunology , Animals , Female , Interferon Regulatory Factor-3/immunology , Interferon Type I/immunology , Interferon-beta/genetics , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Signal Transduction , Transcription, Genetic
3.
Am J Pathol ; 178(3): 1190-200, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21356370

ABSTRACT

Pulmonary infection by Yersinia pestis causes pneumonic plague, a necrotic bronchopneumonia that is rapidly lethal and highly contagious. Acute pneumonic plague accompanies the up-regulation of pro-inflammatory cytokines and chemokines, suggesting that the host innate immune response may contribute to the development of disease. To address this possibility, we sought to understand the consequences of neutrophil recruitment during pneumonic plague, and we studied the susceptibility of C3H-HeN mice lacking the CXC chemokine KC or its receptor CXC receptor 2 (CXCR2) to pulmonary Y. pestis infection. We found that without Kc or Cxcr2, disease progression was accelerated both in bacterial growth and development of primary bronchopneumonia. When examined in an antibody clearance model, Cxcr2(-/-) mice were not protected by neutralizing Y. pestis antibodies, yet bacterial growth in the lungs was delayed in a manner associated with a neutrophil-mediated inflammatory response. After this initial delay, however, robust neutrophil recruitment in Cxcr2(-/-) mice correlated with bacterial growth and the development of fulminant pneumonic and septicemic plague. In contrast, attenuated Y. pestis lacking the conserved pigmentation locus could be cleared from the lungs in the absence of Cxcr2 indicating virulence factors within this locus may inhibit CXCR2-independent pathways of bacterial killing. Together, the data suggest CXCR2 uniquely induces host defense mechanisms that are effective against virulent Y. pestis, raising new insight into the activation of neutrophils during infection.


Subject(s)
Neutrophil Infiltration/immunology , Plague/immunology , Plague/microbiology , Receptors, Interleukin-8B/metabolism , Yersinia pestis/immunology , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Chemokine CXCL1/metabolism , Disease Models, Animal , Disease Progression , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Models, Immunological , Mutation/genetics , Plague/prevention & control , Pore Forming Cytotoxic Proteins/immunology , Signal Transduction , Yersinia pestis/growth & development
4.
Infect Immun ; 78(1): 220-30, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19841077

ABSTRACT

Yersinia pestis causes primary pneumonic plague in many mammalian species, including humans, mice, and rats. Virulent Y. pestis strains undergo frequent spontaneous deletion of a 102-kb chromosomal DNA fragment, known as the pigmentation (pgm) locus, when grown in laboratory media, yet this locus is present in every virulent isolate. The pgm locus encodes, within a high-pathogenicity island, siderophore biosynthesis genes that are required for growth in the mammalian host when inoculated by peripheral routes. Recently, higher challenge doses of nonpigmented Y. pestis were reported to cause fatal pneumonic plague in mice, suggesting a useful model for studies of virulence and immunity. In this work, we show that intranasal infection of BALB/c mice with nonpigmented Yersinia pestis does not result in pneumonic plague. Despite persistent bacterial colonization of the lungs and the eventual death of infected mice, pulmonary inflammation was generally absent, and there was no disease pathology characteristic of pneumonic plague. Iron given to mice at the time of challenge, previously shown to enhance the virulence of pgm-deficient strains, resulted in an accelerated disease course, with less time to bacteremia and lethality, but lung inflammation and pneumonia were still absent. We examined other rodent models and found differences in lung inflammatory responses, some of which led to clearance and survival even when high challenge doses were used. Together, the results suggest that the Y. pestis pgm locus encodes previously unappreciated virulence factors required for the induction of pneumonic plague that are independent of iron scavenging from the mammalian host.


Subject(s)
Bacterial Proteins/metabolism , Inflammation/pathology , Plague/microbiology , Yersinia pestis/classification , Yersinia pestis/pathogenicity , Animals , Bacterial Proteins/genetics , Female , Gene Expression Regulation, Bacterial/physiology , Inflammation/microbiology , Iron/pharmacology , Mice , Mice, Inbred Strains , Pigmentation , Plague/pathology , Rats , Rats, Inbred Strains , Sepsis/microbiology , Sepsis/pathology , Time Factors , Virulence , Yersinia pestis/genetics , Yersinia pestis/physiology
5.
Am J Pathol ; 174(3): 910-21, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19164505

ABSTRACT

The Brown Norway rat was recently described as a bubonic plague model that closely mimics human disease. We therefore evaluated the Brown Norway rat as an alternative small animal model for pneumonic plague and characterized both the efficacy and potency of vaccine candidates. When infected by intranasal instillation, these rats rapidly developed fatal pneumonic plague within 2 to 4 days of infection. Plague disease was characterized by severe alveolar edema and vascular hemorrhage in the lung in addition to fulminant necrotizing pneumonia caused by massive bacterial replication and inflammation. Twenty-four hours before death, animals developed systemic disease with an apparent delayed inflammatory response. We evaluated the ability of the protective antigen, LcrV, and a mutant derivative, V10, to protect these rats from pneumonic plague. Both were highly effective vaccines because complete protection was observed at challenge doses of 7500 LD(50). Antibody analyses suggested stronger potency of V10 immune sera compared with LcrV in the passive transfer of immunity to bubonic plague, with multiple neutralizing epitopes in LcrV. Taken together, these data demonstrate the effectiveness of inhibiting type III secretion in the prevention of pneumonic plague in rats and reveal critical contributions from both the cellular and humoral immune systems. Thus, the Brown Norway rat is an appealing alternative small animal model for the study of pneumonic plague pathogenesis and immunity.


Subject(s)
Plague/immunology , Plague/pathology , Animals , Bacterial Vaccines , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunization, Passive , Inflammation/immunology , Inflammation/pathology , Intradermal Tests , Lethal Dose 50 , Rats , Rats, Inbred BN , Yersinia pestis/genetics , Yersinia pestis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...