Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Negl Trop Dis ; 10(6): e0004787, 2016 06.
Article in English | MEDLINE | ID: mdl-27332553

ABSTRACT

Neurocysticercosis (NCC) is one of the most common helminth parasitic diseases of the central nervous system (CNS) and the leading cause of acquired epilepsy worldwide. NCC is caused by the presence of the metacestode larvae of the tapeworm Taenia solium within brain tissues. NCC patients exhibit a long asymptomatic phase followed by a phase of symptoms including increased intra-cranial pressure and seizures. While the asymptomatic phase is attributed to the immunosuppressive capabilities of viable T. solium parasites, release of antigens by dying organisms induce strong immune responses and associated symptoms. Previous studies in T. solium-infected pigs have shown that the inflammatory response consists of various leukocyte populations including eosinophils, macrophages, and T cells among others. Because the role of eosinophils within the brain has not been investigated during NCC, we examined parasite burden, disease susceptibility and the composition of the inflammatory reaction in the brains of infected wild type (WT) and eosinophil-deficient mice (ΔdblGATA) using a murine model of NCC in which mice were infected intracranially with Mesocestoides corti, a cestode parasite related to T. solium. In WT mice, we observed a time-dependent induction of eosinophil recruitment in infected mice, contrasting with an overall reduced leukocyte infiltration in ΔdblGATA brains. Although, ΔdblGATA mice exhibited an increased parasite burden, reduced tissue damage and less disease susceptibility was observed when compared to infected WT mice. Cellular infiltrates in infected ΔdblGATA mice were comprised of more mast cells, and αß T cells, which correlated with an abundant CD8+ T cell response and reduced CD4+ Th1 and Th2 responses. Thus, our data suggest that enhanced inflammatory response in WT mice appears detrimental and associates with increased disease susceptibility, despite the reduced parasite burden in the CNS. Overall reduced leukocyte infiltration due to absence of eosinophils correlates with attenuated tissue damage and longer survival of ΔdblGATA mice. Therefore, our study suggests that approaches to clear NCC will require strategies to tightly control the host immune response while eradicating the parasite with minimal damage to brain tissue.


Subject(s)
Eosinophilia/genetics , Genetic Predisposition to Disease , Leukocytes/physiology , Neurocysticercosis/pathology , Animals , Female , Mast Cells/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurocysticercosis/genetics , Neutrophils/physiology
2.
Infect Immun ; 81(4): 1052-63, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23319563

ABSTRACT

Neurocysticercosis (NCC) is a central nervous system (CNS) infection caused by the metacestode stage of the parasite Taenia solium. During NCC, the parasites release immunodominant glycan antigens in the CNS environment, invoking immune responses. The majority of the associated pathogenesis is attributed to the immune response against the parasites. Glycans from a number of pathogens, including helminths, act as pathogen-associated molecular pattern molecules (PAMPs), which are recognized by pattern recognition receptors (PRRs) known as C-type lectin receptors (CLRs). Using a mouse model of NCC by infection with the related parasite Mesocestoides corti, we have investigated the role of mannose receptor C type 1 (MRC1), a CLR which recognizes high-mannose-containing glycan antigens. Here we show that MRC1(-/-) mice exhibit increased survival times after infection compared with their wild-type (WT) counterparts. The decreased disease severity correlates with reduced levels of expression of markers implicated in NCC pathology, such as interleukin-1ß (IL-1ß), IL-6, CCL5, and matrix metalloproteinase 9 (MMP9), in addition to induction of an important repair marker, fibroblast growth factor 2 (FGF2). Furthermore, the immune cell subsets that infiltrate the brain of MRC1(-/-) mice are dramatically altered and characterized by reduced numbers of T cells and the accumulation of granulocytic cells with an immune phenotype resembling granulocytic myeloid-dependent suppressor cells (gMDSCs). The results suggest that MRC1 plays a critical role in myeloid plasticity, which in turn affects the adaptive immune response and immunopathogenesis during murine NCC.


Subject(s)
Granulocyte Precursor Cells/immunology , Lectins, C-Type/deficiency , Mannose-Binding Lectins/deficiency , Membrane Glycoproteins/deficiency , Mesocestoides/immunology , Neurocysticercosis/immunology , Receptors, Cell Surface/deficiency , Animals , Brain/immunology , Brain/pathology , Cytokines/metabolism , Female , Lectins, C-Type/metabolism , Mannose Receptor , Mannose-Binding Lectins/metabolism , Membrane Glycoproteins/metabolism , Mesocestoides/pathogenicity , Mice , Mice, Inbred C57BL , Neurocysticercosis/mortality , Neurocysticercosis/pathology , Receptors, Cell Surface/metabolism , Receptors, Immunologic , Severity of Illness Index , Survival Analysis
3.
Microb Pathog ; 51(1-2): 39-47, 2011.
Article in English | MEDLINE | ID: mdl-21440052

ABSTRACT

The virulence mechanisms of Francisella tularensis, the causative agent of severe pneumonia in humans and a CDC category A bioterrorism agent, are not fully defined. As sepsis is the leading cause of mortality associated with respiratory infections, we determined whether, in the absence of any known bacterial toxins, a deregulated host response resulting in sepsis syndrome is associated with lethality of respiratory infection with the virulent human Type A strain SchuS4 of F. tularensis. The C57BL/6 mice infected intranasally with a lethal dose of SchuS4 exhibited high bacterial burden in systemic organs and blood indicative of bacteremia. In correlation, infected mice displayed severe tissue pathology and associated cell death in lungs, liver and spleen. Consistent with our studies with murine model strain Francisella novicida, infection with SchuS4 caused an initial delay in upregulation of inflammatory mediators followed by development of severe sepsis characterized by exaggerated cytokine release, upregulation of cardiovascular injury markers and sepsis mediator alarmins S100A9 and HMGB1. This study shows that pulmonary tularemia caused by the Type A strain of F. tularensis results in a deregulated host response leading to severe sepsis and likely represents the major cause of mortality associated with this virulent pathogen.


Subject(s)
Francisella tularensis/pathogenicity , Lung Diseases/complications , Sepsis/pathology , Tularemia/complications , Animals , Bacteremia/pathology , Cytokines/blood , Humans , Inflammation , Liver/microbiology , Liver/pathology , Lung/microbiology , Lung/pathology , Lung Diseases/immunology , Lung Diseases/microbiology , Lung Diseases/pathology , Mice , Mice, Inbred C57BL , Sepsis/etiology , Sepsis/immunology , Sepsis/microbiology , Spleen/microbiology , Spleen/pathology , Tularemia/immunology , Tularemia/microbiology , Tularemia/pathology , Up-Regulation , Virulence
4.
Immunol Cell Biol ; 89(2): 167-72, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20585334

ABSTRACT

The macrophage is a versatile cell type that can sense and respond to a particular need based on the conditions of the microenvironment. Some studies have recently suggested that pathogens can directly influence the polarization of macrophages. As Francisella infections are characterized by intense necrotic infiltrates in the lung as well as in distal sites of infection, we sought to investigate whether pulmonary Francisella infections could cause the polarization of alternatively activated macrophages (M2/aaMs). Our results indicate that Francisella infections can cause the polarization of M2/aaM in vivo and that macrophages can be polarized toward an M2/aaM phenotype more potently if dead cell debris is used for stimulation in the presence and absence of Francisella infections. Finally, we also demonstrate that efferocytosis is inhibited in macrophages infected with Francisella, thus providing a potential explanation for the lack of clearance and eventual accumulation of dead cell debris associated with this disease.


Subject(s)
Francisella/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/microbiology , Phagocytosis/immunology , Animals , Arginase/biosynthesis , Biomarkers , Lung/immunology , Lung/microbiology , Lung/pathology , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Models, Immunological , Necrosis , Up-Regulation
5.
PLoS One ; 5(11): e14088, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21124895

ABSTRACT

BACKGROUND: Pneumonia and pulmonary infections are major causes of mortality among the growing elderly population. Age associated attenuations of various immune parameters, involved with both innate and adaptive responses are collectively known as immune senescence. These changes are likely to be involved with differences in host susceptibility to disease between young and aged individuals. METHODOLOGY/PRINCIPAL FINDINGS: The objective of this study was to assess potential age related differences in the pulmonary host response in mice to the Gram-negative respiratory pathogen, Francisella novicida. We intranasally infected mice with F. novicida and compared various immune and pathological parameters of the pulmonary host response in both young and aged mice. CONCLUSIONS/SIGNIFICANCE: We observed that 20% of aged mice were able to survive an intranasal challenge with F. novicida while all of their younger cohorts died consistently within 4 to 6 days post infection. Further experiments revealed that all of the aged mice tested were initially able to control bacterial replication in the lungs as well as at distal sites of replication compared with young mice. In addition, the small cohort of aged survivors did not progress to a severe sepsis syndrome with hypercytokinemia, as did all of the young adult mice. Finally, a lack of widespread cell death in potential aged survivors coupled with a difference in cell types recruited to sites of infection within the lung confirmed an altered host response to Francisella in aged mice.


Subject(s)
Aging/immunology , Francisella/immunology , Gram-Negative Bacterial Infections/immunology , Lung Diseases/immunology , Animals , Apoptosis/immunology , Cell Survival/immunology , Cytokines/blood , Cytokines/immunology , Female , Francisella/physiology , Gram-Negative Bacterial Infections/microbiology , Host-Pathogen Interactions/immunology , Inflammation Mediators/blood , Inflammation Mediators/immunology , Kaplan-Meier Estimate , Lung/immunology , Lung/microbiology , Lung/pathology , Lung Diseases/microbiology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence
6.
BMC Microbiol ; 8: 215, 2008 Dec 09.
Article in English | MEDLINE | ID: mdl-19068128

ABSTRACT

BACKGROUND: Francisella tularensis subsp. tularensis is classified as a Category A bioweapon that is capable of establishing a lethal infection in humans upon inhalation of very few organisms. However, the virulence mechanisms of this organism are not well characterized. Francisella tularensis subsp. novicida, which is an equally virulent subspecies in mice, was used in concert with a microPET scanner to better understand its temporal dissemination in vivo upon intranasal infection and how such dissemination compares with other routes of infection. Adult mice were inoculated intranasally with F. tularensis subsp. novicida radiolabeled with 64Cu and imaged by microPET at 0.25, 2 and 20 hours post-infection. RESULTS: 64Cu labeled F. tularensis subsp. novicida administered intranasally or intratracheally were visualized in the respiratory tract and stomach at 0.25 hours post infection. By 20 hours, there was significant tropism to the lung compared with other tissues. In contrast, the images of radiolabeled F. tularensis subsp. novicida when administered intragastrically, intradermally, intraperitoneally and intravenouslly were more generally limited to the gastrointestinal system, site of inoculation, liver and spleen respectively. MicroPET images correlated with the biodistribution of isotope and bacterial burdens in analyzed tissues. CONCLUSION: Our findings suggest that Francisella has a differential tissue tropism depending on the route of entry and that the virulence of Francisella by the pulmonary route is associated with a rapid bacteremia and an early preferential tropism to the lung. In addition, the use of the microPET device allowed us to identify the cecum as a novel site of colonization of Francisella tularensis subsp. novicida in mice.


Subject(s)
Francisella tularensis/pathogenicity , Tularemia/microbiology , Tularemia/pathology , Animals , Copper Radioisotopes/analysis , Francisella tularensis/isolation & purification , Gastrointestinal Tract/microbiology , Liver/microbiology , Lung/microbiology , Mice , Mice, Inbred C57BL , Positron-Emission Tomography/methods , Spleen/microbiology , Staining and Labeling , Whole Body Imaging/methods
7.
Infect Immun ; 76(7): 3001-10, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18411294

ABSTRACT

"Francisella tularensis subsp. novicida" intranasal infection causes a rapid pneumonia in mice with mortality at 4 to 6 days with a low dose of bacteria (10(2) bacteria). The short time to death suggests that there is a failure of the innate immune response. As the neutrophil is often the first cell type to infiltrate sites of infection, we focused on the emigration of neutrophils in this infection, as well as cytokines involved in their recruitment. The results indicated that there was a significant delay in the influx of neutrophils into the bronchoalveolar lavage fluid of F. tularensis subsp. novicida-infected mice. The delay in neutrophil recruitment in F. tularensis subsp. novicida-infected mice correlated with a delay in the upregulation of multiple proinflammatory cytokines and chemokines, as well as a delay in caspase-1 activation. Strikingly, the initial delay in the upregulation of cytokines through 1 day postinfection was followed by profound upregulation of multiple cytokines and chemokines to levels consistent with hypercytokinemia described for severe sepsis. This finding was further supported by a bacteremia and the cellular relocalization and release of high-mobility group box-1 and S100A9, both of which are damage-associated molecular pattern molecules and are known to be mediators of severe sepsis.


Subject(s)
Bacteremia , Calgranulin B/metabolism , Cytokines/metabolism , Francisella tularensis/pathogenicity , HMGB1 Protein/metabolism , Pneumonia, Bacterial , Up-Regulation , Animals , Bacteremia/immunology , Bacteremia/microbiology , Bacteremia/physiopathology , Blood/microbiology , Cell Line , Colony Count, Microbial , Female , Francisella tularensis/immunology , Francisella tularensis/isolation & purification , Humans , Lung/immunology , Lung/microbiology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/physiopathology , Time Factors , Tularemia/immunology , Tularemia/microbiology , Tularemia/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...