Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(6): e0234038, 2020.
Article in English | MEDLINE | ID: mdl-32492075

ABSTRACT

Extracellular adenosine triphosphate (eATP) released by damaged cells, and its purinergic receptors, comprise a crucial signaling network after injury. Purinergic receptor P2X7 (P2RX7), a major driver of NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation and IL-1ß processing, has been shown to play a role in liver injury in murine diet- and chemically-induced liver injury models. It is unclear, however, whether P2RX7 plays a role in non-alcoholic steatohepatitis (NASH) and which cell type is the main target of P2RX7 pharmacological inhibition. Here, we report that P2RX7 is expressed by infiltrating monocytes and resident Kupffer cells in livers from NASH-affected individuals. Using primary isolated human cells, we demonstrate that P2RX7 expression in CD14+ monocytes and Kupffer cells primarily mediates IL-1ß release. In addition, we show that pharmacological inhibition of P2RX7 in monocytes and Kupffer cells, blocks IL-1ß release, reducing hepatocyte caspase 3/7 activity, IL-1ß-mediated CCL2 and CCL5 chemokine gene expression and secretion, and hepatic stellate cell (HSC) procollagen secretion. Consequently, in a chemically-induced nonhuman primate model of liver fibrosis, treatment with a P2RX7 inhibitor improved histological characteristics of NASH, protecting from liver inflammation and fibrosis. Taken together, these findings underscore the critical role of P2RX7 in the pathogenesis of NASH and implicate P2RX7 as a promising therapeutic target for the management of this disease.


Subject(s)
Inflammation/prevention & control , Liver Cirrhosis/drug therapy , Purinergic P2X Receptor Antagonists/therapeutic use , Receptors, Purinergic P2X7/metabolism , Animals , Caspase 3/metabolism , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Inflammation/pathology , Interleukin-1beta/metabolism , Kupffer Cells/cytology , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Lipopolysaccharides/pharmacology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Macaca fascicularis , Male , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Procollagen/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/chemistry , Receptors, Purinergic P2X7/genetics
2.
J Immunol Methods ; 405: 97-108, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24508527

ABSTRACT

New efforts to understand complex interactions between diet, gut microbiota, and intestinal immunity emphasize the need for a standardized murine protocol that has been optimized for the isolation of lamina propria immune cells. In this study multiple mouse strains including BALB/c, 129S6/Sv/EvTac and ICR mice were utilized to develop an optimal protocol for global analysis of lamina propria leukocytes. Incubation temperature was found to significantly improve epithelial cell removal, while changes in media formulation had minor effects. Tissue weight was an effective method for normalization of solution volumes and incubation times. Collagenase digestion in combination with thermolysin was identified as the optimal method for release of leukocytes from tissues and global immunophenotyping, based on the criteria of minimizing marker cleavage, improving cell viability, and reagent cost. The effects of collagenase in combination with dispase or thermolysin on individual cell surface markers revealed diverse marker specific effects. Aggressive formulations cleaved CD8α, CD138, and B220 from the cell surface, and resulted in relatively higher expression levels of CD3, γδ TCR, CD5, DX5, Ly6C, CD11b, CD11c, MHC-II and CD45. Improved collagenase digestion significantly improved viability and reduced debris formation, eliminating the need for density gradient purification. Finally, we demonstrate that two different digestion protocols yield significant differences in detection of CD4(+) and CD8(+) T cells, NK cells, monocytes and interdigitating DC (iDC) populations, highlighting the importance and impact of cell collection protocols on assay outputs. The optimized protocol described herein will help assure the reproducibility and robustness of global assessment of lamina propria immune responses. Moreover, this technique may be applied to isolation of leukocytes from the entire gastrointestinal tract.


Subject(s)
Intestinal Mucosa/immunology , Intestine, Small/immunology , Leukocytes/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Separation/methods , Cell Survival/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Immunophenotyping/methods , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestine, Small/cytology , Intestine, Small/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocytes/cytology , Leukocytes/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred ICR , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Reproducibility of Results
3.
Infect Immun ; 81(12): 4626-34, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24101688

ABSTRACT

Infections with the Gram-negative bacterium Burkholderia pseudomallei (melioidosis) are associated with high mortality, and there is currently no approved vaccine to prevent the development of melioidosis in humans. Infected patients also do not develop protective immunity to reinfection, and some individuals will develop chronic, subclinical infections with B. pseudomallei. At present, our understanding of what constitutes effective protective immunity against B. pseudomallei infection remains incomplete. Therefore, we conducted a study to elucidate immune correlates of vaccine-induced protective immunity against acute B. pseudomallei infection. BALB/c and C57BL/6 mice were immunized subcutaneously with a highly attenuated, Select Agent-excluded purM deletion mutant of B. pseudomallei (strain Bp82) and then subjected to intranasal challenge with virulent B. pseudomallei strain 1026b. Immunization with Bp82 generated significant protection from challenge with B. pseudomallei, and protection was associated with a significant reduction in bacterial burden in lungs, liver, and spleen of immunized mice. Humoral immunity was critically important for vaccine-induced protection, as mice lacking B cells were not protected by immunization and serum from Bp82-vaccinated mice could transfer partial protection to nonvaccinated animals. In contrast, vaccine-induced protective immunity was found to be independent of both CD4 and CD8 T cells. Tracking studies demonstrated uptake of the Bp82 vaccine strain predominately by neutrophils in vaccine-draining lymph nodes and by smaller numbers of dendritic cells (DC) and monocytes. We concluded that protection following cutaneous immunization with a live attenuated Burkholderia vaccine strain was dependent primarily on generation of effective humoral immune responses.


Subject(s)
Antibodies, Bacterial/immunology , Bacterial Vaccines/immunology , Burkholderia pseudomallei/immunology , Melioidosis/immunology , Vaccines, Attenuated/immunology , Animals , Bacterial Vaccines/administration & dosage , Burkholderia pseudomallei/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Female , Immunity, Humoral , Immunization , Melioidosis/microbiology , Melioidosis/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Vaccination
4.
Am J Trop Med Hyg ; 89(5): 973-982, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24062483

ABSTRACT

Burkholderia pseudomallei is a Gram-negative environmental bacterium found in tropical climates that causes melioidosis. Culture remains the diagnostic gold standard, but isolation of B. pseudomallei from heavily contaminated sites, such as fecal specimens, can be difficult. We recently reported that B. pseudomallei is capable of infecting the gastrointestinal tract of mice and suggested that the same may be true in humans. Thus, there is a strong need for new culture techniques to allow for efficient detection of B. pseudomallei in fecal and other specimens. We found that the addition of norfloxacin, ampicillin, and polymyxin B to Ashdown's medium (NAP-A) resulted in increased specificity without affecting the growth of 25 B. pseudomallei strains. Furthermore, recovery of B. pseudomallei from human clinical specimens was not affected by the three additional antibiotics. Therefore, we conclude that NAP-A medium provides a new tool for more sensitive isolation of B. pseudomallei from heavily contaminated sites.


Subject(s)
Burkholderia pseudomallei/isolation & purification , Culture Media/chemistry , Melioidosis/microbiology , Ampicillin/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bacteriological Techniques , Burkholderia pseudomallei/drug effects , Burkholderia pseudomallei/growth & development , Feces/microbiology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Norfloxacin/pharmacology , Polymyxin B/pharmacology , Sensitivity and Specificity
5.
PLoS Negl Trop Dis ; 6(6): e1715, 2012.
Article in English | MEDLINE | ID: mdl-22745846

ABSTRACT

Burkholderia pseudomallei is a mostly saprophytic bacterium, but can infect humans where it causes the difficult-to-manage disease melioidosis. Even with proper diagnosis and prompt therapeutic interventions mortality rates still range from >20% in Northern Australia to over 40% in Thailand. Surprisingly little is yet known about how B. pseudomallei infects, invades and survives within its hosts, and virtually nothing is known about the contribution of critical nutrients such as iron to the bacterium's pathogenesis. It was previously assumed that B. pseudomallei used iron-acquisition systems commonly found in other bacteria, for example siderophores. However, our previous discovery of a clinical isolate carrying a large chromosomal deletion missing the entire malleobactin gene cluster encoding the bacterium's major high-affinity siderophore while still being fully virulent in a murine melioidosis model suggested that other iron-acquisition systems might make contributions to virulence. Here, we deleted the major siderophore malleobactin (mba) and pyochelin (pch) gene clusters in strain 1710b and revealed a residual siderophore activity which was unrelated to other known Burkholderia siderophores such as cepabactin and cepaciachelin, and not due to increased secretion of chelators such as citrate. Deletion of the two hemin uptake loci, hmu and hem, showed that Hmu is required for utilization of hemin and hemoglobin and that Hem cannot complement a Hmu deficiency. Prolonged incubation of a hmu hem mutant in hemoglobin-containing minimal medium yielded variants able to utilize hemoglobin and hemin suggesting alternate pathways for utilization of these two host iron sources. Lactoferrin utilization was dependent on malleobactin, but not pyochelin synthesis and/or uptake. A mba pch hmu hem quadruple mutant could use ferritin as an iron source and upon intranasal infection was lethal in an acute murine melioidosis model. These data suggest that B. pseudomallei may employ a novel ferritin-iron acquisition pathway as a means to sustain in vivo growth.


Subject(s)
Burkholderia pseudomallei/pathogenicity , Hemin/metabolism , Melioidosis/microbiology , Melioidosis/pathology , Siderophores/metabolism , Virulence Factors/metabolism , Animals , Burkholderia pseudomallei/metabolism , Disease Models, Animal , Ferritins/metabolism , Gene Deletion , Melioidosis/mortality , Mice , Mice, Inbred BALB C , Survival Analysis , Virulence , Virulence Factors/genetics
6.
Vaccine ; 30(33): 4977-82, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22652404

ABSTRACT

Successful treatment of pneumonic infection with Francisella tularensis, the causative agent of tularemia, requires rapid initiation of antibiotic therapy, yet even then treatment failures may occur. Consequently, new treatments are needed to enhance the effectiveness of antimicrobial therapy for acute pneumonic tularemia. In a prior study, immunization with F. tularensis membrane protein fraction (MPF) antigens 3 days prior to challenge was reported to induce significant protection from inhalational challenge. We therefore hypothesized that MPF immunization might also be effective in enhancing infection control if combined with antibiotic therapy and administered after infection as post-exposure immunotherapy. To address this question, a 24h post-exposure treatment model of acute pulmonary Schu S4 strain of F. tularensis infection in BALB/c mice was used. Following exposure, mice were immunized with MPF and treated with low-dose gentamicin, alone or in combination and the effects on survival, bacterial burden and dissemination were assessed. We found that immunization with MPF significantly increased the effectiveness of subtherapeutic gentamicin for post-exposure treatment of pneumonic tularemia, with 100% of combination-treated mice surviving long-term. Bacterial burdens in the liver and spleen were significantly reduced in combination MPF-gentamicin treated mice at 7 days after challenge. Passively transferred antibodies against MPF antigens also increased the effectiveness of gentamicin therapy. Thus, we concluded that post-exposure immunization with MPF antigens was an effective means of enhancing conventional antimicrobial therapy for pneumonic tularemia.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Antibiotic Prophylaxis/methods , Bacterial Vaccines/administration & dosage , Gentamicins/administration & dosage , Post-Exposure Prophylaxis/methods , Tularemia/prevention & control , Vaccination/methods , Animals , Bacterial Load , Bacterial Vaccines/immunology , Disease Models, Animal , Female , Francisella tularensis/immunology , Liver/microbiology , Membrane Proteins/administration & dosage , Membrane Proteins/immunology , Mesothelin , Mice , Mice, Inbred BALB C , Spleen/microbiology , Survival Analysis , Treatment Outcome
7.
PLoS One ; 7(5): e37324, 2012.
Article in English | MEDLINE | ID: mdl-22624016

ABSTRACT

Melioidosis is a disease of humans caused by opportunistic infection with the soil and water bacterium Burkholderia pseudomallei. Melioidosis can manifest as an acute, overwhelming infection or as a chronic, recurrent infection. At present, it is not clear where B. pseudomallei resides in the mammalian host during the chronic, recurrent phase of infection. To address this question, we developed a mouse low-dose mucosal challenge model of chronic B. pseudomallei infection and investigated sites of bacterial persistence over 60 days. Sensitive culture techniques and selective media were used to quantitate bacterial burden in major organs, including the gastrointestinal (GI) tract. We found that the GI tract was the primary site of bacterial persistence during the chronic infection phase, and was the only site from which the organism could be consistently cultured during a 60-day infection period. The organism could be repeatedly recovered from all levels of the GI tract, and chronic infection was accompanied by sustained low-level fecal shedding. The stomach was identified as the primary site of GI colonization as determined by fluorescent in situ hybridization. Organisms in the stomach were associated with the gastric mucosal surface, and the propensity to colonize the gastric mucosa was observed with 4 different B. pseudomallei isolates. In contrast, B. pseudomallei organisms were present at low numbers within luminal contents in the small and large intestine and cecum relative to the stomach. Notably, inflammatory lesions were not detected in any GI tissue examined in chronically-infected mice. Only low-dose oral or intranasal inoculation led to GI colonization and development of chronic infection of the spleen and liver. Thus, we concluded that in a mouse model of melioidosis B. pseudomallei preferentially colonizes the stomach following oral inoculation, and that the chronically colonized GI tract likely serves as a reservoir for dissemination of infection to extra-intestinal sites.


Subject(s)
Burkholderia pseudomallei/physiology , Gastric Mucosa/microbiology , Gastrointestinal Tract/microbiology , Melioidosis/microbiology , Viscera/microbiology , Animals , Cell Culture Techniques , Feces/microbiology , Histological Techniques , In Situ Hybridization, Fluorescence , Mice
8.
BMC Microbiol ; 12: 71, 2012 Jul 04.
Article in English | MEDLINE | ID: mdl-22583915

ABSTRACT

BACKGROUND: Dietary rice bran consists of many bioactive components with disease fighting properties; including the capacity to modulate the gut microbiota. Studies point to the important roles of the gut microbiota and the mucosal epithelium in the establishment of protection against enteric pathogens, such as Salmonella. The ability of rice bran to reduce the susceptibility of mice to a Salmonella infection has not been previously investigated. Therefore, we hypothesized that the incorporation of rice bran into the diet would inhibit the colonization of Salmonella in mice through the induction of protective mucosal responses. RESULTS: Mice were fed diets containing 0%, 10% and 20% rice bran for one week prior to being orally infected with Salmonella enterica serovar Typhimurium. We found that mice consuming the 10 and 20% rice bran diets exhibited a reduction in Salmonella fecal shedding for up to nine days post-infection as compared to control diet fed animals (p < 0.05). In addition, we observed decreased concentrations of the pro-inflammatory cytokines, TNF-alpha, IFN-gamma, and IL-12 (p < 0.05) as well as increased colonization of native Lactobacillus spp. in rice bran fed mice (p < 0.05). Furthermore, in vitro experiments revealed the ability of rice bran extracts to reduce Salmonella entry into mouse small intestinal epithelial cells. CONCLUSIONS: Increasing rice bran consumption represents a novel dietary means for reducing susceptibility to enteric infection with Salmonella and potentially via induction of native Lactobacillus spp.


Subject(s)
Diet/methods , Dietary Fiber , Oryza , Salmonella Infections, Animal/prevention & control , Salmonella typhimurium/growth & development , Salmonella typhimurium/immunology , Animals , Bacterial Load , Bacterial Shedding , Cytokines/metabolism , Feces/microbiology , Female , Lactobacillus/growth & development , Lactobacillus/isolation & purification , Mice , Salmonella Infections, Animal/immunology , Salmonella typhimurium/isolation & purification , Salmonella typhimurium/pathogenicity
9.
PLoS Pathog ; 8(3): e1002587, 2012.
Article in English | MEDLINE | ID: mdl-22438809

ABSTRACT

Pulmonary Francisella tularensis and Burkholderia pseudomallei infections are highly lethal in untreated patients, and current antibiotic regimens are not always effective. Activating the innate immune system provides an alternative means of treating infection and can also complement antibiotic therapies. Several natural agonists were screened for their ability to enhance host resistance to infection, and polysaccharides derived from the Acai berry (Acai PS) were found to have potent abilities as an immunotherapeutic to treat F. tularensis and B. pseudomallei infections. In vitro, Acai PS impaired replication of Francisella in primary human macrophages co-cultured with autologous NK cells via augmentation of NK cell IFN-γ. Furthermore, Acai PS administered nasally before or after infection protected mice against type A F. tularensis aerosol challenge with survival rates up to 80%, and protection was still observed, albeit reduced, when mice were treated two days post-infection. Nasal Acai PS administration augmented intracellular expression of IFN-γ by NK cells in the lungs of F. tularensis-infected mice, and neutralization of IFN-γ ablated the protective effect of Acai PS. Likewise, nasal Acai PS treatment conferred protection against pulmonary infection with B. pseudomallei strain 1026b. Acai PS dramatically reduced the replication of B. pseudomallei in the lung and blocked bacterial dissemination to the spleen and liver. Nasal administration of Acai PS enhanced IFN-γ responses by NK and γδ T cells in the lungs, while neutralization of IFN-γ totally abrogated the protective effect of Acai PS against pulmonary B. pseudomallei infection. Collectively, these results demonstrate Acai PS is a potent innate immune agonist that can resolve F. tularensis and B. pseudomallei infections, suggesting this innate immune agonist has broad-spectrum activity against virulent intracellular pathogens.


Subject(s)
Arecaceae/chemistry , Immunity, Innate/drug effects , Melioidosis/prevention & control , Pneumonia/drug therapy , Polysaccharides/pharmacology , Tularemia/prevention & control , Administration, Intranasal , Animals , Burkholderia pseudomallei/drug effects , Burkholderia pseudomallei/immunology , Disease Models, Animal , Female , Francisella tularensis/drug effects , Francisella tularensis/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Longevity/drug effects , Lung/drug effects , Lung/immunology , Lung/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Melioidosis/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Pneumonia/immunology , Pneumonia/microbiology , Polysaccharides/administration & dosage , Polysaccharides/isolation & purification , Tularemia/immunology
10.
Infect Immun ; 80(1): 110-20, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22025508

ABSTRACT

The Gram-negative bacterium Burkholderia mallei causes rapidly fatal illness in equines and humans when contracted by inhalation and also has the potential to be used as a bioweapon. However, little is known regarding the early innate immune responses and signaling mechanisms required to generate protection from pneumonic B. mallei infection. We showed previously that monocyte chemoattractant protein 1 (MCP-1) was a critical chemokine required for protection from pneumonic B. mallei infection. We have now extended those studies to identify key Toll-like receptor (TLR) signaling pathways, effector cells, and cytokines required for protection from respiratory B. mallei infection. We found that MyD88-/- mice were highly susceptible to pulmonary challenge with B. mallei and had significantly short survival times, increased bacterial burdens, and severe organ pathology compared to wild-type mice. Notably, MyD88-/- mice had significantly fewer monocytes and dendritic cells (DCs) in lung tissues and airways than infected wild-type mice despite markedly higher bacterial burdens. The MyD88-/- mice were also completely unable to produce gamma interferon (IFN-γ) at any time points following infection. In wild-type mice, NK cells were the primary cells producing IFN-γ in the lungs following B. mallei infection, while DCs and monocytes were the primary cellular sources of interleukin-12 (IL-12) production. Treatment with recombinant IFN-γ (rIFN-γ) was able to significantly restore protective immunity in MyD88-/- mice. Thus, we conclude that the MyD88-dependent recruitment of inflammatory monocytes and DCs to the lungs and the local production of IL-12, followed by NK cell production of IFN-γ, are the key initial cellular responses required for early protection from B. mallei infection.


Subject(s)
Burkholderia mallei/immunology , Dendritic Cells/immunology , Glanders/immunology , Monocytes/immunology , Myeloid Differentiation Factor 88/metabolism , Pneumonia, Bacterial/immunology , Animals , Bacterial Load , Disease Models, Animal , Female , Interferon-gamma/metabolism , Interleukin-12/metabolism , Lung/microbiology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Pneumonia, Bacterial/microbiology , Survival Analysis
11.
Vaccine ; 28(36): 5924-9, 2010 Aug 16.
Article in English | MEDLINE | ID: mdl-20600517

ABSTRACT

Yersinia pestis is a dangerous bacterial pathogen that when inhaled can rapidly induce fatal pneumonic plague. Thus, there is a need for stable, safe, and easily administered mucosal vaccines capable of eliciting effective protection against pulmonary Y. pestis infections. Cationic liposome-nucleic acid complexes (CLDC) have been shown previously to be effective vaccine adjuvants for parenteral immunization, but have not been previously evaluated for use in oral immunization. Therefore, we investigated the ability of an orally administered CLDC adjuvanted vaccine to elicit protective immunity against lethal pneumonic plague. C57Bl/6 mice were vaccinated orally or subcutaneously using 10mug Y. pestis F1 antigen combined with CLDC and immune responses and protection from challenge was assessed. We found that oral immunization elicited high titers of anti-F1 antibodies, equivalent to those generated by parenteral immunization. Importantly, orally immunized mice were protected from lethal pulmonary challenge with virulent Y. pestis for up to 18 weeks following vaccination. Vaccine-induced protection following oral immunization was found to be dependent primarily on CD4+ T cells, with a partial contribution from CD8+ T cells. Thus, CLDC adjuvanted vaccines represent a new type of orally administered, non-replicating vaccine capable of generating effective protection against pulmonary infection with virulent Y. pestis.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Proteins/immunology , Plague Vaccine/immunology , Plague/prevention & control , Administration, Oral , Animals , Antibodies, Bacterial/blood , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Humoral , Liposomes/immunology , Mice , Mice, Inbred C57BL , Plague/immunology , Plague Vaccine/administration & dosage , Specific Pathogen-Free Organisms , Yersinia pestis/immunology , Yersinia pestis/pathogenicity
12.
J Immunol ; 184(3): 1445-54, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20042590

ABSTRACT

Burkholderia mallei is a gram-negative bacterial pathogen of domestic equidae and humans that can cause severe, rapidly life-threatening pneumonic infections. Little is known regarding the role of chemokines and early cellular immune responses in protective immunity to pulmonary infection with B. mallei. Although the role of MCP-1 in gram-positive bacterial infections has been previously investigated, the role of MCP-1 in immunity to acute pneumonia caused by gram-negative bacteria, such as B. mallei, has not been assessed. In a mouse model of pneumonic B. mallei infection, we found that both MCP-1(-/-) mice and CCR2(-/-) mice were extremely susceptible to pulmonary infection with B. mallei, compared with wild-type (WT) C57Bl/6 mice. Bacterial burden and organ lesions were significantly increased in CCR2(-/-) mice, compared with WT animals, following B. mallei challenge. Monocyte and dendritic cell recruitment into the lungs of CCR2(-/-) mice was significantly reduced in comparison with that in WT mice following B. mallei infection, whereas neutrophil recruitment was actually increased. Depletion of monocytes and macrophages prior to infection also greatly raised the susceptibility of WT mice to infection. Production of IL-12 and IFN-gamma in the lungs after B. mallei infection was significantly impaired in both MCP-1(-/-) and CCR2(-/-) mice, whereas treatment of CCR2(-/-) mice with rIFN-gamma restored protection against lethal challenge with B. mallei. Thus, we conclude that MCP-1 plays a key role in regulating cellular immunity and IFN-gamma production following pneumonic infection with B. mallei and therefore may also figure importantly in other gram-negative pneumonias.


Subject(s)
Burkholderia Infections/immunology , Burkholderia mallei/immunology , Chemokine CCL2/physiology , Pneumonia, Bacterial/immunology , Animals , Burkholderia Infections/genetics , Burkholderia Infections/prevention & control , Cell Movement/immunology , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Genetic Predisposition to Disease , Immunity, Cellular , Interferon-gamma/biosynthesis , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Neutrophil Infiltration/immunology , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/prevention & control , Receptors, CCR2/deficiency , Receptors, CCR2/genetics
13.
Infect Immun ; 77(4): 1579-88, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19179415

ABSTRACT

Burkholderia mallei and B. pseudomallei are important human pathogens and cause the diseases glanders and melioidosis, respectively. Both organisms are highly infectious when inhaled and are inherently resistant to many antimicrobials, thus making it difficult to treat pneumonic Burkholderia infections. We investigated whether it was possible to achieve rapid protection against inhaled Burkholderia infection by using inhaled immunotherapy. For this purpose, cationic liposome DNA complexes (CLDC), which are potent activators of innate immunity, were used to elicit the activation of pulmonary innate immune responses. We found that mucosal CLDC administration before or shortly after bacterial challenge could generate complete or nearly complete protection from inhalational challenge with 100% lethal doses of B. mallei and B. pseudomallei. Protection was found to be dependent on the CLDC-mediated induction of gamma interferon responses in lung tissues and was partially dependent on the activation of NK cells. However, CLDC-mediated protection was not dependent on the induction of inducible nitric oxide synthase, as assessed by depletion studies. We concluded that the potent local activation of innate immune responses in the lung could be used to elicit rapid and nonspecific protection from aerosol exposure to both B. mallei and B. pseudomallei.


Subject(s)
Burkholderia Infections , Burkholderia mallei/pathogenicity , Burkholderia pseudomallei/pathogenicity , Immunotherapy/methods , Lung Diseases , Administration, Inhalation , Animals , Burkholderia Infections/immunology , Burkholderia Infections/microbiology , Burkholderia Infections/prevention & control , Burkholderia Infections/therapy , Cations , Cell Line , DNA, Bacterial/administration & dosage , DNA, Bacterial/genetics , DNA, Bacterial/immunology , Escherichia coli/genetics , Glanders/immunology , Glanders/microbiology , Glanders/prevention & control , Glanders/therapy , Humans , Interferon-gamma/biosynthesis , Liposomes/administration & dosage , Liposomes/immunology , Lung Diseases/immunology , Lung Diseases/microbiology , Lung Diseases/prevention & control , Lung Diseases/therapy , Macrophages, Alveolar/microbiology , Melioidosis/immunology , Melioidosis/microbiology , Melioidosis/prevention & control , Melioidosis/therapy , Mice , Mice, Inbred BALB C , Plasmids/administration & dosage , Plasmids/genetics , Plasmids/immunology
14.
Colloids Surf B Biointerfaces ; 57(2): 161-73, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17324560

ABSTRACT

We explored the bionanofabrication of silicon nanopillar structures using ordered gold nanoparticle arrays generated from microbial surface layer (S-layer) protein templates. The S-layer template used for these thin film processing experiments was isolated from the Gram-positive bacterium Deinococcus radiodurans. In this preliminary work, S-layers preimmobilized onto chemically modified silicon substrates were initially used to template the fabrication of a nanolithographic hard mask pattern comprised of a hexagonally ordered array of 5-nm gold nanoparticles (lattice constant=18 nm). Significantly, the use of the biotemplated gold nanoparticle mask patterns in an inductively coupled plasma (ICP) etching process successfully yielded silicon nanopillar structures. However, it was found that the resultant nanopillars (8-13 nm wide at the tip, 15-20 nm wide at half-height, 20-30 nm wide at the base, and 60-90 nm tall) appeared to lack any significant degree of translational ordering. The results suggest that further studies are needed in order to elucidate the optimal plasma processing parameters that will lead to the generation of long-range ordered arrays of silicon-based nanostructures using S-layer protein templates.


Subject(s)
Colloids/chemistry , Gold/chemistry , Nanostructures , Proteins/chemistry , Silicon/chemistry , Microscopy, Electron, Scanning
15.
J Immunol ; 175(10): 6750-6, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16272331

ABSTRACT

Despite the importance of pneumonic plague, little is known of the early pulmonary immune responses that occur following inhalation of Yersinia pestis. Therefore, we conducted studies to identify the early target cells for uptake of Y. pestis in the lungs following intratracheal or i.v. inoculation. Following intratracheal inoculation, Y. pestis was rapidly internalized primarily by a distinctive population of CD11c+DEC-205+CD11b- cells in the airways, whereas i.v. inoculation resulted in uptake primarily by CD11b+CD11c- macrophages and granulocytes in lung tissues. The airway cells internalized and were infected by Y. pestis, but did not support active replication of the organism. Intratracheal inoculation of Y. pestis resulted in rapid activation of airway CD11c+ cells, followed within 24 h by the selective disappearance of these cells from the airways and lungs and the accumulation of apoptotic CD11c+ cells in draining lymph nodes. When CD11c+ cells in the airways were depleted using liposomal clodronate before infection, this resulted in a significantly increased replication of Y. pestis in the lungs and dissemination to the spleen and draining lymph nodes. These findings suggest that CD11c+ cells in the airways play an important role in suppressing the initial replication and dissemination of inhaled Y. pestis, although these results will also require confirmation using fully virulent strains of Y. pestis. Depletion of these airway cells by Y. pestis may therefore be one strategy the organism uses to overcome pulmonary defenses following inhalation of the organism.


Subject(s)
Antigen-Presenting Cells/immunology , Lung/immunology , Plague/immunology , Yersinia pestis/immunology , Administration, Inhalation , Animals , Antigen-Presenting Cells/microbiology , Antigen-Presenting Cells/pathology , Apoptosis , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , CD11c Antigen/metabolism , Cell Movement , Female , Injections, Intravenous , Lung/microbiology , Lung/pathology , Lymph Nodes/immunology , Lymph Nodes/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plague/microbiology , Plague/pathology , Receptors, CCR7 , Receptors, Chemokine/metabolism , Spleen/immunology , Spleen/microbiology , Yersinia pestis/pathogenicity
16.
Genome Res ; 13(5): 753-63, 2003 May.
Article in English | MEDLINE | ID: mdl-12695326

ABSTRACT

Genes detected by wheat expressed sequence tags (ESTs) were mapped into chromosome bins delineated by breakpoints of 159 overlapping deletions. These data were used to assess the organizational and evolutionary aspects of wheat genomes. Relative gene density and recombination rate increased with the relative distance of a bin from the centromere. Single-gene loci present once in the wheat genomes were found predominantly in the proximal, low-recombination regions, while multigene loci tended to be more frequent in distal, high-recombination regions. One-quarter of all gene motifs within wheat genomes were represented by two or more duplicated loci (paralogous sets). For 40 such sets, ancestral loci and loci derived from them by duplication were identified. Loci derived by duplication were most frequently located in distal, high-recombination chromosome regions whereas ancestral loci were most frequently located proximal to them. It is suggested that recombination has played a central role in the evolution of wheat genome structure and that gradients of recombination rates along chromosome arms promote more rapid rates of genome evolution in distal, high-recombination regions than in proximal, low-recombination regions.


Subject(s)
Chromosomes, Plant/genetics , Evolution, Molecular , Genome, Plant , Recombination, Genetic/genetics , Triticum/genetics , Chromosome Mapping/methods , Chromosome Mapping/statistics & numerical data , Genes, Duplicate/genetics , Genes, Plant/genetics , Genetic Markers/genetics , Multigene Family/genetics , Oryza/genetics , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...