Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
BMC Microbiol ; 18(1): 45, 2018 05 30.
Article in English | MEDLINE | ID: mdl-29848308

ABSTRACT

BACKGROUND: Mice are a natural host for Rodentibacter (R.) pneumotropicus. Despite specific monitoring, it is still one of the most important infectious agents in laboratory animals. The objective of this study was to determine the virulence of a prevalent pathotype of R. pneumotropicus and characterize the host response in a new animal model. RESULTS: Intranasal infection of C57BL/6 and BALB/c mice with a R. pneumotropicus strain (JF4Ni) bearing the genes of the three known repeats in toxin (RTX) toxins resulted in an unprecedented high mortality and morbidity above 50 and 80%, respectively. Morbidity was associated with severe weight loss as well as conjunctivitis and dyspnea. A main pathology was a catarrhal purulent to necrotic bronchopneumonia. Specific immune globuline (Ig) A was detected in tracheonasal lavages of most surviving mice which were still colonized by R. pneumotropicus. Furthermore, all surviving animals showed a distinct production of IgG antibodies. To differentiate T-helper cell (Th) 1 and Th2 immune responses we used subclasses of IgGs as indicators. Mean ratios of IgG2b to IgG1 were below 0.8 in sera drawn from both mice strains prior infection and from BALB/c mice post infection. In contrast, C57BL/6 mice had a mean IgG2b/IgG1 ratio of 1.6 post infection indicating a Th1 immune response in C57BL/6 versus a Th2 response in BALB/c mice associated with a tenfold higher bacterial load in the lung. In accordance with a Th1 response high antigen-specific IgG2c titers were detected in the majority of surviving C57BL/6 mice. CONCLUSIONS: R. pneumotropicus JF4Ni is a highly virulent strain causing severe pneumonia and septicemia after intranasal infection of C57BL/6 and BALB/c mice. Persisting infections in the two mice strains are associated with Th1 and Th2 immune responses, respectively, and differences in the bacterial burden of the lung. The described model is ideally suited for future vaccination studies using the natural host.


Subject(s)
Immunity, Humoral , Immunoglobulin G/metabolism , Pasteurella Infections/immunology , Pasteurella pneumotropica/pathogenicity , Animals , Lung/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pasteurella Infections/mortality , Pasteurella pneumotropica/immunology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Sepsis/immunology , Sepsis/microbiology , Th1 Cells/immunology , Th2 Cells/immunology
2.
Comp Med ; 67(4): 315-329, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28830578

ABSTRACT

A colony of B6.Cg-Rag1tm1Mom Tyrp1B-w Tg(Tcra,Tcrb)9Rest (TRP1/TCR) mice presented with ocular lesions and ulcerative dermatitis. Histopathology, skin scrapes, and fur plucks confirmed the presence of Demodex spp. in all clinically affected and subclinical TRP1/TCR mice examined (n = 48). Pasteurella pneumotropica and Corynebacterium bovis, both opportunistic pathogens, were cultured from the ocular lesions and skin, respectively, and bacteria were observed microscopically in abscesses at various anatomic locations (including retroorbital sites, tympanic bullae, lymph nodes, and reproductive organs) as well as the affected epidermis. The mites were identified as Demodex musculi using the skin fragment digestion technique. Topographic analysis of the skin revealed mites in almost all areas of densely haired skin, indicating a generalized demodecosis. The percentage of infested follicles in 8- to 10-wk-old mice ranged from 0% to 21%, and the number of mites per millimeter of skin ranged from 0 to 3.7. The head, interscapular region, and middorsum had the highest proportions of infested follicles, ranging from 2.3% to 21.1% (median, 4.9%), 2.0% to 16.6% (8.1%), and 0% to 17% (7.6%), respectively. The pinnae and tail skin had few or no mites, with the proportion of follicles infested ranging from 0% to 3.3% (0%) and 0% to 1.4% (0%), respectively. The number of mites per millimeter was strongly correlated with the percentage of infested follicles. After administration of amoxicillin-impregnated feed (0.12%), suppurative infections were eliminated, and the incidence of ulcerative dermatitis was dramatically reduced. We hypothesize that the Rag1-null component of the genotype makes TRP1/TCR mice susceptible to various opportunistic infestations and infections, including Demodex mites, P. pneumotropica, and C. bovis. Therefore, Rag1-null mice may serve as a useful model to study human and canine demodecosis. D. musculi should be ruled out as a contributing factor in immunocompromised mouse strains with dermatologic manifestations.


Subject(s)
Adaptive Immunity , Corynebacterium Infections/veterinary , Corynebacterium/pathogenicity , Mite Infestations/veterinary , Opportunistic Infections/veterinary , Pasteurella Infections/veterinary , Pasteurella pneumotropica/pathogenicity , Skin , Adaptive Immunity/genetics , Animals , Corynebacterium/immunology , Corynebacterium Infections/genetics , Corynebacterium Infections/immunology , Corynebacterium Infections/microbiology , Female , Genetic Predisposition to Disease , Homeodomain Proteins/genetics , Host-Pathogen Interactions , Immunocompromised Host , Male , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Mite Infestations/genetics , Mite Infestations/immunology , Mite Infestations/parasitology , Opportunistic Infections/immunology , Opportunistic Infections/microbiology , Opportunistic Infections/parasitology , Oxidoreductases/genetics , Parasite Load , Pasteurella Infections/genetics , Pasteurella Infections/immunology , Pasteurella Infections/microbiology , Pasteurella pneumotropica/immunology , Phenotype , Receptors, Antigen, T-Cell/genetics , Risk Factors , Skin/immunology , Skin/microbiology , Skin/parasitology , Skin/pathology
3.
Am J Respir Cell Mol Biol ; 54(2): 210-21, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26121027

ABSTRACT

Resident immune cells (e.g., macrophages [MΦs]) and airway mucus clearance both contribute to a healthy lung environment. To investigate interactions between pulmonary MΦ function and defective mucus clearance, a genetic model of lysozyme M (LysM) promoter-mediated MΦ depletion was generated, characterized, and crossed with the sodium channel ß subunit transgenic (Scnn1b-Tg) mouse model of defective mucus clearance. Diphtheria toxin A-mediated depletion of LysM(+) pulmonary MΦs in wild-type mice with normal mucus clearance resulted in lethal pneumonia in 24% of neonates. The pneumonias were dominated by Pasteurella pneumotropica and accompanied by emaciation, neutrophilic inflammation, and elevated Th1 cytokines. The incidence of emaciation and pneumonia reached 51% when LysM(+) MΦ depletion was superimposed on the airway mucus clearance defect of Scnn1b-Tg mice. In LysM(+) MΦ-depleted Scnn1b-Tg mice, pneumonias were associated with a broader spectrum of bacterial species and a significant reduction in airway mucus plugging. Bacterial burden (CFUs) was comparable between Scnn1b-Tg and nonpneumonic LysM(+) MΦ-depleted Scnn1b-Tg mice. However, the nonpneumonic LysM(+) MΦ-depleted Scnn1b-Tg mice exhibited increased airway inflammation, the presence of neutrophilic infiltration, and increased levels of inflammatory cytokines in bronchoalveolar lavage fluid compared with Scnn1b-Tg mice. Collectively, these data identify key MΦ-mucus clearance interactions with respect to both infectious and inflammatory components of muco-obstructive lung disease.


Subject(s)
Lung/immunology , Macrophages/immunology , Mucociliary Clearance , Pasteurella Infections/immunology , Pasteurella pneumotropica/immunology , Pneumonia, Bacterial/immunology , Animals , Animals, Newborn , Cytokines/immunology , Cytokines/metabolism , Diphtheria Toxin/genetics , Diphtheria Toxin/metabolism , Disease Models, Animal , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Genetic Predisposition to Disease , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lung/metabolism , Lung/microbiology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Muramidase/genetics , Pasteurella Infections/genetics , Pasteurella Infections/metabolism , Pasteurella Infections/microbiology , Pasteurella pneumotropica/pathogenicity , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phenotype , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Promoter Regions, Genetic
4.
Immunobiology ; 220(10): 1177-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26074063

ABSTRACT

Ficolins recognize pathogen associated molecular patterns and activate the lectin pathway of complement system. However, our knowledge regarding pathogen recognition of human ficolins is still limited. We therefore set out to explore and investigate the possible interactions of the two main serum ficolins, ficolin-2 and ficolin-3 with different Gram-negative bacteria. We used recombinant ficolin molecules and normal human serum, which were detected with anti-ficolin monoclonal antibodies. In addition we investigated the capacity of these pathogens to activate the lectin pathway of complement system. We show for the first time that human ficolin-2 recognizes the nonpathogenic spirochete Leptospira biflexa serovar Patoc, but not the pathogenic Leptospira interrogans serovar Kennewicki strain Fromm. Additionally, human ficolin-2 and ficolin-3 recognize pathogenic Pasteurella pneumotropica, enteropathogenic Escherichia coli (EPEC) serotype O111ab:H2 and enteroaggregative E. coli (EAEC) serogroup O71 but not four enterohemorrhagic E. coli, three EPEC, three EAEC and two nonpathogenic E. coli strains (DH5α and HB101). The lectin pathway was activated by Pasteurella pneumotropica, EPEC O111ab:H2 and EAEC O71 after incubation with C1q depleted human serum. In conclusion, this study provide novel insight in the binding and complement activating capacity of the lectin pathway initiation molecules ficolin-2 and ficolin-3 towards relevant Gram-negative pathogens of pathophysiological relevance.


Subject(s)
Complement Pathway, Mannose-Binding Lectin/immunology , Escherichia coli/immunology , Glycoproteins/immunology , Lectins/immunology , Leptospira/immunology , Pasteurella pneumotropica/immunology , Humans , Recombinant Proteins/immunology , Ficolins
5.
PLoS One ; 9(10): e111194, 2014.
Article in English | MEDLINE | ID: mdl-25347183

ABSTRACT

Pasteurella pneumotropica is an opportunist Gram negative bacterium responsible for rodent pasteurellosis that affects upper respiratory, reproductive and digestive tracts of mammals. In animal care facilities the presence of P. pneumotropica causes severe to lethal infection in immunodeficient mice, being also a potential source for human contamination. Indeed, occupational exposure is one of the main causes of human infection by P. pneumotropica. The clinical presentation of the disease includes subcutaneous abscesses, respiratory tract colonization and systemic infections. Given the ability of P. pneumotropica to fully disseminate in the organism, it is quite relevant to study the role of the complement system to control the infection as well as the possible evasion mechanisms involved in bacterial survival. Here, we show for the first time that P. pneumotropica is able to survive the bactericidal activity of the human complement system. We observed that host regulatory complement C4BP and Factor H bind to the surface of P. pneumotropica, controlling the activation pathways regulating the formation and maintenance of C3-convertases. These results show that P. pneumotropica has evolved mechanisms to evade the human complement system that may increase the efficiency by which this pathogen is able to gain access to and colonize inner tissues where it may cause severe infections.


Subject(s)
Complement C4b-Binding Protein/immunology , Complement Factor H/immunology , Pasteurella pneumotropica/immunology , Complement C3-C5 Convertases/metabolism , Humans
6.
Vaccine ; 31(48): 5729-35, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24091313

ABSTRACT

Intranasal vaccination is one of the most effective means of protecting against invading and colonizing pathogens because the vaccine elicits a mucosal immune response. The exploitation of vaccine adjuvants and delivery systems for intranasal vaccines is an important way to evoke antigen immunogenicity and elicit a better immune response at the mucosal sites. In the present study, we assessed the potential of intranasal immunization using a non-adjuvanted bacterial adhesive protein toward the host organs. We evaluated intranasal immunization with modified recombinant PnxIIIA (MP3) from Pasteurella pneumotropica and its preventive efficacy against opportunistic infection caused by P. pneumotropica, without using any adjuvants or delivery systems. The 100-kDa MP3 was confirmed to retain its immunogenicity and binding activity to collagen type I similar to the parent PnxIIIA. When MP3 was fused to green-fluorescent protein and inoculated into C57BL/6J mice intranasally, fluorescence intensity in the intranasal airway could be observed until 3 h after inoculation. Mice were intranasally immunized with MP3 at a maximum of 4 doses, with 7-day intervals. The antibody titer of serum IgG and IgA specific for MP3, as well as that of bronchoalveolar lavage fluid IgA, showed more than 9 (log2) after 3 or 4 rounds of immunization. Experimentally infecting immunized mice with P. pneumotropica resulted in the inability to isolate the bacterium from the nasal cavity, trachea, conjunctiva, or cecum with more than 3 doses in the immunized mice. Although the detection in each organ seldom changed with less than 2 rounds of immunization, unlike that observed in the non-immunized mice, the detection remarkably decreased with 3 or more rounds of immunization. These results suggest that intranasal immunization with a non-adjuvanted adhesive protein could have preventive effects against opportunistic infection by P. pneumotropica.


Subject(s)
Adhesins, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Opportunistic Infections/prevention & control , Pasteurella Infections/prevention & control , Pasteurella pneumotropica/immunology , Vaccination/methods , Adhesins, Bacterial/genetics , Administration, Intranasal , Animals , Antibodies, Bacterial/analysis , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Bronchoalveolar Lavage Fluid/chemistry , Disease Models, Animal , Female , Immunity, Mucosal , Immunoglobulin A/analysis , Immunoglobulin G/blood , Mice , Mice, Inbred C57BL , Pasteurella pneumotropica/genetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
7.
Microbes Infect ; 15(6-7): 470-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23624107

ABSTRACT

The ability of recombinant outer membrane proteins of Pasteurella pneumotropica to vaccinate against the infections of mice was studied. The proteins examined were the homologues of the P4, P6, P26, and D15 proteins of Haemophilus influenzae. Intranasal vaccination with P4 and P6 produced protection against pneumonia. P6 vaccination, which was most studied, reduced the peak bacteria load in lungs by 50-fold and caused a rapid resolution of an infection that lasted for at least 5 days in unvaccinated animals. Protection could be partially transferred with CD4(+) T cells and pulmonary challenge with the P6 antigen induced interferon-γ and the Th17 cytokine IL-21. This is the first demonstration of the ability of a recombinant P6 to mediate protective immunity to a pathogen in its natural host and it is proposed that it would not only have utility for mouse breeding but also for investigating how to improve the efficacy of vaccination with homologous proteins for related species.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Pasteurella Infections/prevention & control , Pasteurella pneumotropica/immunology , Administration, Intranasal , Adoptive Transfer , Animals , Bacterial Load , Bacterial Outer Membrane Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Disease Models, Animal , Lung/microbiology , Lung/pathology , Mice , Pasteurella Infections/immunology , Pasteurella Infections/microbiology , Pasteurella Infections/pathology , Pasteurella pneumotropica/genetics , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/prevention & control , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
8.
J Endod ; 38(3): 346-50, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22341072

ABSTRACT

INTRODUCTION: The present study investigated whether bacteria infecting the root canal can activate any infiltrating T cells to produce receptor activator of nuclear factor kappa B (NF-κB) ligand (RANKL). METHODS: Using a mouse model of periapical lesion induced by artificial dental pulp exposure, the presence of RANKL-positive T cells and osteoclasts in the periapical lesion was examined by an immunohistochemical approach. The bacteria colonizing the exposed root canal were identified by 16S ribosomal RNA (rRNA) sequence analysis. The isolated endodontic bacteria were further immunized to normal mice, and soluble activator of NF-κB ligand (sRANKL) production by the T cells isolated from the immunized mice was evaluated by ex vivo culture system. RESULTS: RANKL-positive T cells along with TRAP+ osteoclasts were identified in periapical bone resorption lesions. The gram-negative bacterium Pasteurella pnumotropica, which was most frequently detected from the root canal of exposed pulp, showed remarkably elevated serum immunoglobulin G (IgG)-antibody response in pulp-exposed mice compared with control nontreated mice. Immunization of mice with P. pneumotropica induced not only serum IgG-antibody but also primed bacteria-reactive T cells that produced sRANKL in response to ex vivo exposure to P. pneumotropica. CONCLUSIONS: T cells infiltrating the periapical region express RANKL, and the endodontic bacteria colonizing the root canal appear to induce RANKL expression from bacteria-reactive T cells, suggesting the possible pathogenic engagement of the immune response to endodontic bacteria in the context of developing bone resorptive periapical lesions.


Subject(s)
Alveolar Bone Loss/immunology , Pasteurella Infections/immunology , Pasteurella pneumotropica/immunology , Periapical Diseases/immunology , RANK Ligand/immunology , T-Lymphocytes/immunology , Acid Phosphatase/analysis , Alveolar Bone Loss/microbiology , Animals , Antibodies, Bacterial/blood , Biomarkers/analysis , CD3 Complex/immunology , Dental Pulp Cavity/microbiology , Dental Pulp Exposure/microbiology , Disease Models, Animal , Enterococcus/immunology , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Immunization , Immunoglobulin G/blood , Immunologic Memory/immunology , Isoenzymes/analysis , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Microscopy, Confocal , Osteoclasts/pathology , Pasteurella pneumotropica/classification , Periapical Diseases/microbiology , RNA, Bacterial/analysis , RNA, Ribosomal, 16S/analysis , T-Lymphocytes/pathology , Tartrate-Resistant Acid Phosphatase
9.
Oral Microbiol Immunol ; 22(3): 208-15, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17488448

ABSTRACT

INTRODUCTION: The present study examined whether induction of an adaptive immune response to orally colonizing non-pathogenic Pasteurella pneumotropica by immunization with the phylogenetically closely related bacterium, Actinobacillus actinomycetemcomitans, can result in periodontal bone loss in mice. METHODS: BALB/c mice harboring P. pneumotropica (P. pneumotropica(+) mice) in the oral cavity or control P. pneumotropica-free mice were immunized with fixed A. actinomycetemcomitans. The animals were sacrificed on day 30, and the following measurements were carried out: (i) serum immunoglobulin G and gingival T-cell responses to A. actinomycetemcomitans and P. pneumotropica; (ii) periodontal bone loss; and (iii) identification of receptor activator of nuclear factor-kappaB ligand (RANKL) -positive T cells in gingival tissue. RESULTS: Immunization with A. actinomycetemcomitans induced a significantly elevated serum immunoglobulin G response to the 29-kDa A. actinomycetemcomitans outer membrane protein (Omp29), which showed strong cross-reactivity with P. pneumotropica OmpA compared to results in the control non-immunized mice. The A. actinomycetemcomitans-immunized P. pneumotropica(+) mice developed remarkable periodontal bone loss in a RANKL-dependent manner, as determined by the abrogation of bone loss by treatment with osteoprotegerin-Fc. The T cells isolated from the gingival tissue of A. actinomycetemcomitans-immunized P. pneumotropica(+) mice showed an in vitro proliferative response to both A. actinomycetemcomitans and P. pneumotropica antigen presentation, as well as production of soluble(s)RANKL in the culture supernatant. Double-color confocal microscopy demonstrated that the frequency of RANKL(+) T cells in the gingival tissue of A. actinomycetemcomitans-immunized P. pneumotropica(+) mice was remarkably elevated compared to control mice. CONCLUSION: The induction of an adaptive immune response to orally colonizing non-pathogenic P. pneumotropica results in RANKL-dependent periodontal bone loss in mice.


Subject(s)
Aggregatibacter actinomycetemcomitans/immunology , Alveolar Bone Loss/immunology , Alveolar Bone Loss/microbiology , Pasteurella pneumotropica/immunology , RANK Ligand/immunology , Animals , Antigens, Bacterial , Bacterial Outer Membrane Proteins/immunology , Cross Reactions , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Models, Animal , Osteoprotegerin/pharmacology , Pasteurella pneumotropica/pathogenicity , RANK Ligand/antagonists & inhibitors , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...