Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
PLoS Pathog ; 20(6): e1011777, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38913740

RESUMO

COVID-associated coagulopathy seemly plays a key role in post-acute sequelae of SARS- CoV-2 infection. However, the underlying pathophysiological mechanisms are poorly understood, largely due to the lack of suitable animal models that recapitulate key clinical and pathological symptoms. Here, we fully characterized AC70 line of human ACE2 transgenic (AC70 hACE2 Tg) mice for SARS-CoV-2 infection. We noted that this model is highly permissive to SARS-CoV-2 with values of 50% lethal dose and infectious dose as ~ 3 and ~ 0.5 TCID50 of SARS-CoV-2, respectively. Mice infected with 105 TCID50 of SARS-CoV-2 rapidly succumbed to infection with 100% mortality within 5 days. Lung and brain were the prime tissues harboring high viral titers, accompanied by histopathology. However, viral RNA and inflammatory mediators could be detectable in other organs, suggesting the nature of a systemic infection. Lethal challenge of AC70 hACE2 Tg mice caused acute onset of leukopenia, lymphopenia, along with an increased neutrophil-to-lymphocyte ratio (NLR). Importantly, infected animals recapitulated key features of COVID-19-associated coagulopathy. SARS-CoV-2 could induce the release of circulating neutrophil extracellular traps (NETs), along with activated platelet/endothelium marker. Immunohistochemical staining with anti-platelet factor-4 (PF4) antibody revealed profound platelet aggregates especially within blocked veins of the lungs. We showed that acute SARS-CoV-2 infection triggered a hypercoagulable state coexisting with ill-regulated fibrinolysis. Finally, we highlighted the potential role of Annexin A2 (ANXA2) in fibrinolytic failure. ANXA2 is a calcium-dependent phospholipid-binding protein that forms a heterotertrameric complexes localized at the extracellular membranes with two S100A10 small molecules acting as a co-receptor for tissue-plasminogen activator (t-PA), tightly involved in cell surface fibrinolysis. Thus, our results revealing elevated IgG type anti-ANXA2 antibody production, downregulated de novo ANXA2/S100A10 synthesis, and reduced ANXA2/S100A10 association in infected mice, this protein might serve as druggable targets for development of antithrombotic and/or anti-fibrinolytic agents to attenuate pathogenesis of COVID-19.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Modelos Animais de Doenças , Camundongos Transgênicos , SARS-CoV-2 , Animais , COVID-19/patologia , COVID-19/complicações , COVID-19/virologia , COVID-19/metabolismo , Camundongos , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Humanos , Transtornos da Coagulação Sanguínea/virologia , Transtornos da Coagulação Sanguínea/patologia , Pneumonia Viral/virologia , Pneumonia Viral/patologia , Pneumonia Viral/metabolismo , Betacoronavirus , Pulmão/virologia , Pulmão/patologia , Pulmão/metabolismo , Infecções por Coronavirus/virologia , Infecções por Coronavirus/patologia , Infecções por Coronavirus/complicações , Pandemias , Armadilhas Extracelulares/metabolismo
2.
Mol Immunol ; 74: 106-12, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27174188

RESUMO

We recently described a dominant role for conformational epitopes on the group 1 allergen of the mountain cedar (Juniperus ashei, Cupressaceae), Jun a 1, in pollen hypersensitivity in South Central U.S.A. Since these epitopes are surface exposed and are likely to be flexible, they may be susceptible to molecular or physical perturbations. This may make Jun a 1 a potential target for new forms of therapy for cedar pollinosis. Here, we describe a mouse monoclonal antibody, termed E58, which binds to the group 1 allergens of the cedar pollens from three highly populated regions of the world (central U.S.A., France and Japan). Upon binding to these allergens, E58 strongly reduces the binding of patient's IgE antibodies to these dominant allergens. This characteristic of E58, and potentially other similar antibodies, suggests an opportunity to develop preventative or therapeutic agents that may inhibit cedar pollen sensitization or prevent their allergic reactions.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Plantas/imunologia , Epitopos de Linfócito B/imunologia , Hipersensibilidade/imunologia , Proteínas de Plantas/imunologia , Alérgenos , Animais , Especificidade de Anticorpos , Cedrus/imunologia , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoglobulina E/imunologia , Camundongos , Pólen/imunologia , Ressonância de Plasmônio de Superfície
3.
Nanomedicine ; 11(2): 447-56, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25194998

RESUMO

Burkholderia mallei are Gram-negative bacteria, responsible for the disease glanders. B. mallei has recently been classified as a Tier 1 agent owing to the fact that this bacterial species can be weaponised for aerosol release, has a high mortality rate and demonstrates multi-drug resistance. Furthermore, there is no licensed vaccine available against this pathogen. Lipopolysaccharide (LPS) has previously been identified as playing an important role in generating host protection against Burkholderia infection. In this study, we present gold nanoparticles (AuNPs) functionalised with a glycoconjugate vaccine against glanders. AuNPs were covalently coupled with one of three different protein carriers (TetHc, Hcp1 and FliC) followed by conjugation to LPS purified from a non-virulent clonal relative, B. thailandensis. Glycoconjugated LPS generated significantly higher antibody titres compared with LPS alone. Further, they improved protection against a lethal inhalation challenge of B. mallei in the murine model of infection. FROM THE CLINICAL EDITOR: Burkholderia mallei is associated with multi-drug resistance, high mortality and potentials for weaponization through aerosol inhalation. The authors of this study present gold nanoparticles (AuNPs) functionalized with a glycoconjugate vaccine against this Gram negative bacterium demonstrating promising results in a murine model even with the aerosolized form of B. Mallei.


Assuntos
Vacinas Bacterianas/administração & dosagem , Burkholderia mallei/efeitos dos fármacos , Mormo/tratamento farmacológico , Nanopartículas Metálicas/administração & dosagem , Administração por Inalação , Animais , Vacinas Bacterianas/química , Burkholderia mallei/patogenicidade , Modelos Animais de Doenças , Mormo/imunologia , Mormo/microbiologia , Glicoconjugados/administração & dosagem , Glicoconjugados/química , Ouro/química , Humanos , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/imunologia , Nanopartículas Metálicas/química , Camundongos
5.
Clin Vaccine Immunol ; 21(5): 747-54, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24671550

RESUMO

The environmental Gram-negative encapsulated bacillus Burkholderia pseudomallei is the causative agent of melioidosis, a disease associated with high morbidity and mortality rates in areas of Southeast Asia and northern Australia in which the disease is endemic. B. pseudomallei is also classified as a tier I select agent due to the high level of lethality of the bacterium and its innate resistance to antibiotics, as well as the lack of an effective vaccine. Gram-negative bacteria, including B. pseudomallei, secrete outer membrane vesicles (OMVs) which are enriched with multiple protein, lipid, and polysaccharide antigens. Previously, we demonstrated that immunization with multivalent B. pseudomallei-derived OMVs protects highly susceptible BALB/c mice against an otherwise lethal aerosol challenge. In this work, we evaluated the protective efficacy of OMV immunization against intraperitoneal challenge with a heterologous strain because systemic infection with phenotypically diverse environmental B. pseudomallei strains poses another hazard and a challenge to vaccine development. We demonstrated that B. pseudomallei OMVs derived from strain 1026b afforded significant protection against septicemic infection with B. pseudomallei strain K96243. OMV immunization induced robust OMV-, lipopolysaccharide-, and capsular polysaccharide-specific serum IgG (IgG1, IgG2a, and IgG3) and IgM antibody responses. OMV-immune serum promoted bacterial killing in vitro, and passive transfer of B. pseudomallei OMV immune sera protected naive mice against a subsequent challenge. These results indicate that OMV immunization provides antibody-mediated protection against acute, rapidly lethal sepsis in mice. B. pseudomallei-derived OMVs may represent an efficacious multivalent vaccine strategy against melioidosis.


Assuntos
Vacinas Bacterianas/imunologia , Burkholderia pseudomallei/imunologia , Melioidose/prevenção & controle , Vesículas Secretórias/imunologia , Sepse/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Vacinas Bacterianas/administração & dosagem , Atividade Bactericida do Sangue , Proteção Cruzada , Feminino , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Camundongos Endogâmicos BALB C , Análise de Sobrevida
6.
PLoS One ; 8(5): e63331, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23691024

RESUMO

Mycobacterium tuberculosis (M.tb) is the second leading infectious cause of death worldwide and the primary cause of death in people living with HIV/AIDS. There are several excellent animal models employed to study tuberculosis (TB), but many have limitations for reproducing human pathology and none are amenable to the direct study of HIV/M.tb co-infection. The humanized mouse has been increasingly employed to explore HIV infection and other pathogens where animal models are limiting. Our goal was to develop a small animal model of M.tb infection using the bone marrow, liver, thymus (BLT) humanized mouse. NOD-SCID/γc(null) mice were engrafted with human fetal liver and thymus tissue, and supplemented with CD34(+) fetal liver cells. Excellent reconstitution, as measured by expression of the human CD45 pan leukocyte marker by peripheral blood populations, was observed at 12 weeks after engraftment. Human T cells (CD3, CD4, CD8), as well as natural killer cells and monocyte/macrophages were all observed within the human leukocyte (CD45(+)) population. Importantly, human T cells were functionally competent as determined by proliferative capacity and effector molecule (e.g. IFN-γ, granulysin, perforin) expression in response to positive stimuli. Animals infected intranasally with M.tb had progressive bacterial infection in the lung and dissemination to spleen and liver from 2-8 weeks post infection. Sites of infection in the lung were characterized by the formation of organized granulomatous lesions, caseous necrosis, bronchial obstruction, and crystallization of cholesterol deposits. Human T cells were distributed throughout the lung, liver, and spleen at sites of inflammation and bacterial growth and were organized to the periphery of granulomas. These preliminary results demonstrate the potential to use the humanized mouse as a model of experimental TB.


Assuntos
Modelos Animais de Doenças , Tuberculose/fisiopatologia , Animais , Transplante de Medula Óssea/métodos , Humanos , Fígado/citologia , Fígado/patologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Linfócitos T/imunologia , Timo/citologia
7.
PLoS One ; 7(4): e35386, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22530013

RESUMO

Burkholderia pseudomallei is a Gram-negative bacillus that is the causative agent of melioidosis. The bacterium is inherently resistant to many antibiotics and mortality rates remain high in endemic areas. The lipopolysaccharide (LPS) and capsular polysaccharide (CPS) are two surface-associated antigens that contribute to pathogenesis. We previously developed two monoclonal antibodies (mAbs) specific to the CPS and LPS; the CPS mAb was shown to identify antigen in serum and urine from melioidosis patients. The goal of this study was to determine if passive immunization with CPS and LPS mAbs alone and in combination would protect mice from a lethal challenge with B. pseudomallei. Intranasal (i.n.) challenge experiments were performed with B. pseudomallei strains 1026b and K96423. Both mAbs provided significant protection when administered alone. A combination of mAbs was protective when low doses were administered. In addition, combination therapy provided a significant reduction in spleen colony forming units (cfu) compared to results when either the CPS or LPS mAbs were administered alone.


Assuntos
Anticorpos Monoclonais/imunologia , Burkholderia pseudomallei/imunologia , Imunização Passiva , Melioidose/prevenção & controle , Polissacarídeos/imunologia , Abscesso/patologia , Animais , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/imunologia , Epitopos , Feminino , Melioidose/mortalidade , Melioidose/patologia , Camundongos , Camundongos Endogâmicos BALB C , Baço/microbiologia , Baço/patologia
8.
PLoS One ; 7(3): e34176, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22448290

RESUMO

Prophylactic administration of CpG oligodeoxynucleotides (CpG ODNs) is known to confer protection against lethal sepsis caused by Burkholderia pseudomallei in the mouse model. The mechanisms whereby CpG regulates the innate immune response to provide protection against B. pseudomallei, however, are poorly characterized. In the present study, we demonstrate that intranasal treatment of mice with Class C CpG, results in recruitment of inflammatory monocytes and neutrophils to the lung at 48 h post-treatment. Mice infected with B. pseudomallei 48 h post-CpG treatment had reduced organ bacterial load and significantly altered cytokine and chemokine profiles concomitant with protection as compared to control animals. CpG administration reduced the robust production of chemokines and pro-inflammatory cytokines in blood, lung and spleen, observed following infection of non-treated animals. Death of control animals coincided with the time of peak cytokine production (day 1-3), while a moderate; sustained cytokine production in CpG-treated animals was associated with survival. In general, CpG treatment resulted in diminished expression of cytokines and chemokines post-infection, though IL-12p40 was released in larger quantities in CpG treated animals. In contrast to CpG-treated animals, the lungs of infected control animals were infiltrated with leukocytes, especially neutrophils, and large numbers of necrotic lesions were observed in lung sections. Therapeutic treatment of B. pseudomallei-infected animals with CpG at 24 h post-infection did not impact survival compared to control animals. In summary, protection of CpG-treated animals was associated with recruitment of inflammatory monocytes and neutrophils into the lungs prior to infection. These responses correspond with early control of bacterial growth, a dampened inflammatory cytokine/chemokine response, reduced lung pathology, and greatly increased survival. In contrast, a delay in recruitment of inflammatory cell populations, despite a robust production of pro-inflammatory cytokines, was associated with poorly controlled bacterial growth, severe lung pathology, and death of control animals.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Burkholderia pseudomallei/patogenicidade , Imunidade Inata , Pulmão/imunologia , Melioidose/imunologia , Melioidose/prevenção & controle , Oligodesoxirribonucleotídeos/administração & dosagem , Animais , Burkholderia pseudomallei/isolamento & purificação , Quimiocinas/metabolismo , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Pulmão/microbiologia , Pulmão/patologia , Melioidose/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/imunologia , Monócitos/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Baço/imunologia , Baço/microbiologia , Baço/patologia , Taxa de Sobrevida
9.
Front Microbiol ; 2: 227, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22125550

RESUMO

Burkholderia are highly evolved Gram-negative bacteria that primarily infect solipeds but are transmitted to humans by ingestion and cutaneous or aerosol exposures. Heightened concern over human infections of Burkholderia mallei and the very closely related species B. pseudomallei is due to the pathogens' proven effectiveness as bioweapons, and to the increased potential for natural opportunistic infections in the growing diabetic and immuno-compromised populations. These Burkholderia species are nearly impervious to antibiotic treatments and no vaccine exists. In this study, the genome of the highly virulent B. mallei ATCC23344 strain was examined by expression library immunization for gene-encoded protective antigens. This protocol for genomic-scale functional screening was customized to accommodate the unusually large complexity of Burkholderia, and yielded 12 new putative vaccine candidates. Five of the candidates were individually tested as protein immunogens and three were found to confer significant partial protection against a lethal pulmonary infection in a murine model of disease. Determinations of peripheral blood cytokine and chemokine profiles following individual protein immunizations show that interleukin-2 (IL-2) and IL-4 are elicited by the three confirmed candidates, but unexpectedly interferon-γ and tumor necrosis factor-α are not. We suggest that these pathogen components, discovered using genetic immunization and confirmed in a conventional protein format, will be useful toward the development of a safe and effective glanders vaccine.

10.
Front Microbiol ; 2: 174, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21904535

RESUMO

Bioluminescent imaging (BLI) technology is a powerful tool for monitoring infectious disease progression and treatment approaches. BLI is particularly useful for tracking fastidious intracellular pathogens that might be difficult to recover from certain organs. Burkholderia mallei, the causative agent of glanders, is a facultative intracellular pathogen and has been classified by the CDC as a Category B select agent due to its highly infectious nature and potential use as a biological weapon. Very little is known regarding pathogenesis or treatment of glanders. We investigated the use of bioluminescent reporter constructs to monitor the dynamics of infection as well as the efficacy of therapeutics for B. mallei in real-time. A stable luminescent reporter B. mallei strain was created using the pUTmini-Tn5::luxKm2 plasmid and used to monitor glanders in the BALB/c murine model. Mice were infected via the intranasal route with 5 × 10(3) bacteria and monitored by BLI at 24, 48, and 72 h. We verified that our reporter construct maintained similar virulence and growth kinetics compared to wild-type B. mallei and confirmed that it maintains luminescent stability in the presence or absence of antibiotic selection. The luminescent signal was initially seen in the lungs, and progressed to the liver and spleen over the course of infection. We demonstrated that antibiotic treatment 24 h post-infection resulted in reduction of bioluminescence that can be attributed to decreased bacterial burden in target organs. These findings suggest that BLI can be used to monitor disease progression and efficacy of therapeutics during glanders infections. Finally, we report an alternative method to mini-Tn5::luxKm2 transposon using mini-Tn7-lux elements that insert site-specifically at known genomic attachment sites and that can also be used to tag bacteria.

11.
Faraday Discuss ; 149: 23-36; discussion 63-77, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21413172

RESUMO

Rapid detection of the category B biothreat agents Burkholderia pseudomallei and Burkholderia mallei in acute infections is critical to ensure that appropriate treatment is administered quickly to reduce an otherwise high probability of mortality (ca. 40% for B. pseudomallei). We are developing assays that can be used in clinical laboratories or security applications for the direct detection of surface-localized and secreted macromolecules produced by these organisms. We present our current medium-throughout approach for target selection and production of Burkholderia macromolecules and describe the generation of a Fab molecule targeted to the B. mallei BimA protein. We also present development of prototype assays for detecting Burkholderia species using anti-lipopolysaccharide antibodies.


Assuntos
Burkholderia mallei/isolamento & purificação , Burkholderia pseudomallei/isolamento & purificação , Ensaio de Imunoadsorção Enzimática/métodos , Mormo/microbiologia , Melioidose/microbiologia , Animais , Burkholderia mallei/metabolismo , Burkholderia pseudomallei/metabolismo , Chaperonina 60/química , Chaperonina 60/metabolismo , Mormo/diagnóstico , Humanos , Melioidose/diagnóstico , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/metabolismo
12.
Artigo em Inglês | MEDLINE | ID: mdl-24379895

RESUMO

Burkholderia mallei and B. pseudomallei are Gram-negative pathogenic bacteria, responsible for the diseases glanders and melioidosis, respectively. Furthermore, there is currently no vaccine available against these Burkholderia species. In this study, we aimed to identify protective proteins against these pathogens. Immunization with recombinant B. mallei Hcp1 (type VI secreted/structural protein), BimA (autotransporter protein), BopA (type III secreted protein), and B. pseudomallei LolC (ABC transporter protein) generated significant protection against lethal inhaled B. mallei ATCC23344 and B. pseudomallei 1026b challenge. Immunization with BopA elicited the greatest protective activity, resulting in 100% and 60% survival against B. mallei and B. pseudomallei challenge, respectively. Moreover, sera from recovered mice demonstrated reactivity with the recombinant proteins. Dendritic cells stimulated with each of the different recombinant proteins showed distinct cytokine patterns. In addition, T cells from immunized mice produced IFN-γ following in vitro re-stimulation. These results indicated therefore that it was possible to elicit cross-protective immunity against both B. mallei and B. pseudomallei by vaccinating animals with one or more novel recombinant proteins identified in B. mallei.

13.
BMC Microbiol ; 9: 88, 2009 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-19426516

RESUMO

BACKGROUND: Burkholderia mallei is a zoonotic Gram negative bacterium which primarily infects solipeds but can cause lethal disease in humans if left untreated. The effect of two antibiotics with different modes of action on Burkholderia mallei strain ATCC23344 was investigated by using in vitro and in vivo studies. RESULTS: Determination of minimal inhibitory concentrations (MICs) in vitro was done by the agar diffusion method and the dilution method. The MICs of levofloxacin and ceftazidime were in the similar range, 2.5 and 5.0 microg/ml, respectively. Intracellular susceptibility of the bacterium to these two antibiotics in J774A.1 mouse macrophages in vitro was also investigated. Macrophages treated with antibiotics demonstrated uptake of the drugs and reduced bacterial loads in vitro. The efficacy of ceftazidime and levofloxacin were studied in BALB/c mice as post-exposure treatment following intranasal B. mallei infection. Intranasal infection with 5 x 10(5) CFUs of B. mallei resulted in 90% death in non-treated control mice. Antibiotic treatments 10 days post-infection proved to be effective in vivo with all antibiotic treated mice surviving to day 34 post-infection. The antibiotics did not result in complete clearance of the bacterial infection and presence of the bacteria was found in lungs and spleens of the survivors, although bacterial burden recovered from levofloxacin treated animals appeared reduced compared to ceftazidime. CONCLUSION: Both antibiotics demonstrated utility for the treatment of glanders, including the ability for intracellular penetration and clearance of organisms in vitro.


Assuntos
Antibacterianos/farmacologia , Burkholderia mallei/efeitos dos fármacos , Ceftazidima/farmacologia , Levofloxacino , Ofloxacino/farmacologia , Animais , Linhagem Celular , Farmacorresistência Bacteriana , Feminino , Mormo/tratamento farmacológico , Mormo/microbiologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana
14.
Vaccine ; 27(30): 4064-73, 2009 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-19446933

RESUMO

Studying the mechanisms of host survival resulting from viral encephalitis is critical to the development of vaccines. Here we have shown in several independent studies that high dose treatment with neutralizing antibody prior to intranasal infection with Venezuelan equine encephalitis virus had an antiviral effect in the visceral organs and prolonged survival time of infected mice, even in the absence of alphabeta T cells. Nevertheless, antibody treatment did not prevent the development of lethal encephalitis. On the contrary, the adoptive transfer of primed CD4(+) T cells was necessary to prevent lethal encephalitis in mice lacking alphabeta T cell receptor.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/imunologia , Transferência Adotiva , Animais , Anticorpos Antivirais/administração & dosagem , Anticorpos Antivirais/imunologia , Encefalomielite Equina Venezuelana/prevenção & controle , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Testes de Neutralização , Receptores de Antígenos de Linfócitos T/deficiência , Análise de Sobrevida
15.
J Med Microbiol ; 58(Pt 5): 554-562, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19369515

RESUMO

Burkholderia mallei is a facultative intracellular pathogen that survives and replicates in phagocytic cell lines. The bacterial burden recovered from naïve BALB/c mice infected by intranasal delivery indicated that B. mallei persists in the lower respiratory system. To address whether B. mallei invades respiratory non-professional phagocytes, this study utilized A549 and LA-4 respiratory epithelial cells and demonstrated that B. mallei possesses the capacity to adhere poorly to, but not to invade, these cells. Furthermore, it was found that B. mallei was taken up by the murine alveolar macrophage cell line MH-S following serum coating, an attribute suggestive of complement- or Fc receptor-mediated uptake. Invasion/intracellular survival assays of B. mallei-infected MH-S cells demonstrated decreased intracellular survival, whilst a type III secretion system effector bopA mutant strain survived longer than the wild-type. Evaluation of the potential mechanism(s) responsible for efficient clearing of intracellular organisms demonstrated comparable levels of caspase-3 in both the wild-type and bopA mutant with characteristics consistent with apoptosis of infected MH-S cells. Furthermore, challenge of BALB/c mice with the bopA mutant by the intranasal route resulted in increased survival. Overall, these data suggest that B. mallei induces apoptotic cell death, whilst the BopA effector protein participates in intracellular survival.


Assuntos
Infecções por Burkholderia/patologia , Burkholderia mallei , Adenocarcinoma , Animais , Aderência Bacteriana , Burkholderia mallei/citologia , Burkholderia mallei/genética , Burkholderia mallei/isolamento & purificação , Burkholderia mallei/patogenicidade , Morte Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Primers do DNA , Humanos , Pulmão/microbiologia , Neoplasias Pulmonares , Macrófagos Alveolares/microbiologia , Macrófagos Alveolares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase , Mucosa Respiratória/microbiologia , Virulência
16.
Eur J Inorg Chem ; 2009(13): 1739-1745, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20160993

RESUMO

A series of N-heterocyclic carbene silver complexes have been synthesized and tested against the select group of bio-safety level 3 bacteria Burkholderia pseudomallei, Burkholderia mallei, Bacillus anthracis, methicillin-resistant Staphylococcus aureus and Yersinia pestis. Minimal inhibitory concentrations, minimal bactericidal and killing assays demonstrated the exceptional efficacy of the complexes against these potentially weaponizable pathogens.

17.
BMC Immunol ; 9: 55, 2008 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-18823549

RESUMO

BACKGROUND: We performed initial cell, cytokine and complement depletion studies to investigate the possible role of these effectors in response to vaccination with heat-killed Burkholderia mallei in a susceptible BALB/c mouse model of infection. RESULTS: While protection with heat-killed bacilli did not result in sterilizing immunity, limited protection was afforded against an otherwise lethal infection and provided insight into potential host protective mechanisms. Our results demonstrated that mice depleted of either B cells, TNF-alpha or IFN-gamma exhibited decreased survival rates, indicating a role for these effectors in obtaining partial protection from a lethal challenge by the intraperitoneal route. Additionally, complement depletion had no effect on immunoglobulin production when compared to non-complement depleted controls infected intranasally. CONCLUSION: The data provide a basis for future studies of protection via vaccination using either subunit or whole-organism vaccine preparations from lethal infection in the experimental BALB/c mouse model. The results of this study demonstrate participation of B220+ cells and pro-inflammatory cytokines IFN-gamma and TNF-alpha in protection following HK vaccination.


Assuntos
Vacinas Bacterianas/imunologia , Burkholderia mallei/imunologia , Mormo/imunologia , Imunidade Ativa , Vacinas de Produtos Inativados/imunologia , Animais , Anticorpos Bloqueadores/imunologia , Linfócitos B/imunologia , Vacinas Bacterianas/administração & dosagem , Interferon gama/imunologia , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Vacinação , Vacinas de Produtos Inativados/administração & dosagem
18.
Trans R Soc Trop Med Hyg ; 102 Suppl 1: S58-60, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19121690

RESUMO

We report the successful purification of lipopolysaccharide (LPS) from Burkholderia thailandensis, a Gram-negative bacterium, closely related to the highly pathogenic organisms B. pseudomallei and B. mallei. Burkholderia thailandensis LPS is shown to cross-react with rabbit and mouse sera obtained from inoculation with B. pseudomallei or B. mallei, respectively. These data suggest that B. thailandensis LPS shares similar structural features with LPS molecules from highly pathogenic Burkholderia species. This information may prove useful in ongoing efforts to develop novel vaccines and/or diagnostic reagents.


Assuntos
Burkholderia/química , Lipopolissacarídeos/isolamento & purificação , Animais , Lipopolissacarídeos/química , Camundongos , RNA Bacteriano/isolamento & purificação , Coelhos , Especificidade da Espécie
19.
Virology ; 367(2): 307-23, 2007 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-17610927

RESUMO

We evaluated the safety and immunogenicity of a chimeric alphavirus vaccine candidate in mice with selective immunodeficiencies. This vaccine candidate was highly attenuated in mice with deficiencies in the B and T cell compartments, as well as in mice with deficient gamma-interferon responsiveness. However, the level of protection varied among the strains tested. Wild type mice were protected against lethal VEEV challenge. In contrast, alpha/beta (alphabeta) TCR-deficient mice developed lethal encephalitis following VEEV challenge, while mice deficient in gamma/delta (gammadelta) T cells were protected. Surprisingly, the vaccine potency was diminished by 50% in animals lacking interferon-gamma receptor alpha chain (R1)-chain and a minority of vaccinated immunoglobulin heavy chain-deficient (microMT) mice survived challenge, which suggests that neutralizing antibody may not be absolutely required for protection. Prolonged replication of encephalitic VEEV in the brain of pre-immunized mice is not lethal and adoptive transfer experiments indicate that CD3(+) T cells are required for protection.


Assuntos
Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/prevenção & controle , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Subpopulações de Linfócitos T/imunologia , Vacinas Virais/imunologia , Animais , Modelos Animais de Doenças , Vírus da Encefalite Equina Venezuelana/metabolismo , Vírus da Encefalite Equina Venezuelana/fisiologia , Encefalomielite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/patologia , Encefalomielite Equina Venezuelana/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Segurança , Vacinação , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
20.
J Infect Dis ; 195(5): 680-3, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17262709

RESUMO

The protective effect of estrogen replacement on ascending urinary-tract infection (UTI) is controversial. We designed a study using an experimental model of UTI in which surgically menopausal mice were supplemented with estrogen and the susceptibility to UTI was evaluated after experimental Escherichia coli infection. The mean rate of E. coli infection in the group not treated with estrogen was 2 x 10(4) cfu/g of renal tissue, compared with 9 x 10(8) cfu/g (P<.001) in the estrogen-treated group. Surprisingly, despite the hypothesis that estrogen would protect mice from infection, estrogen treatment significantly increased the susceptibility of the mice to ascending UTI.


Assuntos
Cistite/prevenção & controle , Infecções por Escherichia coli/prevenção & controle , Estradiol/farmacologia , Estrogênios/metabolismo , Infecções Urinárias/prevenção & controle , Animais , Suscetibilidade a Doenças , Escherichia coli/classificação , Escherichia coli/metabolismo , Infecções por Escherichia coli/metabolismo , Feminino , Proteínas de Fímbrias , Menopausa , Camundongos , Camundongos Endogâmicos C3H , Ovariectomia , Infecções Urinárias/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA