Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Infect Immun ; 90(8): e0022222, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35862715

RESUMO

Burkholderia pseudomallei, the causative agent of melioidosis, is a facultative intracellular, Gram-negative pathogen that is highly infectious via the respiratory route and can cause severe, debilitating, and often fatal diseases in humans and animals. At present, no licensed vaccines for immunization against this CDC Tier 1 select agent exist. Studies in our lab have previously demonstrated that subunit vaccine formulations consisting of a B. pseudomallei capsular polysaccharide (CPS)-based glycoconjugate (CPS-CRM197) combined with hemolysin-coregulated protein (Hcp1) provided C57BL/6 mice with high-level protection against an acute inhalational challenge of B. pseudomallei. In this study, we evaluated the immunogenicity and protective capacity of B. pseudomallei alkyl hydroperoxide reductase subunit C (AhpC) in combination with CPS-CRM197. AhpC is a peroxiredoxin involved in oxidative stress reduction and is a potential protective antigen. To facilitate our studies and maximize safety in animals, recombinant B. pseudomallei AhpC harboring an active site mutation (AhpCC57G) was expressed in Escherichia coli and purified using tandem nickel-cobalt affinity chromatography. Immunization of C57BL/6 mice with CPS-CRM197 combined with AhpCC57G stimulated high-titer IgG responses against the CPS component of the glycoconjugate as well as stimulated high-titer IgG and robust interferon gamma (IFN-γ)-, interleukin-5 (IL-5)-, and IL-17-secreting T cell responses against AhpCC57G. When challenged via an inhalational route with a high dose (~27 50% lethal doses [LD50s]) of B. pseudomallei, 70% of the immunized mice survived 35 days postchallenge. Collectively, our findings demonstrate that AhpCC57G is a potent activator of cellular and humoral immune responses and may be a promising candidate to include in future melioidosis subunit vaccines.


Assuntos
Burkholderia pseudomallei , Melioidose , Animais , Anticorpos Antibacterianos , Vacinas Bacterianas , Burkholderia pseudomallei/genética , Glicoconjugados , Humanos , Imunoglobulina G , Melioidose/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vacinas de Subunidades Antigênicas/genética
2.
Infect Immun ; 86(1)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29109172

RESUMO

Burkholderia pseudomallei, the etiologic agent of melioidosis, causes severe disease in humans and animals. Diagnosis and treatment of melioidosis can be challenging, and no licensed vaccines currently exist. Several studies have shown that this pathogen expresses a variety of structurally conserved protective antigens that include cell surface polysaccharides and cell-associated and cell-secreted proteins. Based on those findings, such antigens have become important components of the subunit vaccine candidates that we are currently developing. In the present study, the 6-deoxyheptan capsular polysaccharide (CPS) from B. pseudomallei was purified, chemically activated, and covalently linked to recombinant CRM197 diphtheria toxin mutant (CRM197) to produce CPS-CRM197. Additionally, tandem nickel-cobalt affinity chromatography was used to prepare highly purified recombinant B. pseudomallei Hcp1 and TssM proteins. Immunization of C57BL/6 mice with CPS-CRM197 produced high-titer IgG and opsonizing antibody responses against the CPS component of the glycoconjugate, while immunization with Hcp1 and TssM produced high-titer IgG and robust gamma interferon-secreting T cell responses against the proteins. Extending upon these studies, we found that when mice were vaccinated with a combination of CPS-CRM197 and Hcp1, 100% of the mice survived a lethal inhalational challenge with B. pseudomallei Remarkably, 70% of the survivors had no culturable bacteria in their lungs, livers, or spleens, indicating that the vaccine formulation had generated sterilizing immune responses. Collectively, these studies help to better establish surrogates of antigen-induced immunity against B. pseudomallei as well as provide valuable insights toward the development of a safe, affordable, and effective melioidosis vaccine.


Assuntos
Vacinas Bacterianas/imunologia , Melioidose/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Burkholderia pseudomallei , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Subunidades Proteicas/imunologia , Vacinas de Subunidades Antigênicas
3.
Nat Commun ; 8(1): 115, 2017 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-28740137

RESUMO

Burkholderia pseudomallei (Bp) and Burkholderia mallei (Bm), the etiologic agents of melioidosis and glanders, respectively, cause severe disease in both humans and animals. Studies have highlighted the importance of Bp and Bm lipopolysaccharides (LPS) as vaccine candidates. Here we describe the synthesis of seven oligosaccharides as the minimal structures featuring all of the reported acetylation/methylation patterns associated with Bp and Bm LPS O-antigens (OAgs). Our approach is based on the conversion of an L-rhamnose into a 6-deoxy-L-talose residue at a late stage of the synthetic sequence. Using biochemical and biophysical methods, we demonstrate the binding of several Bp and Bm LPS-specific monoclonal antibodies with terminal OAg residues. Mice immunized with terminal disaccharide-CRM197 constructs produced high-titer antibody responses that crossreacted with Bm-like OAgs. Collectively, these studies serve as foundation for the development of novel therapeutics, diagnostics, and vaccine candidates to combat diseases caused by Bp and Bm.Melioidosis and glanders are multifaceted infections caused by gram-negative bacteria. Here, the authors synthesize a series of oligosaccharides that mimic the lipopolysaccharides present on the pathogens' surface and use them to develop novel glycoconjugates for vaccine development.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Bactérias/metabolismo , Burkholderia mallei/metabolismo , Burkholderia pseudomallei/metabolismo , Epitopos/imunologia , Lipopolissacarídeos/imunologia , Animais , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Vacinas Bacterianas/imunologia , Burkholderia mallei/genética , Burkholderia pseudomallei/genética , Feminino , Regulação Bacteriana da Expressão Gênica/fisiologia , Lipopolissacarídeos/química , Lipopolissacarídeos/metabolismo , Melioidose/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C
4.
PLoS One ; 10(4): e0124181, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25860021

RESUMO

Burkholderia mallei is a host-adapted bacterium that does not persist outside of its equine reservoir. The organism causes the zoonosis glanders, which is endemic in Asia, Africa, the Middle East and South America. Infection by B. mallei typically occurs via the respiratory or percutaneous route, and the most common manifestations are life-threatening pneumonia and bacteremia. Glanders is difficult to diagnose and requires prolonged antibiotic therapy with low success rates. There is no vaccine to protect against B. mallei and there is concern regarding its use as a biothreat agent. Thus, experiments were performed to establish a non-human primate model of intranasal infection to study the organism and develop countermeasures. Groups of marmosets (Callithrix jacchus) were inoculated intranasally with B. mallei strain ATCC 23344 and monitored for clinical signs of illness for up to 13 days. We discovered that 83% of marmosets inoculated with doses of 2.5 X 10(4) to 2.5 X 10(5) bacteria developed acute lethal infection within 3-4 days. Signs of disease were severe and included lethargy, inappetence, conjunctivitis, mucopurulent and hemorrhagic nasal discharges, and increased respiratory effort with abdominal lifts. Burkholderia mallei was cultured from the lungs, spleen and liver of these animals, and pathologic examination of tissues revealed lesions characteristic of glanders. Challenge experiments also revealed that 91% of animals infected with doses ranging from 25 to 2.5 X 10(3) bacteria exhibited mild non-specific signs of illness and were culture negative. One marmoset inoculated with 2.5 X 10(3) organisms developed moderate signs of disease and reached humane end-points 8 days post-infection. The liver and spleen of this animal were colonized with the agent and pathological analysis of tissues showed nasal, splenic and hepatic lesions. Taken together, these data indicate that the marmoset is a suitable model to study respiratory infection by B. mallei.


Assuntos
Burkholderia mallei/patogenicidade , Callithrix/microbiologia , Mormo/etiologia , Administração Intranasal , Animais , Carga Bacteriana , Modelos Animais de Doenças , Feminino , Mormo/patologia , Mormo/transmissão , Cavalos , Humanos , Fígado/microbiologia , Fígado/patologia , Pulmão/microbiologia , Pulmão/patologia , Masculino , Especificidade da Espécie , Baço/microbiologia , Baço/patologia , Zoonoses/etiologia , Zoonoses/patologia , Zoonoses/transmissão
5.
PLoS One ; 8(10): e76804, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098563

RESUMO

Burkholderia pseudomallei, the etiologic agent of melioidosis, is a saprophytic bacterium readily isolated from wet soils of countries bordering the equator. Burkholderia mallei is a host-adapted clone of B. pseudomallei that does not persist outside of its equine reservoir and causes the zoonosis glanders, which is endemic in Asia, Africa, the Middle East and South America. Infection by these organisms typically occurs via percutaneous inoculation or inhalation of aerosols, and the most common manifestation is severe pneumonia leading to fatal bacteremia. Glanders and melioidosis are difficult to diagnose and require prolonged antibiotic therapy with low success rates. There are no vaccines available to protect against either Burkholderia species, and there is concern regarding their use as biological warfare agents given that B. mallei has previously been utilized in this manner. Hence, experiments were performed to establish a mouse model of aerosol infection to study the organisms and develop countermeasures. Using a hand-held aerosolizer, BALB/c mice were inoculated intratracheally with strains B. pseudomallei 1026b and B. mallei ATCC23344 and growth of the agents in the lungs, as well as dissemination to the spleen, were examined. Mice infected with 10(2), 10(3) and 10(4) organisms were unable to control growth of B. mallei in the lungs and bacteria rapidly disseminated to the spleen. Though similar results were observed in mice inoculated with 10(3) and 10(4) B. pseudomallei cells, animals infected with 10(2) organisms controlled bacterial replication in the lungs, dissemination to the spleen, and the extent of bacteremia. Analysis of sera from mice surviving acute infection revealed that animals produced antibodies against antigens known to be targets of the immune response in humans. Taken together, these data show that small volume aerosol inoculation of mice results in acute disease, dose-dependent chronic infection, and immune responses that correlate with those seen in human infections.


Assuntos
Aerossóis/administração & dosagem , Anticorpos Antibacterianos/sangue , Bacteriemia/imunologia , Burkholderia mallei/imunologia , Burkholderia pseudomallei/imunologia , Mormo/imunologia , Melioidose/imunologia , Administração por Inalação , Animais , Bacteriemia/sangue , Bacteriemia/microbiologia , Bacteriemia/patologia , Armas Biológicas , Burkholderia mallei/patogenicidade , Burkholderia pseudomallei/patogenicidade , Modelos Animais de Doenças , Feminino , Mormo/sangue , Mormo/microbiologia , Mormo/patologia , Cavalos , Humanos , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Melioidose/sangue , Melioidose/microbiologia , Melioidose/patologia , Camundongos , Camundongos Endogâmicos BALB C , Baço/imunologia , Baço/microbiologia , Baço/patologia
6.
PLoS One ; 8(7): e67881, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23844117

RESUMO

Moraxella catarrhalis causes significant health problems, including 15-20% of otitis media cases in children and ~10% of respiratory infections in adults with chronic obstructive pulmonary disease. The lack of an efficacious vaccine, the rapid emergence of antibiotic resistance in clinical isolates, and high carriage rates reported in children are cause for concern. In addition, the effectiveness of conjugate vaccines at reducing the incidence of otitis media caused by Streptococcus pneumoniae and nontypeable Haemophilus influenzae suggest that M. catarrhalis infections may become even more prevalent. Hence, M. catarrhalis is an important and emerging cause of infectious disease for which the development of a vaccine is highly desirable. Studying the pathogenesis of M. catarrhalis and the testing of vaccine candidates have both been hindered by the lack of an animal model that mimics human colonization and infection. To address this, we intranasally infected chinchilla with M. catarrhalis to investigate colonization and examine the efficacy of a protein-based vaccine. The data reveal that infected chinchillas produce antibodies against antigens known to be major targets of the immune response in humans, thus establishing immune parallels between chinchillas and humans during M. catarrhalis infection. Our data also demonstrate that a mutant lacking expression of the adherence proteins MhaB1 and MhaB2 is impaired in its ability to colonize the chinchilla nasopharynx, and that immunization with a polypeptide shared by MhaB1 and MhaB2 elicits antibodies interfering with colonization. These findings underscore the importance of adherence proteins in colonization and emphasize the relevance of the chinchilla model to study M. catarrhalis-host interactions.


Assuntos
Adesinas Bacterianas/imunologia , Proteínas de Bactérias/imunologia , Chinchila/imunologia , Moraxella catarrhalis/imunologia , Infecções por Moraxellaceae/imunologia , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Animais , Anticorpos Antibacterianos/imunologia , Aderência Bacteriana/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Vacinas Bacterianas/metabolismo , Western Blotting , Linhagem Celular Tumoral , Chinchila/microbiologia , Modelos Animais de Doenças , Haemophilus influenzae/imunologia , Haemophilus influenzae/fisiologia , Hemaglutininas/genética , Hemaglutininas/imunologia , Hemaglutininas/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Moraxella catarrhalis/genética , Moraxella catarrhalis/fisiologia , Infecções por Moraxellaceae/microbiologia , Mutação , Nasofaringe/imunologia , Nasofaringe/microbiologia , Otite Média/imunologia , Otite Média/microbiologia , Streptococcus pneumoniae/imunologia , Streptococcus pneumoniae/fisiologia , Vacinação/métodos
7.
BMC Microbiol ; 13: 140, 2013 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-23782650

RESUMO

BACKGROUND: Moraxella catarrhalis is a human-specific gram-negative bacterium readily isolated from the respiratory tract of healthy individuals. The organism also causes significant health problems, including 15-20% of otitis media cases in children and ~10% of respiratory infections in adults with chronic obstructive pulmonary disease. The lack of an efficacious vaccine, the rapid emergence of antibiotic resistance in clinical isolates, and high carriage rates reported in children are cause for concern. Virtually all Moraxella catarrhalis isolates are resistant to ß-lactam antibiotics, which are generally the first antibiotics prescribed to treat otitis media in children. The enzymes responsible for this resistance, BRO-1 and BRO-2, are lipoproteins and the mechanism by which they are secreted to the periplasm of M. catarrhalis cells has not been described. RESULTS: Comparative genomic analyses identified M. catarrhalis gene products resembling the TatA, TatB, and TatC proteins of the well-characterized Twin Arginine Translocation (TAT) secretory apparatus. Mutations in the M. catarrhalis tatA, tatB and tatC genes revealed that the proteins are necessary for optimal growth and resistance to ß-lactams. Site-directed mutagenesis was used to replace highly-conserved twin arginine residues in the predicted signal sequence of M. catarrhalis strain O35E BRO-2, which abolished resistance to the ß-lactam antibiotic carbanecillin. CONCLUSIONS: Moraxella catarrhalis possesses a TAT secretory apparatus, which plays a key role in growth of the organism and is necessary for secretion of BRO-2 into the periplasm where the enzyme can protect the peptidoglycan cell wall from the antimicrobial activity of ß-lactam antibiotics.


Assuntos
Proteínas de Membrana Transportadoras/metabolismo , Moraxella catarrhalis/metabolismo , beta-Lactamases/metabolismo , Adulto , Criança , Pré-Escolar , Biologia Computacional , Análise Mutacional de DNA , Técnicas de Inativação de Genes , Genoma Bacteriano , Humanos , Proteínas de Membrana Transportadoras/genética , Moraxella catarrhalis/efeitos dos fármacos , Moraxella catarrhalis/genética , Moraxella catarrhalis/crescimento & desenvolvimento , Moraxella catarrhalis/isolamento & purificação , Infecções por Moraxellaceae/microbiologia , Mutagênese Sítio-Dirigida , Resistência beta-Lactâmica
8.
Artigo em Inglês | MEDLINE | ID: mdl-20426171

RESUMO

Our long term research goal is to develop a fully automated, image-based diagnostic system for early diagnosis of pulmonary nodules that may lead to lung cancer. In this paper, we focus on generating new probabilistic models for the estimated growth rate of the detected lung nodules from Low Dose Computed Tomography (LDCT). We propose a new methodology for 3D LDCT data registration which is non-rigid and involves two steps: (i) global target-to-prototype alignment of one scan to another using the learned prior appearance model followed by (ii) local alignment in order to correct for intricate relative deformations. Visual appearance of these chest images is described using a Markov-Gibbs random field (MGRF) model with multiple pairwise interaction. An affine transformation that globally registers a target to a prototype is estimated by the gradient ascent-based maximization of a special Gibbs energy function. To handle local deformations, we displace each voxel of the target over evolving closed equi-spaced surfaces (iso-surfaces) to closely match the prototype. The evolution of the iso-surfaces is guided by a speed function in the directions that minimize distances between the corresponding voxel pairs on the iso-surfaces in both the data sets. Preliminary results show that the proposed accurate registration could lead to precise diagnosis and identification of the development of the detected pulmonary nodules.


Assuntos
Algoritmos , Imageamento Tridimensional/métodos , Neoplasias Pulmonares/diagnóstico por imagem , Reconhecimento Automatizado de Padrão/métodos , Interpretação de Imagem Radiográfica Assistida por Computador/métodos , Tomografia Computadorizada por Raios X/métodos , Humanos , Intensificação de Imagem Radiográfica/métodos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
9.
Med Image Comput Comput Assist Interv ; 11(Pt 1): 322-30, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18979763

RESUMO

New techniques for more accurate unsupervised segmentation of lung tissues from Low Dose Computed Tomography (LDCT) are proposed. In this paper we describe LDCT images and desired maps of regions (lung and the other chest tissues) by a joint Markov-Gibbs random field model (MGRF) of independent image signals and interdependent region labels but focus on most accurate model identification. To better specify region borders, each empirical distribution of signals is precisely approximated by a Linear Combination of Discrete Gaussians (LCDG) with positive and negative components. We modify a conventional Expectation-Maximization (EM) algorithm to deal with the LCDG and develop a sequential EM-based technique to get an initial LCDG-approximation for the modified EM algorithm. The initial segmentation based on the LCDG-models is then iteratively refined using a MGRF model with analytically estimated potentials. Experiments on real data sets confirm high accuracy of the proposed approach.


Assuntos
Algoritmos , Inteligência Artificial , Pulmão/diagnóstico por imagem , Reconhecimento Automatizado de Padrão/métodos , Interpretação de Imagem Radiográfica Assistida por Computador/métodos , Tomografia Computadorizada por Raios X/métodos , Simulação por Computador , Interpretação Estatística de Dados , Humanos , Modelos Biológicos , Modelos Estatísticos , Intensificação de Imagem Radiográfica/métodos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Processos Estocásticos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...