Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Microorganisms ; 11(7)2023 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-37512916

RESUMEN

One pathogen that commonly causes gastrointestinal illnesses from the consumption of contaminated food is Escherichia coli O157:H7. In 2011 in Germany, however, there was a prominent outbreak of bloody diarrhea with a high incidence of hemolytic uremic syndrome (HUS) caused by an atypical, more virulent E. coli O104:H4 strain. To facilitate the identification of this lesser-known, atypical E. coli O104:H4 strain, we wanted to identify phenotypic differences between it and a strain of O157:H7 in different media and culture conditions. We found that E. coli O104:H4 strains produced considerably more biofilm than the strain of O157:H7 at 37 °C (p = 0.0470-0.0182) Biofilm production was significantly enhanced by the presence of 5% CO2 (p = 0.0348-0.0320). In our study on the innate immune response to the E. coli strains, we used HEK293 cells that express Toll-like receptors (TLRs) 2 or 4. We found that E. coli O104:H4 strains had the ability to grow in a novel HEK293 cell culture medium, while the E. coli O157:H7 strain could not. Thus, we uncovered previously unknown phenotypic properties of E. coli O104:H4 to further differentiate this pathogen from E. coli O157:H7.

2.
Front Cell Infect Microbiol ; 11: 745325, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34888257

RESUMEN

Successful bacterial pathogens have evolved to avoid activating an innate immune system in the host that responds to the pathogen through distinct Toll-like receptors (TLRs). The general class of biochemical components that activate TLRs has been studied extensively, but less is known about how TLRs interact with the class of compounds that are still associated with the live pathogen. Accordingly, we examined the activation of surface assembled TLR 2, 4, and 5 with live Tier 1 Gram-negative pathogens that included Yersinia pestis (plague), Burkholderia mallei (glanders), Burkholderia pseudomallei (melioidosis), and Francisella tularensis (tularemia). We found that Y. pestis CO92 grown at 28°C activated TLR2 and TLR4, but at 37°C the pathogen activated primarily TLR2. Although B. mallei and B. pseudomallei are genetically related, the former microorganism activated predominately TLR4, while the latter activated predominately TLR2. The capsule of wild-type B. pseudomallei 1026b was found to mitigate the activation of TLR2 and TLR4 when compared to a capsule mutant. Live F. tularensis (Ft) Schu S4 did not activate TLR2 or 4, although the less virulent Ft LVS and F. novicida activated only TLR2. B. pseudomallei purified flagellin or flagella attached to the microorganism activated TLR5. Activation of TLR5 was abolished by an antibody to TLR5, or a mutation of fliC, or elimination of the pathogen by filtration. In conclusion, we have uncovered new properties of the Gram-negative pathogens, and their interaction with TLRs of the host. Further studies are needed to include other microorganism to extend our observations with their interaction with TLRs, and to the possibility of leading to new efforts in therapeutics against these pathogens.


Asunto(s)
Melioidosis , Receptor Toll-Like 4 , Animales , Flagelos , Receptor Toll-Like 4/genética , Receptor Toll-Like 5 , Receptores Toll-Like
3.
Front Immunol ; 12: 726416, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34512658

RESUMEN

Relatively recent advances in plague vaccinology have produced the recombinant fusion protein F1-V plague vaccine. This vaccine has been shown to readily protect mice from both bubonic and pneumonic plague. The protection afforded by this vaccine is solely based upon the immune response elicited by the F1 or V epitopes expressed on the F1-V fusion protein. Accordingly, questions remain surrounding its efficacy against infection with non-encapsulated (F1-negative) strains. In an attempt to further optimize the F1-V elicited immune response and address efficacy concerns, we examined the inclusion of multiple toll-like receptor agonists into vaccine regimens. We examined the resulting immune responses and also any protection afforded to mice that were exposed to aerosolized Yersinia pestis. Our data demonstrate that it is possible to further augment the F1-V vaccine strategy in order to optimize and augment vaccine efficacy.


Asunto(s)
Adyuvantes Inmunológicos , Antígenos Bacterianos/inmunología , Vacuna contra la Peste/inmunología , Peste/prevención & control , Receptores Toll-Like/fisiología , Animales , Femenino , Ratones , Ratones Endogámicos BALB C , Peste/inmunología , Vacunación , Eficacia de las Vacunas , Vacunas Sintéticas/inmunología , Yersinia pestis/inmunología
4.
Microb Pathog ; 155: 104919, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33915206

RESUMEN

Burkholderia mallei is a gram-negative obligate animal pathogen that causes glanders, a highly contagious and potentially fatal disease of solipeds including horses, mules, and donkeys. Humans are also susceptible, and exposure can result in a wide range of clinical forms, i.e., subclinical infection, chronic forms with remission and exacerbation, or acute and potentially lethal septicemia and/or pneumonia. Due to intrinsic antibiotic resistance and the ability of the organisms to survive intracellularly, current treatment regimens are protracted and complicated; and no vaccine is available. As a consequence of these issues, and since B. mallei is infectious by the aerosol route, B. mallei is regarded as a major potential biothreat agent. To develop optimal medical countermeasures and diagnostic tests, well characterized animal models of human glanders are needed. The goal of this study was to perform a head-to-head comparison of models employing three commonly used nonhuman primate (NHP) species, the African green monkey (AGM), Rhesus macaque, and the Cynomolgus macaque. The natural history of infection and in vitro clinical, histopathological, immunochemical, and bacteriological parameters were examined. The AGMs were the most susceptible NHP to B. mallei; five of six expired within 14 days. Although none of the Rhesus or Cynomolgus macaques succumbed, the Rhesus monkeys exhibited abnormal signs and clinical findings associated with B. mallei infection; and the latter may be useful for modeling chronic B. mallei infection. Based on the disease progression observations, gross and histochemical pathology, and humoral and cellular immune response findings, the AGM appears to be the optimal model of acute, lethal glanders infection. AGM models of infection by B. pseudomallei, the etiologic agent of melioidosis, have been characterized recently. Thus, the selection of the AGM species provides the research community with a single NHP model for investigations on acute, severe, inhalational melioidosis and glanders.


Asunto(s)
Burkholderia mallei , Burkholderia pseudomallei , Muermo , Melioidosis , Aerosoles , Animales , Chlorocebus aethiops , Modelos Animales de Enfermedad , Muermo/diagnóstico , Caballos , Macaca mulatta
5.
PLoS Negl Trop Dis ; 15(2): e0009125, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33571211

RESUMEN

Melioidosis, caused by the Gram-negative bacterium Burkholderia pseudomallei, is a major cause of sepsis and mortality in endemic regions of Southeast Asia and Northern Australia. B. pseudomallei is a potential bioterrorism agent due to its high infectivity, especially via inhalation, and its inherent resistance to antimicrobials. There is currently no vaccine for melioidosis and antibiotic treatment can fail due to innate drug resistance, delayed diagnosis and treatment, or insufficient duration of treatment. A well-characterized animal model that mimics human melioidosis is needed for the development of new medical countermeasures. This study first characterized the disease progression of melioidosis in the African green monkey (AGM) and rhesus macaque (RM) for non-human primate model down-selection. All AGMs developed acute lethal disease similar to that described in human acute infection following exposure to aerosolized B. pseudomallei strain HBPUB10134a. Only 20% of RMs succumbed to acute disease. Disease progression, immune response and pathology of two other strains of B. pseudomallei, K96243 and MSHR5855, were also compared using AGMs. These three B. pseudomallei strains represent a highly virulent strain from Thailand (HBPUB101034a), a highly virulent strains from Australia (MSHR5855), and a commonly used laboratory strains originating from Thailand (K96243). Animals were observed for clinical signs of infection and blood samples were analyzed for cytokine responses, blood chemistry and leukocyte changes in order to characterize bacterial infection. AGMs experienced fever after exposure to aerosolized B. pseudomallei at the onset of acute disease. Inflammation, abscesses and/or pyogranulomas were observed in lung with all three strains of B. pseudomallei. Inflammation, abscesses and/or pyogranulomas were observed in lymph nodes, spleen, liver and/or kidney with B. pseudomallei, HBPUB10134a and K96243. Additionally, the Australian strain MSHR5855 induced brain lesions in one AGM similar to clinical cases of melioidosis seen in Australia. Elevated serum levels of IL-1ß, IL-1 receptor antagonist, IL-6, MCP-1, G-CSF, HGF, IFNγ, MIG, I-TAC, and MIP-1ß at terminal end points can be significantly correlated with non-survivors with B. pseudomallei infection in AGM. The AGM model represents an acute model of B. pseudomallei infection for all three strains from two geographical locations and will be useful for efficacy testing of vaccines and therapeutics against melioidosis. In summary, a dysregulated immune response leading to excessive persistent inflammation and inflammatory cell death is the key driver of acute melioidosis. Early intervention in these pathways will be necessary to counter B. pseudomallei and mitigate the pathological consequences of melioidosis.


Asunto(s)
Aerosoles , Burkholderia pseudomallei , Melioidosis/microbiología , Melioidosis/patología , Animales , Asia Sudoriental , Australia , Bacteriemia , Médula Ósea/patología , Quimiocinas/metabolismo , Chlorocebus aethiops , Citocinas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Hígado/patología , Pulmón/patología , Macaca mulatta , Bazo/patología , Telemetría , Tailandia , Virulencia
6.
Antibodies (Basel) ; 9(3)2020 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-32756297

RESUMEN

Plague is a zoonotic disease that is caused by Yersinia pestis. Monoclonal antibodies (mAbs) that bind to the V-antigen, a virulence factor that is produced by Y. pestis, can passively protect mice from plague. An analysis of protective mAbs that bind to V-antigen was made to assess binding sites, avidities, and affinities. Anti-V mAbs were screened for their efficacy in a murine model of plague. Antigen-binding sites of protective V mAbs were determined with a linear peptide library, V-antigen fragment, competitive binding, and surface plasmon resonance. The avidities to the V-antigen was determined by ELISA, and affinities of the mAbs to the V-antigen were determined by surface plasmon resonance. The most protective mAb 7.3 bound to a unique conformational site on the V-antigen, while a less protective mAb bound to a different conformational site located on the same V-antigen fragment as mAb 7.3. The avidity of mAb 7.3 for the V-antigen was neither the strongest overall nor did it have the highest affinity for the V-antigen. The binding site of the most protective mAb was critical in its ability to protect against a lethal plague challenge.

7.
Trop Med Infect Dis ; 5(2)2020 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-32365605

RESUMEN

Burkholderia pseudomallei and B. mallei are Gram-negative, facultative intracellular bacteria that cause melioidosis and glanders, respectively. Currently, there are no vaccines for these two diseases. Animal models have been developed to evaluate vaccines and therapeutics. Tissues from infected animals, however, must be fixed in formalin and embedded in paraffin (FFPE) before analysis. A brownish staining material in infected tissues that represents the exopolysaccharide of the pathogen was seen by bright field microscopy but not the actual microorganism. Because of these results, FFPE tissue was examined by laser scanning confocal microscopy (LSCM) in an attempt to see the microorganism. Archival FFPE tissues were examined from ten mice, and five nonhuman primates after exposure to B. pseudomallei or B. mallei by LSCM. Additionally, a historical spleen biopsy from a human suspected of exposure to B. mallei was examined. B. pseudomallei was seen in many of the infected tissues from mice. Four out of five nonhuman primates were positive for the pathogen. In the human sample, B. mallei was seen in pyogranulomas in the spleen biopsy. Thus, the presence of the pathogen was validated by LSCM in murine, nonhuman primate, and human FFPE tissues.

8.
BMC Immunol ; 21(1): 5, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-32013893

RESUMEN

BACKGROUND: Melioidosis is endemic in Southeast Asia and Northern Australia and is caused by the Gram-negative, facultative intracellular pathogen Burkholderia pseudomallei. Diagnosis of melioidosis is often difficult because of the protean clinical presentation of the disease, and it may mimic other diseases, such as tuberculosis. There are many different strains of B. pseudomallei that have been isolated from patients with melioidosis, but it was not clear if they could cause a similar disease in a chronic BALB/c murine model of melioidosis. Hence, we wanted to examine chronically infected mice exposed to different strains of B. pseudomallei to determine if there were differences in the host immune response to the pathogen. RESULTS: We identified common host immune responses exhibited in chronically infected BALB/c mice, although there was some heterogeneity in the host response in chronically infected mice after exposure to different strains of B. pseudomallei. They all displayed pyogranulomatous lesions in their spleens with a large influx of monocytes/macrophages, NK cells, and neutrophils identified by flow cytometry. Sera from chronically infected mice by ELISA exhibited elevated IgG titers to the pathogen, and we detected by Luminex micro-bead array technology a significant increase in the expression of inflammatory cytokines/chemokines, such as IFN-γ, IL-1α, IL-1ß, KC, and MIG. By immunohistochemical and in situ RNA hybridization analysis we found that the increased expression of proinflammatory cytokines (IL-1α, IL-1ß, TNF-α, IFN-γ) was confined primarily to the area with the pathogen within pyogranulomatous lesions. We also found that cultured splenocytes from chronically infected mice could express IFN-γ, TNF-α, and MIP-1α ex vivo without the need for additional exogenous stimulation. In addition by flow cytometry, we detected significant amounts of intracellular expression of TNF-α and IFN-γ without a protein transport blocker in monocytes/macrophages, NK cells, and neutrophils but not in CD4+ or CD8+ T cells in splenocytes from chronically infected mice. CONCLUSION: Taken together the common features we have identified in chronically infected mice when 10 different human clinical strains of B. pseudomallei were examined could serve as biomarkers when evaluating potential therapeutic agents in mice for the treatment of chronic melioidosis in humans.


Asunto(s)
Burkholderia pseudomallei/fisiología , Interferón gamma/metabolismo , Melioidosis/inmunología , Bazo/patología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Humanos , Inmunidad Celular , Ratones , Ratones Endogámicos BALB C
9.
Vaccines (Basel) ; 7(4)2019 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-31779073

RESUMEN

Melioidosis is an emerging disease that is caused by the facultative intracellular pathogen Burkholderia pseudomallei. It is intrinsically resistant to many antibiotics and host risk factors play a major role in susceptibility to infection. Currently, there is no human or animal vaccine against melioidosis. In this study, multiple B. pseudomallei MSHR668 deletion mutants were evaluated as live attenuated vaccines in the sensitive BALB/c mouse model of melioidosis. The most efficacious vaccines after an intraperitoneal challenge with 50-fold over the 50% median lethal dose (MLD50) with B. pseudomallei K96243 were 668 ΔhisF and 668 ΔilvI. Both vaccines completely protected mice in the acute phase of infection and showed significant protection (50% survivors) during the chronic phase of infection. The spleens of the survivors that were examined were sterile. Splenocytes from mice vaccinated with 668 ΔhisF and 668 ΔilvI expressed higher amounts of IFN-γ after stimulation with B. pseudomallei antigens than splenocytes from mice vaccinated with less protective candidates. Finally, we demonstrate that 668 ΔhisF is nonlethal in immunocompromised NOD/SCID mice. Our results show that 668 ΔhisF and 668 ΔilvI provide protective cell-mediated immune responses in the acute phase of infection and promote long term survival in the sensitive BALB/c mouse model of melioidosis.

10.
J Med Microbiol ; 68(2): 263-278, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30628877

RESUMEN

PURPOSE: Burkholderia pseudomallei, the tier 1 agent of melioidosis, is a saprophytic microbe that causes endemic infections in tropical regions such as South-East Asia and Northern Australia. It is globally distributed, challenging to diagnose and treat, infectious by several routes including inhalation, and has potential for adversarial use. B. pseudomallei strain MSHR5848 produces two colony variants, smooth (S) and rough (R), which exhibit a divergent range of morphological, biochemical and metabolic phenotypes, and differ in macrophage and animal infectivity. We aimed to characterize two major phenotypic differences, analyse gene expression and study the regulatory basis of the variation. METHODOLOGY: Phenotypic expression was characterized by DNA and RNA sequencing, microscopy, and differential bacteriology. Regulatory genes were identified by cloning and bioinformatics.Results/Key findings. Whereas S produced larger quantities of extracellular DNA, R was upregulated in the production of a unique chromosome 1-encoded Siphoviridae-like bacteriophage, φMSHR5848. Exploratory transcriptional analyses revealed significant differences in variant expression of genes encoding siderophores, pili assembly, type VI secretion system cluster 4 (T6SS-4) proteins, several exopolysaccharides and secondary metabolites. A single 3 base duplication in S was the only difference that separated the variants genetically. It occurred upstream of a cluster of bacteriophage-associated genes on chromosome 2 that were upregulated in S. The first two genes were involved in regulating expression of the multiple phenotypes distinguishing S and R. CONCLUSION: Bacteriophage-associated proteins have a major role in the phenotypic expression of MSHR5848. The goals are to determine the regulatory basis of this phenotypic variation and its role in pathogenesis and environmental persistence of B. pseudomallei.


Asunto(s)
Bacteriófagos/genética , Burkholderia pseudomallei/genética , Melioidosis/microbiología , Bacteriófagos/aislamiento & purificación , Bacteriófagos/ultraestructura , Burkholderia pseudomallei/clasificación , Burkholderia pseudomallei/virología , Clonación Molecular , Biología Computacional , ADN Bacteriano/química , ADN Bacteriano/aislamiento & purificación , ADN Viral/análisis , ADN Viral/química , ADN Viral/aislamiento & purificación , Duplicación de Gen/genética , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Genes Reguladores , Humanos , Microscopía Electrónica , Familia de Multigenes , Myoviridae/genética , Myoviridae/aislamiento & purificación , Myoviridae/ultraestructura , Fenotipo , ARN Bacteriano/análisis , ARN Bacteriano/química , ARN Bacteriano/aislamiento & purificación , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN
11.
Immun Inflamm Dis ; 7(1): 7-21, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30569531

RESUMEN

INTRODUCTION: Burkholderia mallei (B. mallei) and Burkholderia pseudomallei (B. pseudomallei), causative agents of glanders and melioidosis, respectively, are invasive intracellular pathogens that actively multiply in phagocytic and non-phagocytic cells. Activation of cell-autonomous autophagy mechanism eliminate intracellular pathogens in which p62 a cytosolic cargo protein is selectively degraded, and an accumulation of this marker occurs if autophagy is deficient. Recurrent, relapsed and reinfection of B. pseudomallei in melioidosis patients in endemic area indicative of lack of complete of clearance and persistence of the pathogen. Reasoning that abundance in the levels of p62 may provide an indication of the intracellular infection, we sought to examine whether increase in intracellular p62 and bacterial burden with Burkholderia infection are linked to autophagy deficiency. METHODS: In this study, we investigated cell culture and mouse models of disease to identify an association between autophagy biomarkers (p62/NBR1) accumulation and intracellular persistence of B. mallei and B. pseudomallei. RESULTS: We demonstrate, that elevated levels of intracellular p62/NBR1 correlated with bacterial persistence, while pre-treatment with a pharmacological inducer of autophagy, rapamycin, reduced both intracellular p62, and bacterial survival. Our results showed an elevated p62 levels (2-5 fold) in spleen and liver cells of Burkholderia-infected BALB/c mice, as well as in spleen cells of Burkholderia-infected C57BL/6 mice, suggesting that an increase in p62/NBR1 was due to an autophagy deficiency. Similar to p62, cytosolic LC3-I levels were also elevated, while the characteristic conversion to the autophagosome-associated membrane bound form LC3-II was low in spleens of the infected mice further supporting the conclusion that autophagy was deficient. CONCLUSION: Taken together, our results suggest that an increase in intracellular p62/NBR1 may be a potential host cell biomarker of B. mallei or B. pseudomallei infections, and identifying autophagy manipulation may potentially aid to therapeutic approach for complete clearance of the pathogen.


Asunto(s)
Autofagia/genética , Burkholderia mallei/fisiología , Burkholderia pseudomallei/fisiología , Desensibilización Inmunológica/métodos , Muermo/metabolismo , Melioidosis/metabolismo , Animales , Quimasas/metabolismo , Modelos Animales de Enfermedad , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas/genética , Proteínas/metabolismo , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/metabolismo
12.
PLoS One ; 13(11): e0208277, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30500862

RESUMEN

Mouse models have been essential to generate supporting data for the research of infectious diseases. Burkholderia pseudomallei, the etiological agent of melioidosis, has been studied using mouse models to investigate pathogenesis and efficacy of novel medical countermeasures to include both vaccines and therapeutics. Previous characterization of mouse models of melioidosis have demonstrated that BALB/c mice present with an acute infection, whereas C57BL/6 mice have shown a tendency to be more resistant to infection and may model chronic disease. In this study, either BALB/c or C57BL/6 mice were exposed to aerosolized human clinical isolates of B. pseudomallei. The bacterial strains included HBPUB10134a (virulent isolate from Thailand), MSHR5855 (virulent isolate from Australia), and 1106a (relatively attenuated isolate from Thailand). The LD50 values were calculated and serial sample collections were performed in order to examine the bacterial burdens in tissues, histopathological features of disease, and the immune response mounted by the mice after exposure to aerosolized B. pseudomallei. These data will be important when utilizing these models for testing novel medical countermeasures. Additionally, by comparing highly virulent strains with attenuated isolates, we hope to better understand the complex disease pathogenesis associated with this bacterium.


Asunto(s)
Burkholderia pseudomallei/fisiología , Melioidosis/patología , Animales , Formación de Anticuerpos , Australia/epidemiología , Bronquios/inmunología , Bronquios/microbiología , Bronquios/patología , Burkholderia pseudomallei/patogenicidad , Citocinas/sangre , Citocinas/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Melioidosis/sangre , Melioidosis/epidemiología , Melioidosis/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Tailandia/epidemiología , Virulencia
13.
PLoS One ; 12(2): e0172627, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28235018

RESUMEN

Burkholderia pseudomallei, the etiologic agent of melioidosis, is a Gram negative bacterium designated as a Tier 1 threat. This bacterium is known to be endemic in Southeast Asia and Northern Australia and can infect humans and animals by several routes. Inhalational melioidosis has been associated with monsoonal rains in endemic areas and is also a significant concern in the biodefense community. There are currently no effective vaccines for B. pseudomallei and antibiotic treatment can be hampered by non-specific symptomology and also the high rate of naturally occurring antibiotic resistant strains. Well-characterized animal models will be essential when selecting novel medical countermeasures for evaluation prior to human clinical trials. Here, we further characterize differences between the responses of BALB/c and C57BL/6 mice when challenged with low doses of a low-passage and well-defined stock of B. pseudomallei K96243 via either intraperitoneal or aerosol routes of exposure. Before challenge, mice were implanted with a transponder to collect body temperature readings, and daily body weights were also recorded. Mice were euthanized on select days for pathological analyses and determination of the bacterial burden in selected tissues (blood, lungs, liver, and spleen). Additionally, spleen homogenate and sera samples were analyzed to better characterize the host immune response after infection with aerosolized bacteria. These clinical, pathological, and immunological data highlighted and confirmed important similarities and differences between these murine models and exposure routes.


Asunto(s)
Burkholderia pseudomallei/inmunología , Inmunidad Innata , Hígado/inmunología , Pulmón/inmunología , Melioidosis/inmunología , Bazo/inmunología , Administración por Inhalación , Animales , Carga Bacteriana , Temperatura Corporal , Peso Corporal , Burkholderia pseudomallei/crecimiento & desarrollo , Burkholderia pseudomallei/patogenicidad , Recuento de Colonia Microbiana , Citocinas/biosíntesis , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Granulocitos/inmunología , Granulocitos/microbiología , Humanos , Inyecciones Intraperitoneales , Hígado/microbiología , Pulmón/microbiología , Subgrupos Linfocitarios/clasificación , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/microbiología , Melioidosis/microbiología , Melioidosis/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/microbiología , Especificidad de la Especie , Bazo/microbiología
14.
J Clin Microbiol ; 55(4): 1205-1210, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28179407

RESUMEN

Melioidosis is a neglected tropical disease that is caused by the bacterium Burkholderia pseudomallei and is underreported in many countries where the disease is endemic. A long and costly administration of antibiotics is needed to clear infections, and there is an unmet need for biomarkers to guide antibiotic treatment and increase the number of patients that complete therapy. We identified calprotectin as a lead biomarker of B. pseudomallei infections and examined correlations between this serum protein and the antibiotic treatment outcomes of patients with melioidosis. Serum levels of calprotectin and C-reactive protein were significantly higher in patients with melioidosis and nonmelioidosis sepsis than in healthy controls. Median calprotectin levels were higher in patients with melioidosis than in those with nonmelioidosis sepsis, whereas C-reactive protein levels were similar in both groups. Notably, intensive intravenous antibiotic treatment of patients with melioidosis resulted in lower levels of calprotectin and C-reactive protein (P < 0.0001), coinciding with recovery. The median percent reduction of calprotectin and C-reactive protein was 71% for both biomarkers after antibacterial therapy. In contrast, we found no significant differences in calreticulin levels between the two melioidosis treatment phases. Thus, reductions in serum calprotectin levels were linked to therapeutic responses to antibiotics. Our results suggest that calprotectin may be a sensitive indicator of melioidosis disease activity and illustrate the potential utility of this biomarker in guiding the duration of antibiotic therapy.


Asunto(s)
Biomarcadores/sangre , Complejo de Antígeno L1 de Leucocito/sangre , Melioidosis/diagnóstico , Administración Intravenosa , Adolescente , Adulto , Anciano , Antibacterianos/administración & dosificación , Proteína C-Reactiva/análisis , Estudios de Cohortes , Progresión de la Enfermedad , Monitoreo de Drogas/métodos , Femenino , Humanos , Masculino , Melioidosis/tratamiento farmacológico , Persona de Mediana Edad , Embarazo , Suero/química , Adulto Joven
15.
Artículo en Inglés | MEDLINE | ID: mdl-26955620

RESUMEN

Burkholderia mallei (Bm) is a highly infectious intracellular pathogen classified as a category B biological agent by the Centers for Disease Control and Prevention. After respiratory exposure, Bm establishes itself within host macrophages before spreading into major organ systems, which can lead to chronic infection, sepsis, and death. Previously, we combined computational prediction of host-pathogen interactions with yeast two-hybrid experiments and identified novel virulence factor genes in Bm, including BMAA0553, BMAA0728 (tssN), and BMAA1865. In the present study, we used recombinant allelic exchange to construct deletion mutants of BMAA0553 and tssN (ΔBMAA0553 and ΔTssN, respectively) and showed that both deletions completely abrogated virulence at doses of >100 times the LD50 of the wild-type Bm strain. Analysis of ΔBMAA0553- and ΔTssN-infected mice showed starkly reduced bacterial dissemination relative to wild-type Bm, and subsequent in vitro experiments characterized pathogenic phenotypes with respect to intracellular growth, macrophage uptake and phagosomal escape, actin-based motility, and multinucleated giant cell formation. Based on observed in vitro and in vivo phenotypes, we explored the use of ΔTssN as a candidate live-attenuated vaccine. Mice immunized with aerosolized ΔTssN showed a 21-day survival rate of 67% after a high-dose aerosol challenge with the wild-type Bm ATCC 23344 strain, compared to a 0% survival rate for unvaccinated mice. However, analysis of histopathology and bacterial burden showed that while the surviving vaccinated mice were protected from acute infection, Bm was still able to establish a chronic infection. Vaccinated mice showed a modest IgG response, suggesting a limited potential of ΔTssN as a vaccine candidate, but also showed prolonged elevation of pro-inflammatory cytokines, underscoring the role of cellular and innate immunity in mitigating acute infection in inhalational glanders.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Vacunas Bacterianas/inmunología , Burkholderia mallei/inmunología , Burkholderia mallei/patogenicidad , Muermo/inmunología , Inmunoglobulina G/inmunología , Administración por Inhalación , Aerosoles , Animales , Burkholderia mallei/genética , Citocinas/metabolismo , Femenino , Eliminación de Gen , Muermo/microbiología , Interacciones Huésped-Patógeno , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Vacunación , Vacunas Atenuadas/inmunología , Virulencia/genética
16.
PLoS One ; 10(9): e0137578, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26356298

RESUMEN

Burkholderia pseudomallei and Burkholderia mallei, classified as category B priority pathogens, are significant human and animal pathogens that are highly infectious and broad-spectrum antibiotic resistant. Currently, the pathogenicity mechanisms utilized by Burkholderia are not fully understood, and correct diagnosis of B. pseudomallei and B. mallei infection remains a challenge due to limited detection methods. Here, we provide a comprehensive analysis of a set of 13 novel Burkholderia collagen-like proteins (Bucl) that were identified among B. pseudomallei and B. mallei select agents. We infer that several Bucl proteins participate in pathogenesis based on their noncollagenous domains that are associated with the components of a type III secretion apparatus and membrane transport systems. Homology modeling of the outer membrane efflux domain of Bucl8 points to a role in multi-drug resistance. We determined that bucl genes are widespread in B. pseudomallei and B. mallei; Fischer's exact test and Cramer's V2 values indicate that the majority of bucl genes are highly associated with these pathogenic species versus nonpathogenic B. thailandensis. We designed a bucl-based quantitative PCR assay which was able to detect B. pseudomallei infection in a mouse with a detection limit of 50 CFU. Finally, chromosomal mapping and phylogenetic analysis of bucl loci revealed considerable genomic plasticity and adaptation of Burkholderia spp. to host and environmental niches. In this study, we identified a large set of phylogenetically unrelated bucl genes commonly found in Burkholderia select agents, encoding predicted pathogenicity factors, detection targets, and vaccine candidates.


Asunto(s)
Adaptación Biológica , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Infecciones por Burkholderia/microbiología , Burkholderia/fisiología , Evolución Molecular , Genoma Bacteriano , Animales , Proteínas Bacterianas/química , Burkholderia/clasificación , Burkholderia/patogenicidad , Biología Computacional , Eliminación de Gen , Reordenamiento Génico , Genes Bacterianos , Humanos , Ratones , Modelos Moleculares , Filogenia , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Selección Genética , Termodinámica
17.
Microb Pathog ; 86: 53-63, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26162294

RESUMEN

Burkholderia pseudomallei is the etiologic agent of melioidosis, which is endemic in Southeast Asia and Northern Australia. We previously found by the intraperitoneal (IP) route that we could discern differences in virulence in mice amongst different strains of B. pseudomallei. We report an early immune response study comparing two strains in our collection which represent the least, B. pseudomallei 1106a, and one of the most, HBPUB10134a, virulent strains in BALB/c mice. B. pseudomallei HBPUB10134a infected mouse spleens contained a 2-3 log higher bacterial burden than mice infected with B. pseudomallei 1106a 3 days post-infection (PI). More and higher amounts of inflammatory cytokines/chemokines were detected in sera and spleen extracts from B. pseudomallei HBPUB10134a than B. pseudomallei 1106a infected mice. The most prominent were IFNγ, IL-1α, IL-1ß, IL-6, IL-10, IP-10, and MIG. After 7 days PI, there was a decrease in bacterial burden in spleens from 1106a infected mice and a decrease in cytokines/chemokines in sera and spleen extracts from both sets of mice. By day 14 PI we saw an increase in monocytes/macrophages, NK cells, and granulocytes in spleens from both sets of mice. No B. pseudomallei HBPUB10134a infected mice survived after this time. In summary, B. pseudomallei HBPUB10134a was more virulent and induced host innate immune responses typical of a more acute-type infection than did B. pseudomallei 1106a which produced a more chronic infection in mice.


Asunto(s)
Burkholderia pseudomallei/inmunología , Melioidosis/inmunología , Melioidosis/patología , Animales , Asia Sudoriental , Australia , Carga Bacteriana , Burkholderia pseudomallei/crecimiento & desarrollo , Enfermedad Crónica , Citocinas/análisis , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Humanos , Leucocitos Mononucleares/inmunología , Ratones Endogámicos BALB C , Suero/química , Bazo/microbiología , Bazo/patología , Virulencia , Adulto Joven
18.
Bull Tokyo Dent Coll ; 56(2): 113-20, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26084999

RESUMEN

We report bone augmentation for alveolar bone loss at the bottom of the nasal cavity in conjunction with simultaneous implant placement in the same operative field in the esthetic zone. The patient was a 33-year-old man who was referred to us requesting implant treatment after undergoing tooth extraction (#12) due to root fracture. An examination was performed using cone beam computed tomography (CT) and simulation software. The results indicated insufficient volume of labial bone for the requested procedure, especially at the planned site of the implant neck. Therefore, bone augmentation was performed at the apical site of the implant socket (alveolar bone at the bottom of the nasal cavity). Because the surgical line of the harvest site formed a trapezoidal shape, the procedure was named the "Trapezial Design Technique". Assessment of complications (Barone & Covane classification), success (Albrektsson classification), and observation of labial bone using cone beam CT were performed postoperatively. No com-plications were observed at 27 months after prosthetic treatment. The implant and the tissue surrounding it were in a stable condition. This indicates that this procedure is effective in placing an implant with simultaneous bone augmentation in the esthetic zone.


Asunto(s)
Pérdida de Hueso Alveolar/etiología , Aumento de la Cresta Alveolar , Implantación Dental Endoósea , Adulto , Trasplante Óseo , Estética , Humanos , Masculino
19.
PLoS One ; 10(4): e0124667, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25909629

RESUMEN

Burkholderia pseudomallei, the etiologic agent of melioidosis, is a gram-negative facultative intracellular bacterium. This bacterium is endemic in Southeast Asia and Northern Australia and can infect humans and animals by several routes. It has also been estimated to present a considerable risk as a potential biothreat agent. There are currently no effective vaccines for B. pseudomallei, and antibiotic treatment can be hampered by nonspecific symptomology, the high incidence of naturally occurring antibiotic resistant strains, and disease chronicity. Accordingly, there is a concerted effort to better characterize B. pseudomallei and its associated disease. Before novel vaccines and therapeutics can be tested in vivo, a well characterized animal model is essential. Previous work has indicated that mice may be a useful animal model. In order to develop standardized animal models of melioidosis, different strains of bacteria must be isolated, propagated, and characterized. Using a murine intraperitoneal (IP) infection model, we tested the virulence of 11 B. pseudomallei strains. The IP route offers a reproducible way to rank virulence that can be readily reproduced by other laboratories. This infection route is also useful in distinguishing significant differences in strain virulence that may be masked by the exquisite susceptibility associated with other routes of infection (e.g., inhalational). Additionally, there were several pathologic lesions observed in mice following IP infection. These included varisized abscesses in the spleen, liver, and haired skin. This model indicated that commonly used laboratory strains of B. pseudomallei (i.e., K96243 and 1026b) were significantly less virulent as compared to more recently acquired clinical isolates. Additionally, we characterized in vitro strain-associated differences in virulence for macrophages and described a potential inverse relationship between virulence in the IP mouse model of some strains and in the macrophage phagocytosis assay. Strains which were more virulent for mice (e.g., HBPU10304a) were often less virulent in the macrophage assays, as determined by several parameters such as intracellular bacterial replication and host cell cytotoxicity.


Asunto(s)
Burkholderia pseudomallei/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Melioidosis/inmunología , Melioidosis/microbiología , Absceso/inmunología , Absceso/microbiología , Absceso/patología , Animales , Burkholderia pseudomallei/patogenicidad , Modelos Animales de Enfermedad , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Melioidosis/metabolismo , Melioidosis/patología , Ratones , Fenotipo
20.
Microb Pathog ; 78: 20-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25450887

RESUMEN

Burkholderia pseudomallei infection causes melioidosis and is often characterized by severe sepsis. Although rare in humans, Burkholderia mallei has caused infections in laboratory workers, and the early innate cellular response to B. mallei in human and nonhuman primates has not been characterized. In this study, we examined the primary cellular immune response to B. mallei in PBMC cultures of non-human primates (NHPs), Chlorocebus aethiops (African Green Monkeys), Macaca fascicularis (Cynomolgus macaque), and Macaca mulatta (Rhesus macaque) and humans. Our results demonstrated that B. mallei elicited strong primary pro-inflammatory cytokines (IFN-γ, TNF-α, IL-1ß, and IL-6) equivalent to the levels of B. pseudomallei in primary PBMC cultures of NHPs and humans. When we examined IL-1ß and other cytokine responses by comparison to Escherichia coli LPS, African Green Monkeys appears to be most responsive to B. mallei than Cynomolgus or Rhesus. Characterization of the immune signaling mechanism for cellular response was conducted by using a ligand induced cell-based reporter assay, and our results demonstrated that MyD88 mediated signaling contributed to the B. mallei and B. pseudomallei induced pro-inflammatory responses. Notably, the induced reporter activity with B. mallei, B. pseudomallei, or purified LPS from these pathogens was inhibited and cytokine production was attenuated by a MyD88 inhibitor. Together, these results show that in the scenario of severe hyper-inflammatory responses to B. mallei infection, MyD88 targeted therapeutic intervention may be a successful strategy for therapy.


Asunto(s)
Burkholderia mallei/inmunología , Muermo/inmunología , Inmunidad Innata , Leucocitos Mononucleares/inmunología , Animales , Burkholderia mallei/fisiología , Chlorocebus aethiops , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Muermo/genética , Muermo/microbiología , Humanos , Inmunidad Celular , Leucocitos Mononucleares/microbiología , Macaca fascicularis , Macaca mulatta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...