Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Dis Markers ; 2021: 6166492, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34691288

RESUMEN

Melioidosis is a serious infectious disease caused by the environmental Gram-negative bacillus Burkholderia pseudomallei. It has been shown that the host immune system, mainly comprising various types of immune cells, fights against the disease. The present study was to specify correlation between septicemic melioidosis and the levels of multiple immune cells. First, the genes with differential expression patterns between patients with septicemic melioidosis (B. pseudomallei) and health donors (control/healthy) were identified. These genes being related to cytokine binding, cell adhesion molecule binding, and MHC relevant proteins may influence immune response. The Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed 23 enriched immune response pathways. We further leveraged the microarray data to investigate the relationship between immune response and septicemic melioidosis, using the CIBERSORT analysis. Comparison of the percentages of 22 immune cell types in B. pseudomallei vs. control/healthy revealed that those of CD4 memory resting cells, CD8+ T cells, B memory cells, and CD4 memory activated cells were low, whereas those of M0 macrophages, neutrophils, and gamma delta T cells were high. The multivariate logistic regression analysis further revealed that CD8+ T cells, M0 macrophages, neutrophils, and naive CD4+ cells were strongly associated with the onset of septicemic melioidosis, and M2 macrophages and neutrophils were associated with the survival in septicemic melioidosis. Taken together, these data point to a complex role of immune cells on the development and progression of melioidosis.


Asunto(s)
Bacteriemia/inmunología , Bacteriemia/mortalidad , Proteínas Sanguíneas/genética , Melioidosis/inmunología , Melioidosis/mortalidad , Bacteriemia/sangre , Bacteriemia/genética , Sangre/inmunología , Fenómenos Fisiológicos Sanguíneos , Proteínas Sanguíneas/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/fisiología , Estudios de Casos y Controles , Perfilación de la Expresión Génica , Ontología de Genes , Humanos , Macrófagos/inmunología , Macrófagos/fisiología , Melioidosis/sangre , Melioidosis/genética
2.
Emerg Microbes Infect ; 10(1): 8-18, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33256556

RESUMEN

Melioidosis is an often lethal tropical disease caused by the Gram-negative bacillus, Burkholderia pseudomallei. The study objective was to characterize transcriptomes in melioidosis patients and identify genes associated with outcome. Whole blood RNA-seq was performed in a discovery set of 29 melioidosis patients and 3 healthy controls. Transcriptomic profiles of patients who did not survive to 28 days were compared with patients who survived and healthy controls, showing 65 genes were significantly up-regulated and 218 were down-regulated in non-survivors compared to survivors. Up-regulated genes were involved in myeloid leukocyte activation, Toll-like receptor cascades and reactive oxygen species metabolic processes. Down-regulated genes were hematopoietic cell lineage, adaptive immune system and lymphocyte activation pathways. RT-qPCR was performed for 28 genes in a validation set of 60 melioidosis patients and 20 healthy controls, confirming differential expression. IL1R2, GAS7, S100A9, IRAK3, and NFKBIA were significantly higher in non-survivors compared with survivors (P < 0.005) and healthy controls (P < 0.0001). The AUROCC of these genes for mortality discrimination ranged from 0.80-0.88. In survivors, expression of IL1R2, S100A9 and IRAK3 genes decreased significantly over 28 days (P < 0.05). These findings augment our understanding of this severe infection, showing expression levels of specific genes are potential biomarkers to predict melioidosis outcomes.


Asunto(s)
Biomarcadores/sangre , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Melioidosis/mortalidad , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Melioidosis/sangre , Melioidosis/genética , Persona de Mediana Edad , Estudios Prospectivos , Análisis de Secuencia de ARN , Análisis de Supervivencia
3.
PLoS Negl Trop Dis ; 14(9): e0008659, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32986699

RESUMEN

BACKGROUND: Melioidosis therapy is divided into an intravenous intensive phase and an oral eradication phase. The Darwin melioidosis treatment guideline has evolved over two decades, with over 1150 consecutive patients with culture-confirmed melioidosis managed under the Darwin Prospective Melioidosis Study. The current guideline, published in 2015, has been associated with low rates of recrudescence, relapse and mortality, and together with the treatment trials in Thailand, forms the basis for consensus global guidelines. We aimed to reassess the Darwin guideline and determine if any adjustments to the recommendations better reflect current practice in melioidosis therapy at Royal Darwin Hospital. METHODOLOGY/PRINCIPAL FINDINGS: This retrospective cohort study reviews the characteristics, admission duration, duration of intravenous antibiotics, recrudescence, recurrence and mortality in all patients presenting with first episode culture-confirmed melioidosis in the tropical north of Australia's Northern Territory from 1st October 2012 until 1st January 2017. 234 patients were available for analysis. 16 (6.8%) died during the intensive phase treatment and 6 (2.6%) did not have complete treatment at Royal Darwin Hospital, leaving 212 patients for analysis. Six (2.8%) patients had recrudescence during therapy and 10 (4.7%) had recurrent melioidosis (relapse or new infection) after completion of therapy. Persisting osteomyelitis requiring surgery was an important reason for recrudescence as was unrecognized osteomyelitis for relapse. For patients presenting with an antibiotic duration determining focus of pneumonia, durations of intravenous antibiotics were often prolonged beyond the current 2-week minimum treatment recommendation. Prolongation of therapy in pneumonia mostly occurred in patients presenting with multi-lobar disease or with concurrent blood culture positivity. CONCLUSIONS/SIGNIFICANCE: The 2015 Darwin melioidosis guideline is working well with low rates of recrudescence, relapse and mortality. Based on the practice of the treating clinicians, the 2020 revision of the guideline has been adjusted to include a duration of a minimum of 3 weeks of intravenous antibiotics for those with concurrent bacteraemia and pneumonia involving only a single lobe and those with bilateral and unilateral multi-lobar pneumonias who do not have bacteraemia. We also extend to a minimum of 4 weeks intravenous therapy for those with concurrent bacteraemia and bilateral or unilateral multi-lobar pneumonia.


Asunto(s)
Antibacterianos/uso terapéutico , Melioidosis/tratamiento farmacológico , Melioidosis/genética , Selección Genética , Administración Intravenosa/métodos , Adulto , Bacteriemia/tratamiento farmacológico , Burkholderia pseudomallei , Femenino , Guías como Asunto , Humanos , Masculino , Melioidosis/mortalidad , Persona de Mediana Edad , Northern Territory , Osteomielitis/cirugía , Estudios Prospectivos , Recurrencia , Estudios Retrospectivos , Tailandia
4.
PLoS Negl Trop Dis ; 13(5): e0007354, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31067234

RESUMEN

BACKGROUND: Toll-like receptors (TLRs) are sentinel receptors of the innate immune system. TLR4 detects bacterial lipopolysaccharide (LPS) and TLR5 detects bacterial flagellin. A common human nonsense polymorphism, TLR5:c.1174C>T, results in a non-functional TLR5 protein. Individuals carrying this variant have decreased mortality from melioidosis, infection caused by the flagellated Gram-negative bacterium Burkholderia pseudomallei. Although impaired flagellin-dependent signaling in carriers of TLR5:c.1174C>T is well established, this study tested the hypothesis that a functional effect of TLR5:c.1174C>T is flagellin-independent and involves LPS-TLR4 pathways. METHODOLOGY/PRINCIPAL FINDINGS: Whole blood from two independent cohorts of individuals genotyped at TLR5:c.1174C>T was stimulated with wild type or aflagellated B. pseudomallei or purified bacterial motifs followed by plasma cytokine measurements. Blood from individuals carrying the TLR5:c.1174C>T variant produced less IL-6 and IL-10 in response to an aflagellated B. pseudomallei mutant and less IL-8 in response to purified B. pseudomallei LPS than blood from individuals without the variant. TLR5 expression in THP1 cells was silenced using siRNA; these cells were stimulated with LPS before cytokine levels in cell supernatants were quantified by ELISA. In these cells following LPS stimulation, silencing of TLR5 with siRNA reduced both TNF-α and IL-8 levels. These effects were not explained by differences in TLR4 mRNA expression or NF-κB or IRF activation. CONCLUSIONS/SIGNIFICANCE: The effects of the common nonsense TLR5:c.1174C>T polymorphism on the host inflammatory response to B. pseudomallei may not be restricted to flagellin-driven pathways. Moreover, TLR5 may modulate TLR4-dependent cytokine production. While these results may have broader implications for the role of TLR5 in the innate immune response in melioidosis and other conditions, further studies of the mechanisms underlying these observations are required.


Asunto(s)
Burkholderia pseudomallei/inmunología , Flagelina/inmunología , Melioidosis/genética , Melioidosis/inmunología , Polimorfismo Genético , Receptor Toll-Like 5/genética , Adolescente , Adulto , Anciano , Burkholderia pseudomallei/genética , Codón sin Sentido , Estudios de Cohortes , Femenino , Flagelina/genética , Humanos , Inmunidad Innata , Interleucina-10/genética , Interleucina-10/inmunología , Masculino , Melioidosis/microbiología , Persona de Mediana Edad , FN-kappa B/genética , FN-kappa B/inmunología , Mutación Puntual , Receptor Toll-Like 5/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Adulto Joven
5.
Infect Immun ; 86(10)2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30037795

RESUMEN

Burkholderia pseudomallei causes the severe disease melioidosis. The bacterium subverts the host immune system and replicates inside cells, and host mortality results primarily from sepsis-related complications. Lipopolysaccharide (LPS) is a major virulence factor and mediator of sepsis that many pathogens capable of intracellular growth modify to reduce their immunological "footprint." The binding strength of B. pseudomallei LPS for human LPS binding protein (hLBP) was measured using surface plasmon resonance. The structures of lipid A isolated from B. pseudomallei under different temperatures were analyzed by matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS), and the gene expression of two lipid A remodeling genes, lpxO and pagL, was investigated. The LPS was characterized for its ability to trigger tumor necrosis factor alpha (TNF-α) release and to activate caspase-11-triggered pyroptosis by introduction of LPS into the cytosol. Lipid A from long-term chronic-infection isolates was isolated and characterized by MALDI-TOF MS and also by the ability to trigger caspase-11-mediated cell death. Lipid A from B. pseudomallei 1026b lpxO and pagL mutants were characterized by positive- and negative-mode MALDI-TOF MS to ultimately identify their role in lipid A structural modifications. Replication of lpxO and pagL mutants and their complements within macrophages showed that lipid A remodeling can effect growth in host cells and activation of caspase-11-mediated cytotoxicity.


Asunto(s)
Burkholderia pseudomallei/metabolismo , Burkholderia pseudomallei/patogenicidad , Lípido A/metabolismo , Lipopolisacáridos/metabolismo , Melioidosis/microbiología , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo , Animales , Apoptosis , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/crecimiento & desarrollo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Humanos , Lípido A/química , Melioidosis/genética , Melioidosis/metabolismo , Melioidosis/fisiopatología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Viabilidad Microbiana , Unión Proteica , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
6.
PLoS One ; 12(7): e0180203, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28686630

RESUMEN

Caspase-6 is a member of the executioner caspases and known to play a role in innate and adaptive immune processes. However, its role in infectious diseases has rarely been addressed yet. We here examined the impact of caspase-6 in an in vivo infection model using the Gram-negative rod Burkholderia pseudomallei, causing the infectious disease melioidosis that is endemic in tropical and subtropical areas around the world. Caspase-6-/- and C57BL/6 wild type mice were challenged with B. pseudomallei for comparing mortality, bacterial burden and inflammatory cytokine expression. Bone-marrow derived macrophages were used to analyse the bactericidal activity in absence of caspase-6. Caspase-6 deficiency was associated with higher mortality and bacterial burden in vivo after B. pseudomallei infection. The bactericidal activity of caspase-6-/- macrophages was impaired compared to wild type cells. Caspase-6-/- mice showed higher expression of the IL-1ß gene, known to be detrimental in murine melioidosis. Expression of the IL-10 gene was also increased in caspase-6-/- mice as early as 6 hours after infection. Treatment with exogenous IL-10 rendered mice more susceptible against B. pseudomallei challenge. Thus, caspase-6 seems to play a crucial role for determining resistance against the causative agent of melioidosis. To our knowledge this is the first report showing that caspase-6 is crucial for mediating resistance in an in vivo infection model. Caspase-6 influences the expression of detrimental cytokines and therefore seems to be important for achieving a well-balanced immune response that contributes for an efficient elimination of the pathogen.


Asunto(s)
Burkholderia pseudomallei/genética , Caspasa 6/genética , Interleucina-10/administración & dosificación , Interleucina-1beta/biosíntesis , Melioidosis/genética , Animales , Burkholderia pseudomallei/patogenicidad , Farmacorresistencia Microbiana/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-10/biosíntesis , Interleucina-1beta/genética , Macrófagos/metabolismo , Macrófagos/patología , Melioidosis/microbiología , Melioidosis/patología , Ratones , Ratones Noqueados
7.
Infect Immun ; 84(7): 1941-1956, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27091931

RESUMEN

Melioidosis, caused by the bacterium Burkholderia pseudomallei, is an often severe infection that regularly involves respiratory disease following inhalation exposure. Intranasal (i.n.) inoculation of mice represents an experimental approach used to study the contributions of bacterial capsular polysaccharide I (CPS I) to virulence during acute disease. We used aerosol delivery of B. pseudomallei to establish respiratory infection in mice and studied CPS I in the context of innate immune responses. CPS I improved B. pseudomallei survival in vivo and triggered multiple cytokine responses, neutrophil infiltration, and acute inflammatory histopathology in the spleen, liver, nasal-associated lymphoid tissue, and olfactory mucosa (OM). To further explore the role of the OM response to B. pseudomallei infection, we infected human olfactory ensheathing cells (OECs) in vitro and measured bacterial invasion and the cytokine responses induced following infection. Human OECs killed >90% of the B. pseudomallei in a CPS I-independent manner and exhibited an antibacterial cytokine response comprising granulocyte colony-stimulating factor, tumor necrosis factor alpha, and several regulatory cytokines. In-depth genome-wide transcriptomic profiling of the OEC response by RNA-Seq revealed a network of signaling pathways activated in OECs following infection involving a novel group of 378 genes that encode biological pathways controlling cellular movement, inflammation, immunological disease, and molecular transport. This represents the first antimicrobial program to be described in human OECs and establishes the extensive transcriptional defense network accessible in these cells. Collectively, these findings show a role for CPS I in B. pseudomallei survival in vivo following inhalation infection and the antibacterial signaling network that exists in human OM and OECs.


Asunto(s)
Cápsulas Bacterianas/inmunología , Burkholderia pseudomallei/inmunología , Interacciones Huésped-Patógeno/inmunología , Melioidosis/inmunología , Melioidosis/microbiología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/microbiología , Animales , Cápsulas Bacterianas/genética , Carga Bacteriana , Burkholderia pseudomallei/genética , Células Cultivadas , Biología Computacional/métodos , Citocinas/metabolismo , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunidad Innata , Melioidosis/genética , Melioidosis/metabolismo , Ratones , Mutación , Infiltración Neutrófila , Neuronas Receptoras Olfatorias/inmunología , Neuronas Receptoras Olfatorias/metabolismo , Neuronas Receptoras Olfatorias/microbiología , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/metabolismo , Transducción de Señal , Virulencia , Factores de Virulencia
8.
Artículo en Ruso | MEDLINE | ID: mdl-30695386

RESUMEN

AIM: Determine an optimal set of the most effective methods of identification and intraspecies typing ofcausative agents ofglanders and melioidosis. Materials andmethods. Bacteriologic, immunochemical, molecular-genetic methods were used. RESULTS: A possibility to identify collection strains of pathogenic and closely related Burkholderia in semiautomatic systems is studied. Means of detection of informative variable genome segments ofthe specified microorganisms were developed, methods of their genetic typing were selected. Effectiveness of application of precipitating mAbs for differentiation of Burkholderia was established. Data on diagnostic possibilities of immunoglobulins fluorescing based on monoclonal antibodies of various etiotropic directionality for detection and identification of B. mallei and B. pseudomallei are generalized. Experimental series of amplification test-systems for identification of glanders and melioidosis causative agents in real-time PCR format are created. CONCLUSION: A number of methods for identification and typing of glanders and melioidosis causative agents is proposed.


Asunto(s)
Burkholderia mallei/genética , Burkholderia pseudomallei/genética , Muermo , Melioidosis , Reacción en Cadena en Tiempo Real de la Polimerasa , Animales , Muermo/diagnóstico , Muermo/genética , Humanos , Melioidosis/diagnóstico , Melioidosis/genética
9.
Immunol Cell Biol ; 93(1): 18-24, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25331551

RESUMEN

Autophagy has become increasingly viewed as an important component of the eukaryotic innate immune system. The elimination of intracellular pathogens by autophagy in mammalian cells (xenophagy) results not only in the degradation of invading bacteria, viruses, fungi and parasites, but also liberation of metabolites that may have been utilized during pathogen infection, thus promoting cell survival. After gaining entry into the cell, intracellular bacterial pathogens attempt to escape from phagosomes (or endosomes) into the cytosol where they endeavour to continue the infection cycle unhindered by host cell protective mechanisms. Bacterial recognition resulting from either their cytosolic location, the secretion of bacterial products, or phagosomal membrane damage, can induce autophagy. In this context, induction of autophagy results in the clearance of some bacterial pathogens, whereas other bacteria are able to manipulate autophagy for their own benefit and appear to effectively replicate within autophagosome-like vesicles. Some bacteria are seemingly able to evade autophagy and Burkholderia pseudomallei is one of them. This review will discuss the autophagic processes that may be activated by host cells to provide protection against infection by this bacterial pathogen.


Asunto(s)
Autofagia/inmunología , Proteínas Bacterianas/inmunología , Burkholderia pseudomallei/inmunología , Macrófagos/inmunología , Melioidosis/inmunología , Fagosomas/inmunología , Autofagia/genética , Proteínas Bacterianas/genética , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Inmunidad Innata , Macrófagos/microbiología , Macrófagos/patología , Melioidosis/genética , Melioidosis/microbiología , Melioidosis/patología , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/inmunología , Fagosomas/química , Fagosomas/genética , Transducción de Señal , Ubiquitina/genética , Ubiquitina/inmunología
10.
J Immunol ; 192(1): 300-7, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24298015

RESUMEN

Nucleotide-binding oligomerization domain 2 (NOD2) is a cytosolic pathogen recognition receptor that regulates susceptibility to a variety of infections and chronic diseases. Burkholderia pseudomallei, a facultative intracellular bacterium, causes the tropical infection melioidosis. We hypothesized that NOD2 may participate in host defense in melioidosis. We performed a series of in vitro assays and in vivo experiments and analyzed the association of human genetic variation with infection to delineate the contribution of NOD2 to the host response to B. pseudomallei. We found that transfection with NOD2 mediated NF-κB activation induced by B. pseudomallei stimulation of HEK293 cells. After low-dose inoculation with aerosolized B. pseudomallei, Nod2-deficient mice showed impaired clinical responses and permitted greater bacterial replication in the lung and dissemination to the spleen compared with wild-type mice. IL-6 and KC levels were higher in the lungs of Nod2-deficient mice. In a cohort of 1562 Thai subjects, a common genetic polymorphism in the NOD2 region, rs7194886, was associated with melioidosis, and this effect was most pronounced in women. rs7194886 was not associated with differences in cytokine production induced by whole-blood stimulation with the NOD2 ligand, muramyl dipeptide, or B. pseudomallei. To our knowledge, these findings are the first to characterize the role of NOD2 in host defense in mammalian melioidosis.


Asunto(s)
Burkholderia pseudomallei/inmunología , Melioidosis/genética , Melioidosis/inmunología , Proteína Adaptadora de Señalización NOD2/genética , Animales , Línea Celular Tumoral , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Inmunidad Innata/genética , Interleucina-6/sangre , Interleucina-6/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Melioidosis/metabolismo , Melioidosis/mortalidad , Ratones , Ratones Noqueados , Proteína Adaptadora de Señalización NOD2/deficiencia , Proteína Adaptadora de Señalización NOD2/metabolismo , Polimorfismo de Nucleótido Simple
11.
Infect Immun ; 81(9): 3463-71, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23836818

RESUMEN

Burkholderia pseudomallei, the causative agent of melioidosis, evades macrophage killing by suppressing the TRIF-dependent pathway, leading to inhibition of inducible nitric oxide synthase (iNOS) expression. We previously demonstrated that virulent wild-type B. pseudomallei inhibits the TRIF-dependent pathway by upregulating sterile-α- and armadillo motif-containing protein (SARM) and by inhibiting downregulation of signal regulatory protein α (SIRPα); both molecules are negative regulators of Toll-like receptor signaling. In contrast, the less virulent lipopolysaccharide (LPS) mutant of B. pseudomallei is unable to exhibit these features and is susceptible to macrophage killing. However, the functional relationship of these two negative regulators in the evasion of macrophage defense has not been elucidated. We demonstrated here that SIRPα downregulation was observed after inhibition of SARM expression by small interfering RNA in wild-type-infected macrophages, indicating that SIRPα downregulation is regulated by SARM. Furthermore, this downregulation requires activation of the TRIF signaling pathway, as we observed abrogation of SIRPα downregulation as well as restricted bacterial growth in LPS mutant-infected TRIF-depleted macrophages. Although inhibition of SARM expression is correlated to SIRPα downregulation and iNOS upregulation in gamma interferon-activated wild-type-infected macrophages, these phenomena appear to bypass the TRIF-dependent pathway. Similar to live bacteria, the wild-type LPS is able to upregulate SARM and to prevent SIRPα downregulation, implying that the LPS of B. pseudomallei may play a crucial role in regulating the expression of these two negative regulators. Altogether, our findings show a previously unrecognized role of B. pseudomallei-induced SARM in inhibiting SIRPα downregulation-mediated iNOS upregulation, facilitating the ability of the bacterium to multiply in macrophages.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas del Dominio Armadillo/genética , Burkholderia pseudomallei/genética , Proteínas del Citoesqueleto/genética , Macrófagos/metabolismo , Melioidosis/genética , Receptores Inmunológicos/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Proteínas del Dominio Armadillo/inmunología , Proteínas del Dominio Armadillo/metabolismo , Burkholderia pseudomallei/inmunología , Burkholderia pseudomallei/metabolismo , Proteínas del Citoesqueleto/inmunología , Proteínas del Citoesqueleto/metabolismo , Regulación hacia Abajo/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Melioidosis/inmunología , Melioidosis/metabolismo , Melioidosis/microbiología , Ratones , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Óxido Nítrico Sintasa de Tipo II/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
12.
Infect Immun ; 81(1): 271-7, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23115045

RESUMEN

LC3-associated phagocytosis (LAP) of Burkholderia pseudomallei by murine macrophage (RAW 264.7) cells is an intracellular innate defense mechanism. Beclin 1, a protein with several roles in autophagic processes, is known to be recruited to phagosomal membranes as a very early event in LAP. We sought to determine whether knockdown of Beclin 1 by small interfering RNA (siRNA) would affect recruitment of LC3 and subsequent LAP of infecting B. pseudomallei. Both starvation and rapamycin treatment can induce Beclin 1-dependent autophagy. Therefore, we analyzed the consequences of Beclin 1 knockdown for LAP in infected cells that had been either starved or treated with rapamycin by determining the levels of bacterial colocalization with LC3 and intracellular survival. Concurrently, we confirmed the location of bacteria as either contained in phagosomes or free in the cytoplasm. We found that both rapamycin and starvation treatment enhanced LAP of B. pseudomallei but that the rapamycin response is Beclin 1 independent whereas the starvation response is Beclin 1 dependent.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Burkholderia pseudomallei/inmunología , Proteínas Asociadas a Microtúbulos/inmunología , Fagocitosis/inmunología , Inanición/fisiopatología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/genética , Autofagia/inmunología , Proteínas Bacterianas/genética , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/metabolismo , Línea Celular , Macrófagos/inmunología , Macrófagos/metabolismo , Melioidosis/genética , Melioidosis/inmunología , Melioidosis/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Fagocitosis/genética , Fagosomas/genética , Fagosomas/inmunología , Fagosomas/metabolismo , Sirolimus/farmacología , Inanición/inmunología
13.
Infect Immun ; 79(7): 2921-7, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21555400

RESUMEN

Burkholderia pseudomallei, a causative agent of melioidosis, is a Gram-negative facultative intracellular bacterium that can survive and multiply in macrophages. Previously, we demonstrated that B. pseudomallei failed to activate gene expression downstream of the MyD88-independent pathway, particularly the expression of beta interferon (IFN-ß) and inducible nitric oxide synthase (iNOS), leading to the inability of macrophages to kill this bacterium. In the present report, we extended our study to show that B. pseudomallei was able to activate sterile-α and Armadillo motif (SARM)-containing protein, a known negative regulator of the MyD88-independent pathway. Both live B. pseudomallei and heat-killed B. pseudomallei were able to upregulate SARM expression in a time-dependent manner in mouse macrophage cell line RAW 264.7. The expression of SARM required bacterial internalization, as it could be inhibited by cytochalasin D. In addition, the intracellular survival of B. pseudomallei was suppressed in SARM-deficient macrophages. Increased expression of IFN-ß and iNOS and degradation of IκBα correlated with enhanced macrophage killing capability. These results demonstrated that B. pseudomallei modulated macrophage defense mechanisms by upregulating SARM, thus leading to the suppression of IFN-ß and iNOS needed for bacterial elimination.


Asunto(s)
Proteínas del Dominio Armadillo/biosíntesis , Proteínas del Dominio Armadillo/genética , Burkholderia pseudomallei/metabolismo , Burkholderia pseudomallei/patogenicidad , Macrófagos/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Animales , Línea Celular , Citocalasina D/farmacología , Proteínas I-kappa B/metabolismo , Immunoblotting , Interferón beta/metabolismo , Macrófagos/microbiología , Melioidosis/genética , Melioidosis/metabolismo , Melioidosis/microbiología , Ratones , Inhibidor NF-kappaB alfa , Óxido Nítrico Sintasa de Tipo II/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
BMC Immunol ; 12: 20, 2011 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-21410970

RESUMEN

BACKGROUND: Burkholderia pseudomallei is the causative agent of melioidosis, an emerging bacterial infectious disease in tropical and subtropical areas. We recently showed that NADPH oxidase but not nitric oxide (NO) contributes to resistance in innately resistant C57BL/6 mice in a B. pseudomallei respiratory infection model. However, the function of NO for resistance was shown to differ among distinct strains of mice and proved also to be stage dependent in various infection models. The present study therefore aimed to examine the role of NO in a systemic infection model of melioidosis and to test whether the function of NO differs among innately resistant C57BL/6 and susceptible BALB/c mice after B. pseudomallei infection. RESULTS: C57BL/6 iNOS-/- mice that were intravenously infected with B. pseudomallei survived several weeks, whereas most of the wild type animals succumbed during this period. The bacterial burden in liver and spleen was significantly higher in wild type animals compared to iNOS-/- mice 13 days after challenge. In contrast, BALB/c mice that were treated with amminoguanidine to inhibit NO expression in vivo showed significantly enhanced mortality rates and higher bacterial loads in liver and spleen compared to control animals. The bactericidal function of IFN-γ stimulated C57BL/6 iNOS-/- macrophages were not altered after B. pseudomallei infection, but BALB/c macrophages exhibited reduced killing activity against the pathogen when NO was inhibited. CONCLUSION: Our present data indicate a dual role of NO among resistant and susceptible mouse strains after B. pseudomallei infection. NO mediated mechanisms are an essential component to control the infection in susceptible BALB/c mice. In contrast, NO production in B. pseudomallei infected C57BL/6 mice rather harmed the host likely due to its detrimental effects.


Asunto(s)
Burkholderia pseudomallei/inmunología , Susceptibilidad a Enfermedades , Melioidosis/inmunología , Óxido Nítrico/metabolismo , Animales , Burkholderia pseudomallei/patogenicidad , Células Cultivadas , Citotoxicidad Inmunológica/efectos de los fármacos , Modelos Animales de Enfermedad , Guanidina/administración & dosificación , Guanidina/análogos & derivados , Guanidina/farmacología , Humanos , Inmunidad Activa , Activación de Macrófagos/efectos de los fármacos , Melioidosis/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/inmunología , Óxido Nítrico Sintasa de Tipo II/genética , Especificidad de la Especie
15.
Infect Immun ; 79(4): 1512-25, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21300775

RESUMEN

The Burkholderia pseudomallei K96243 genome encodes six type VI secretion systems (T6SSs), but little is known about the role of these systems in the biology of B. pseudomallei. In this study, we purified recombinant Hcp proteins from each T6SS and tested them as vaccine candidates in the BALB/c mouse model of melioidosis. Recombinant Hcp2 protected 80% of mice against a lethal challenge with K96243, while recombinant Hcp1, Hcp3, and Hcp6 protected 50% of mice against challenge. Hcp6 was the only Hcp constitutively produced by B. pseudomallei in vitro; however, it was not exported to the extracellular milieu. Hcp1, on the other hand, was produced and exported in vitro when the VirAG two-component regulatory system was overexpressed in trans. We also constructed six hcp deletion mutants (Δhcp1 through Δhcp6) and tested them for virulence in the Syrian hamster model of infection. The 50% lethal doses (LD(50)s) for the Δhcp2 through Δhcp6 mutants were indistinguishable from K96243 (<10 bacteria), but the LD(50) for the Δhcp1 mutant was >10(3) bacteria. The hcp1 deletion mutant also exhibited a growth defect in RAW 264.7 macrophages and was unable to form multinucleated giant cells in this cell line. Unlike K96243, the Δhcp1 mutant was only weakly cytotoxic to RAW 264.7 macrophages 18 h after infection. The results suggest that the cluster 1 T6SS is essential for virulence and plays an important role in the intracellular lifestyle of B. pseudomallei.


Asunto(s)
Perfilación de la Expresión Génica , Melioidosis/microbiología , Factores de Virulencia/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/metabolismo , Cricetinae , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Femenino , Técnica del Anticuerpo Fluorescente , Expresión Génica , Genes Bacterianos , Humanos , Immunoblotting , Hígado/microbiología , Hígado/patología , Macrófagos/microbiología , Macrófagos/patología , Melioidosis/genética , Melioidosis/metabolismo , Mesocricetus , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Virulencia/genética , Factores de Virulencia/genética
16.
FEMS Immunol Med Microbiol ; 52(3): 379-88, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18294191

RESUMEN

Melioidosis is caused by the facultative intracellular bacterium, Burkholderia pseudomallei. Using C57BL/6 mice, we investigated the role of macrophages, TNF-alpha, TNF receptor-1 (TNFR1) and TNF receptor-2 (TNFR2) in host defense against B. pseudomallei using an experimental model of melioidosis. This study has demonstrated that in vivo depletion of macrophages renders C57BL/6 mice highly susceptible to intranasal infection with B. pseudomallei, with significant mortality occurring within 5 days of infection. Using knockout mice, we have also shown that TNF-alpha and both TNFR1 and TNFR2 are required for optimal control of B. pseudomallei infection. Compared with control mice, increased bacterial loads were demonstrated in spleen and liver of knockout mice at day 2 postinfection, correlating with increased inflammatory infiltrates comprised predominantly of neutrophils and widespread necrosis. Following infection with B. pseudomallei, mortality rates of 85.7%, 70% and 91.7% were observed for mice deficient in TNF-alpha, TNFR1 and TNFR2, respectively. Comparison of survival, bacterial loads and histology indicate that macrophages, TNF-alpha, TNFR1 or TNFR2 play a role in controlling rapid dissemination of B. pseudomallei.


Asunto(s)
Burkholderia pseudomallei/inmunología , Burkholderia pseudomallei/patogenicidad , Melioidosis/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Animales , Burkholderia pseudomallei/genética , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Hígado/inmunología , Hígado/microbiología , Macrófagos/microbiología , Macrófagos/fisiología , Melioidosis/genética , Melioidosis/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores del Factor de Necrosis Tumoral , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Bazo/inmunología , Bazo/microbiología
17.
J Infect Dis ; 195(1): 99-107, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17152013

RESUMEN

Inhalation is an important route of infection with Burkholderia pseudomallei, the causative agent of melioidosis. In resistant C57BL/6 mice, activated neutrophils are rapidly recruited to the lungs after intranasal B. pseudomallei infection. Prevention of this response by use of the anti-Gr-1+ cell-depleting monoclonal antibody RB6-8C5 severely exacerbated disease, resulting in an acute lethal infection associated with a 1000-fold increase in lung bacterial loads within 4 days. C57BL/6 interferon (IFN)-gamma(-/-) mice were also acutely susceptible to pulmonary B. pseudomallei infection, dying within 3 days of challenge; this suggests that IFN-gamma is essential for control in the lungs and precedes the protective role of neutrophils in resistance. In neutrophil-depleted mice, lung concentrations of tumor necrosis factor (TNF)-alpha, IFN-gamma, and interleukin-6 were decreased by up to 98%. Natural killer cells were the principle source of IFN-gamma, and monocytes were the principle source of TNF-alpha, suggesting that neutrophils play an important indirect role in the generation of the early cytokine environment in the lungs.


Asunto(s)
Burkholderia pseudomallei/inmunología , Burkholderia pseudomallei/patogenicidad , Citocinas/biosíntesis , Inmunidad Innata/inmunología , Melioidosis/inmunología , Neutrófilos/fisiología , Animales , Citocinas/genética , Modelos Animales de Enfermedad , Melioidosis/genética , Melioidosis/fisiopatología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neutrófilos/metabolismo
18.
Infect Immun ; 68(4): 2034-42, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10722599

RESUMEN

Production of cytokines including gamma interferon (IFN-gamma) and tumor necrosis factor alpha (TNF-alpha) is an important early-stage host response following infection with intracellular pathogens. Development of immunity to these pathogens is determined to a large extent by the timing and relative level of expression of the cytokines. Numerous studies have shown that early cytokine responses involving interleukin-12 (IL-12) and IFN-gamma are important for resistance to intracellular pathogens, whereas responses involving IL-4 and IL-10 increase host susceptibility. These often-indistinct early cytokine responses influence the differentiation of naïve CD4(+) T helper cells, which later develop into what have commonly been termed Th1- and Th2-type cells. The characterization of CD4(+) T-helper-cell responses as Th1 or Th2 type is based largely on the cytokine profiles during the specific phase and has been used in recent years to account for the innate resistance and susceptibility of different inbred strains of mice to several intracellular pathogens. Studies investigating cytokine production in terms of CD4(+) T-helper-cell polarization in Burkholderia pseudomallei infection have not been undertaken. In this study, we used semiquantitative reverse transcription-PCR to assess induction of cytokine mRNA in liver and spleen of B. pseudomallei-susceptible BALB/c and relatively resistant C57BL/6 mice following infection with virulent B. pseudomallei. The levels of mRNA for IFN-gamma, TNF-alpha, IL-1beta, IL-6, IL-10, and IL-12 increased in both BALB/c and C57BL/6 mice 24 to 36 h after infection. A comparison of BALB/c and C57BL/6 responses revealed the relative levels of expression of mRNA for several of these cytokines, including IFN-gamma, were greater in BALB/c mice, suggesting a role for endotoxic shock and cytokine-mediated immunopathology in the development of acute melioidosis. Early induction of mRNA for the cytokines classically associated with development of Th1- and Th2-type responses was absent or minimal, and induction levels in both strains of mice were similar. During the specific phase, cytokine mRNA profiles occurred as a combination of Th1- and Th2-type patterns. Collectively, these results demonstrate that cytokine mRNA responses in BALB/c and C57BL/6 mice following infection with virulent B. pseudomallei do not develop as polarized Th1- or Th2-type profiles. Considering the role of TNF-alpha and IFN-gamma in the processes of endotoxic shock, these results also indicate that selected cytokines, while important for resistance to B. pseudomallei infection, are also potential contributors to immunopathology and the development of acute fulminating disease.


Asunto(s)
Burkholderia pseudomallei/inmunología , Burkholderia pseudomallei/patogenicidad , Citocinas/biosíntesis , Melioidosis/metabolismo , Animales , Citocinas/genética , Femenino , Expresión Génica , Interferón gamma/biosíntesis , Interleucina-1/biosíntesis , Interleucina-10/biosíntesis , Interleucina-12/biosíntesis , Interleucina-2/biosíntesis , Interleucina-4/biosíntesis , Interleucina-6/biosíntesis , Hígado/metabolismo , Hígado/microbiología , Masculino , Melioidosis/genética , Melioidosis/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/metabolismo , Bazo/microbiología , Factor de Necrosis Tumoral alfa/biosíntesis , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA