Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Cell Host Microbe ; 32(6): 913-924.e7, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38870903

RESUMO

Aspects of how Burkholderia escape the host's intrinsic immune response to replicate in the cell cytosol remain enigmatic. Here, we show that Burkholderia has evolved two mechanisms to block the activity of Ring finger protein 213 (RNF213)-mediated non-canonical ubiquitylation of bacterial lipopolysaccharide (LPS), thereby preventing the initiation of antibacterial autophagy. First, Burkholderia's polysaccharide capsule blocks RNF213 association with bacteria and second, the Burkholderia deubiquitylase (DUB), TssM, directly reverses the activity of RNF213 through a previously unrecognized esterase activity. Structural analysis provides insight into the molecular basis of TssM esterase activity, allowing it to be uncoupled from its isopeptidase function. Furthermore, a putative TssM homolog also displays esterase activity and removes ubiquitin from LPS, establishing this as a virulence mechanism. Of note, we also find that additional immune-evasion mechanisms exist, revealing that overcoming this arm of the host's immune response is critical to the pathogen.


Assuntos
Proteínas de Bactérias , Burkholderia , Lipopolissacarídeos , Ubiquitinação , Lipopolissacarídeos/metabolismo , Humanos , Burkholderia/imunologia , Proteínas de Bactérias/metabolismo , Esterases/metabolismo , Evasão da Resposta Imune , Ubiquitina-Proteína Ligases/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Autofagia , Virulência
2.
mBio ; 12(4): e0205421, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34399626

RESUMO

Inflammasomes are cytosolic multiprotein signaling complexes that are activated upon pattern recognition receptor-mediated recognition of pathogen-derived ligands or endogenous danger signals. Their assembly activates the downstream inflammatory caspase-1 and caspase-4/5 (human) or caspase-11 (mouse), which induces cytokine release and pyroptotic cell death through the cleavage of the pore-forming effector gasdermin D. Pathogen detection by host cells also results in the production and release of interferons (IFNs), which fine-tune inflammasome-mediated responses. IFN-induced guanylate-binding proteins (GBPs) have been shown to control the activation of the noncanonical inflammasome by recruiting caspase-4 on the surface of cytosolic Gram-negative bacteria and promoting its interaction with lipopolysaccharide (LPS). The Gram-negative opportunistic bacterial pathogen Burkholderia thailandensis infects epithelial cells and macrophages and hijacks the host actin polymerization machinery to spread into neighboring cells. This process causes host cell fusion and the formation of so-called multinucleated giant cells (MNGCs). Caspase-1- and IFN-regulated caspase-11-mediated inflammasome pathways play an important protective role against B. thailandensis in mice, but little is known about the role of IFNs and inflammasomes during B. thailandensis infection of human cells, particularly epithelial cells. Here, we report that IFN-γ priming of human epithelial cells restricts B. thailandensis-induced MNGC formation in a GBP1-dependent manner. Mechanistically, GBP1 does not promote bacteriolysis or impair actin-based bacterial motility but acts by inducing caspase-4-dependent pyroptosis of the infected cell. In addition, we show that IFN-γ priming of human primary macrophages confers a more efficient antimicrobial effect through inflammasome activation, further confirming the important role that interferon signaling plays in restricting Burkholderia replication and spread. IMPORTANCE The Gram-negative bacteria of the Burkholderia species are associated with human diseases ranging from pneumonia to life-threatening melioidosis. Upon infection through inhalation, ingestion, or the percutaneous route, these bacteria can spread and establish granuloma-like lesions resulting from the fusion of host cells to form multinucleated giant cells (MNGCs). Burkholderia resistance to several antibiotics highlights the importance to better understand how the innate immune system controls infections. Here, we report that interferons protect human epithelial cells against Burkholderia-induced MNGC formation, specifically through the action of the interferon-induced GBP1 protein. Mechanistically, GBP1 acts by inducing caspase-4-dependent cell death through pyroptosis, allowing the infected cells to be quickly eliminated before bacterial spread and the formation of MNGCs. This study provides evidence that interferon-induced innate immune activation, through GBP1 and caspase-4, confers protection against Burkholderia infection, potentially opening new perspectives for therapeutic approaches.


Assuntos
Burkholderia/imunologia , Células Epiteliais/microbiologia , Proteínas de Ligação ao GTP/genética , Células Gigantes/microbiologia , Inflamassomos/imunologia , Interferon gama/metabolismo , Burkholderia/química , Burkholderia/genética , Fusão Celular , Citosol , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Células Gigantes/fisiologia , Células HeLa , Humanos , Inflamassomos/genética , Interferon gama/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/fisiologia , Fagocitose , Transdução de Sinais/imunologia
3.
mBio ; 12(3): e0122721, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34182777

RESUMO

Burkholderia pseudomallei is the causative agent of melioidosis, a fatal disease with a high mortality rate. The intrinsic resistance to commonly used antibiotics combined with the complex bacterial life cycle has hampered the development of preventive and therapeutic interventions and vaccines. Furthermore, the need of humoral and cell-mediated immunity in protection against B. pseudomallei has complicated the development of effective vaccines. Antigen delivery vaccine platforms that promote humoral and cellular responses while maintaining a safe profile are a roadblock to developing subunit vaccines against intracellular pathogens. Gold nanoparticles (AuNPs) were used for the delivery of multicomponent antigens with the goal of inducing vaccine-mediated immunity, promoting protection against melioidosis disease. Different nanoglycoconjugates using predicted immunogenic protein candidates, Hcp1, FlgL, OpcP, OpcP1, OmpW, and hemagglutinin, were covalently coupled to AuNPs, together with the lipopolysaccharide (LPS) from Burkholderia thailandensis, which acted as an additional antigen. Animals immunized with individually coupled (AuNP-protein-LPS) formulations containing OpcP or OpcP1, together with CpG as an adjuvant, showed a significant increase in protection, whereas a nanovaccine combination (AuNP-Combo2-LPS) showed significant and complete protection against a lethal intranasal B. pseudomallei challenge. Animals immunized with AuNP-Combo2-LPS showed robust humoral antigen-specific (IgG and IgA) responses with higher IgG2c titer, indicating a TH1-skewed response and promotion of macrophage uptake. In addition, immunization with the nanovaccine combination resulted in a mixed antigen-specific TH1-TH17 cytokine profile after immunization. This study provides the basis for an elegant and refined multicomponent glycoconjugate vaccine formulation capable of eliciting both humoral and cell-mediated responses against lethal B. pseudomallei challenge. IMPORTANCE Melioidosis is a complex human disease associated with a wide range of complications caused by the Gram-negative bacillus Burkholderia pseudomallei. The global burden of melioidosis is estimated to have 165,000 cases per year and 89,000 fatal outcomes. The endemicity of B. pseudomallei includes a wide range of tropical regions in Asia, Africa, Latin America, and Australia. Therefore, a viable alternative to prevent human infections is the development of an effective vaccine; however, no approved vaccine for human use is available. This study provides a vaccine strategy against B. pseudomallei and an immune-stimulatory platform to induce strong humoral and T-cell-mediated immunity.


Assuntos
Vacinas Bacterianas/imunologia , Burkholderia pseudomallei/imunologia , Ouro , Imunidade Humoral , Melioidose/prevenção & controle , Células Th1/imunologia , Células Th17/imunologia , Adjuvantes Imunológicos , Animais , Anticorpos Antibacterianos/sangue , Vacinas Bacterianas/administração & dosagem , Burkholderia/imunologia , Feminino , Glicoconjugados/química , Imunidade Celular , Melioidose/imunologia , Nanopartículas Metálicas/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vacinação
4.
mBio ; 12(3): e0105921, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34154417

RESUMO

Burkholderia infections can result in serious diseases with high mortality, such as melioidosis, and they are difficult to treat with antibiotics. Innate immunity is critical for cell-autonomous clearance of intracellular pathogens like Burkholderia by regulating programmed cell death. Inflammasome-dependent inflammatory cytokine release and cell death contribute to host protection against Burkholderia pseudomallei and Burkholderia thailandensis; however, the contribution of apoptosis and necroptosis to protection is not known. Here, we found that bone marrow-derived macrophages (BMDMs) lacking key components of pyroptosis died via apoptosis during infection. BMDMs lacking molecules required for pyroptosis, apoptosis, and necroptosis (PANoptosis), however, were significantly resistant to B. thailandensis-induced cell death until later stages of infection. Consequently, PANoptosis-deficient BMDMs failed to limit B. thailandensis-induced cell-cell fusion, which permits increased intercellular spread and replication compared to wild-type or pyroptosis-deficient BMDMs. Respiratory B. thailandensis infection resulted in higher mortality in PANoptosis-deficient mice than in pyroptosis-deficient mice, indicating that, in the absence of pyroptosis, apoptosis is essential for efficient control of infection in vivo. Together, these findings suggest both pyroptosis and apoptosis are necessary for host-mediated control of Burkholderia infection. IMPORTANCEBurkholderia infections result in a high degree of mortality when left untreated; therefore, understanding the host immune response required to control infection is critical. In this study, we found a hierarchical cell death program utilized by infected cells to disrupt the intracellular niche of Burkholderia thailandensis, which limits bacterial intercellular spread, host cell-cell fusion, and bacterial replication. In macrophages, combined loss of key PANoptosis components results in extensive B. thailandensis infection-induced cell-cell fusion, bacterial replication, and increased cell death at later stages of infection compared with both wild-type (WT) and pyroptosis-deficient cells. During respiratory infection, mortality was increased in PANoptosis-deficient mice compared to pyroptosis-deficient mice, identifying an essential role for multiple cell death pathways in controlling B. thailandensis infection. These findings advance our understanding of the physiological role of programmed cell death in controlling Burkholderia infection.


Assuntos
Apoptose/imunologia , Infecções por Burkholderia/imunologia , Burkholderia/patogenicidade , Imunidade Inata , Macrófagos/microbiologia , Macrófagos/patologia , Animais , Burkholderia/imunologia , Caspases/classificação , Caspases/genética , Caspases/imunologia , Feminino , Masculino , Camundongos , Necroptose/imunologia , Piroptose/imunologia
5.
Cells ; 10(3)2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33668922

RESUMO

Increasing antimicrobial resistance due to misuse and overuse of antimicrobials, as well as a lack of new and innovative antibiotics in development has become an alarming global threat. Preventative therapeutics, like vaccines, are combative measures that aim to stop infections at the source, thereby decreasing the overall use of antibiotics. Infections due to Gram-negative pathogens pose a significant treatment challenge because of substantial multidrug resistance that is acquired and spread throughout the bacterial population. Burkholderia spp. are Gram-negative intrinsically resistant bacteria that are responsible for environmental and nosocomial infections. The Burkholderia cepacia complex are respiratory pathogens that primarily infect immunocompromised and cystic fibrosis patients, and are acquired through contaminated products and equipment, or via patient-to-patient transmission. The Burkholderia pseudomallei complex causes percutaneous wound, cardiovascular, and respiratory infections. Transmission occurs through direct exposure to contaminated water, water-vapors, or soil, leading to the human disease melioidosis, or the equine disease glanders. Currently there is no licensed vaccine against any Burkholderia pathogen. This review will discuss Burkholderia vaccine candidates derived from outer membrane proteins, OmpA, OmpW, Omp85, and Bucl8, encompassing their structures, conservation, and vaccine formulation.


Assuntos
Vacinas Bacterianas/imunologia , Burkholderia/imunologia , Proteínas de Membrana/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Bactérias/imunologia , Humanos , Proteínas de Membrana/química , Modelos Biológicos
6.
STAR Protoc ; 2(1): 100244, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33458706

RESUMO

Characterizing cytokine production in situ is important for properly understanding immunologic responses. Cytokine reporter mice are limited by the need to cross markers into various knockout backgrounds and by availability of reporters of interest. To overcome this, we utilize injection of brefeldin A into mice to enable flow cytometric analysis of in situ cytokine production during a bacterial infection. While we evaluate IFN-γ production during Burkholderia thailandensis infection, this protocol can be applied to other cytokines and other mouse models. For complete details on the use and execution of this protocol, please refer to Kovacs et al. (2020) and Liu and Whitton (2005).


Assuntos
Brefeldina A/química , Infecções por Burkholderia/imunologia , Burkholderia/imunologia , Citometria de Fluxo , Interferon gama/imunologia , Animais , Camundongos
7.
Cells ; 9(12)2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33322641

RESUMO

The genus Burkholderia includes a wide range of Gram-negative bacterial species some of which are pathogenic to humans and other vertebrates. The most pathogenic species are Burkholderia mallei, Burkholderia pseudomallei, and the members of the Burkholderia cepacia complex (Bcc). B. mallei and B. pseudomallei, the cause of glanders and melioidosis, respectively, are considered potential bioweapons. The Bcc comprises a subset of Burkholderia species associated with respiratory infections in people with chronic granulomatous disease and cystic fibrosis. Antimicrobial treatment of Burkholderia infections is difficult due to the intrinsic multidrug antibiotic resistance of these bacteria; prophylactic vaccines provide an attractive alternative to counteract these infections. Although commercial vaccines against Burkholderia infections are still unavailable, substantial progress has been made over recent years in the development of vaccines against B. pseudomallei and B. mallei. This review critically discusses the current advances in vaccine development against B. mallei, B. pseudomallei, and the Bcc.


Assuntos
Vacinas Bacterianas/administração & dosagem , Infecções por Burkholderia/prevenção & controle , Burkholderia/imunologia , Animais , Vacinas Bacterianas/imunologia , Burkholderia/genética , Burkholderia/patogenicidade , Infecções por Burkholderia/imunologia , Infecções por Burkholderia/microbiologia , Humanos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
8.
PLoS Pathog ; 16(3): e1008364, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32150572

RESUMO

Innate immunity responds to pathogens by producing alarm signals and activating pathways that make host cells inhospitable for pathogen replication. The intracellular bacterium Burkholderia thailandensis invades the cytosol, hijacks host actin, and induces cell fusion to spread to adjacent cells, forming multinucleated giant cells (MNGCs) which promote bacterial replication. We show that type I interferon (IFN) restricts macrophage MNGC formation during B. thailandensis infection. Guanylate-binding proteins (GBPs) expressed downstream of type I IFN were required to restrict MNGC formation through inhibition of bacterial Arp2/3-dependent actin motility during infection. GTPase activity and the CAAX prenylation domain were required for GBP2 recruitment to B. thailandensis, which restricted bacterial actin polymerization required for MNGC formation. Consistent with the effects in in vitro macrophages, Gbp2-/-, Gbp5-/-, GbpChr3-KO mice were more susceptible to intranasal infection with B. thailandensis than wildtype mice. Our findings reveal that IFN and GBPs play a critical role in restricting cell-cell fusion and bacteria-induced pathology during infection.


Assuntos
Infecções por Burkholderia/imunologia , Burkholderia/imunologia , Proteínas de Ligação ao GTP/imunologia , Células Gigantes/imunologia , Macrófagos/imunologia , Doenças Nasais/imunologia , Prenilação de Proteína/imunologia , Animais , Infecções por Burkholderia/genética , Infecções por Burkholderia/patologia , Fusão Celular , Proteínas de Ligação ao GTP/genética , Células Gigantes/microbiologia , Células Gigantes/patologia , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , Doenças Nasais/genética , Doenças Nasais/microbiologia , Doenças Nasais/patologia
9.
Emerg Infect Dis ; 26(3): 463-471, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32091359

RESUMO

Melioidosis is a neglected tropical disease with an estimated annual mortality rate of 89,000 in 45 countries across tropical regions. The causative agent is Burkholderia pseudomallei, a gram-negative soil-dwelling bacterium. In Thailand, B. pseudomallei can be found across multiple regions, along with the low-virulence B. thailandensis and the recently discovered B. thailandensis variant (BTCV), which expresses B. pseudomallei-like capsular polysaccharide. Comprehensive studies of human immune responses to B. thailandensis variants and cross-reactivity to B. pseudomallei are not complete. We evaluated human immune responses to B. pseudomallei, B. thailandensis, and BTCV in melioidosis patients and healthy persons in B. pseudomallei-endemic areas using a range of humoral and cellular immune assays. We found immune cross-reactivity to be strong for both humoral and cellular immunity among B. pseudomallei, B. thailandensis, and BTCV. Our findings suggest that environmental exposure to low-virulence strains may build cellular immunity to B. pseudomallei.


Assuntos
Burkholderia/imunologia , Melioidose/epidemiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Burkholderia/patogenicidade , Estudos de Coortes , Reações Cruzadas , Feminino , Humanos , Imunidade , Masculino , Melioidose/microbiologia , Pessoa de Meia-Idade , Estudos Prospectivos , Tailândia/epidemiologia , Virulência , Adulto Jovem
10.
Infect Immun ; 87(12)2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31548320

RESUMO

Lipopolysaccharides (LPSs) of Gram-negative bacteria comprise lipid A, core, and O-polysaccharide (OPS) components. Studies have demonstrated that LPSs isolated from the pathogenic species Burkholderia pseudomallei and Burkholderia mallei and from less-pathogenic species, such as Burkholderia thailandensis, are potent immune stimulators. The LPS structure of B. pseudomallei, the causative agent of melioidosis, is highly conserved in isolates from Thailand; however, the LPSs isolated from other, related species have not been characterized to enable understanding of their immune recognition and antigenicities. Here, we describe the structural and immunological characteristics of the LPSs isolated from eight Burkholderia species and compare those for B. pseudomallei to those for the other seven species. Matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS), gas chromatography (GC), SDS-PAGE, Toll-like receptor 4 (TLR4) stimulation, and immunoblot analysis were performed on these Burkholderia species. MALDI-TOF profiles demonstrated that Burkholderia lipid A contains predominantly penta-acylated species modified with 4-amino-4-deoxy-arabinose residues at both terminal phosphate groups. The lipid A could be differentiated based on mass differences at m/z 1,511, 1,642, 1,773, and 1,926 and on fatty acid composition. LPSs of all species induced TLR4-dependent NF-κB responses; however, while SDS-PAGE analysis showed similar LPS ladder patterns for B. pseudomallei, B. thailandensis, and B. mallei, these patterns differed from those of other Burkholderia species. Interestingly, immunoblot analysis demonstrated that melioidosis patient sera cross-reacted with OPSs of other Burkholderia species. These findings can be used to better understand the characteristics of LPS in Burkholderia species, and they have implications for serological diagnostics based on the detection of antibodies to OPS.


Assuntos
Burkholderia mallei/imunologia , Burkholderia pseudomallei/imunologia , Burkholderia/imunologia , Lipídeo A/imunologia , Receptor 4 Toll-Like/metabolismo , Amino Açúcares/química , Anticorpos Antibacterianos/imunologia , Reações Cruzadas/imunologia , Humanos , Lipídeo A/química , Melioidose/imunologia , Melioidose/microbiologia , Conformação Molecular , Polissacarídeos Bacterianos/imunologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
11.
BMC Med Genomics ; 12(1): 127, 2019 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-31492148

RESUMO

BACKGROUND: Burkholderia mallei (Bm) is a facultative intracellular bacterial pathogen causing highly-fatal glanders in solipeds and humans. The ability of Bm to thrive intracellularly is thought to be related to exploitation of host immune response-related genes and pathways. Relatively little is known of the molecular strategies employed by this pathogen to modulate these pathways and evade intracellular killing. This manuscript seeks to fill gaps in the understanding of the interface between Bm and innate immunity by examining gene expression changes during infection of host monocytes. METHODS: The transcriptome of Bm-infected human Mono Mac-6 (MM6) monocytes was profiled on Affymetrix Human Transcriptome GeneChips 2.0. Gene expression changes in Bm-infected monocytes were compared to those of Burkholderia thailandensis (Bt)-infected monocytes and to uninfected monocytes. The resulting dataset was normalized using Robust Multichip Average and subjected to statistical analyses employing a univariate F test with a random variance model. Differentially expressed genes significant at p < 0.001 were subjected to leave-one-out cross-validation studies and 1st and 3rd nearest neighbor prediction model. Significant probe sets were used to populate human pathways in Ingenuity Pathway Analysis, with statistical significance determined by Fisher's exact test or z-score. RESULTS: The Pattern Recognition Receptor (PRR) pathway was represented among significantly enriched immune response-related human canonical pathways, with evidence of upregulation across both infections. Among members of this pathway, pentraxin-3 was significantly upregulated by Bm- or Bt-infected monocytes. Pentraxin-3 (PTX3) was demonstrated to bind to both Bt and Burkholderia pseudomallei (Bp), but not Bm. Subsequent assays did not identify a role for PTX3 in potentiating complement-mediated lysis of Bt or in enhancing phagocytosis or replication of Bt in human monocytes. CONCLUSION: We report on the novel binding of PTX3 to Bt and Bp, with lack of interaction with Bm, suggesting that a possible evasive mechanism by Bm warrants further exploration. We determined that (1) PTX3 may not play a role in activating the lytic pathway of complement in different bacterial species and that (2) the opsonophagocytic properties of PTX3 should be investigated in different primary or immortalized cell lines representing host phagocytes, given lack of binding of PTX3 to MM6 monocytes.


Assuntos
Burkholderia/imunologia , Proteína C-Reativa/metabolismo , Perfilação da Expressão Gênica , Imunidade Inata , Monócitos/imunologia , Monócitos/microbiologia , Componente Amiloide P Sérico/metabolismo , Anticorpos/metabolismo , Burkholderia/crescimento & desenvolvimento , Linhagem Celular , Proteínas do Sistema Complemento/metabolismo , Humanos , Imunidade Inata/genética , Viabilidade Microbiana , Proteínas Opsonizantes/metabolismo , Fagocitose , Ligação Proteica , Regulação para Cima/genética
12.
J Proteome Res ; 18(7): 2848-2858, 2019 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-31244210

RESUMO

In Thailand, diabetes mellitus is the most significant risk factor for melioidosis, a severe disease caused by Burkholderia pseudomallei. In this study, neutrophils isolated from healthy or diabetic subjects were infected with B. thailandensis E555, a variant strain with a B. pseudomallei-like capsular polysaccharide used here as a surrogate micro-organism for B. pseudomallei. At 2 h post-infection, neutrophil proteins were subjected to 4-plex iTRAQ-based comparative proteomic analysis. A total of 341 proteins were identified in two or more samples, of which several proteins involved in oxidative stress and inflammation were enriched in infected diabetic neutrophils. We validated this finding by demonstrating that infected diabetic neutrophils generated significantly elevated levels of pro-inflammatory cytokines TNFα, IL-6, IL-1ß, and IL-17 compared to healthy neutrophils. Our data also revealed that infected neutrophils from healthy or diabetic individuals undergo apoptotic cell death at distinctly different rates, with infected diabetic neutrophils showing a diminished ability to delay apoptosis and an increased likelihood of undergoing a lytic form of cell death, compared to infected neutrophils from healthy individuals. Increased expression of inflammatory proteins by infected neutrophils could contribute to the increased susceptibility to infection and inflammation in diabetic patients in melioidosis-endemic areas.


Assuntos
Infecções por Burkholderia/imunologia , Burkholderia/imunologia , Diabetes Mellitus/patologia , Neutrófilos/imunologia , Proteômica , Estudos de Casos e Controles , Morte Celular , Células Cultivadas , Citocinas/metabolismo , Diabetes Mellitus/microbiologia , Humanos , Inflamação/metabolismo , Melioidose/etiologia , Neutrófilos/metabolismo , Neutrófilos/microbiologia
13.
Sci Signal ; 12(571)2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30837305

RESUMO

Bacterial lectins are typically multivalent and bind noncovalently to specific carbohydrates on host tissues to facilitate bacterial adhesion. Here, we analyzed the effects of two fucose-binding lectins, BambL from Burkholderia ambifaria and LecB from Pseudomonas aeruginosa, on specific signaling pathways in B cells. We found that these bacterial lectins induced B cell activation, which, in vitro, was dependent on the cell surface expression of the B cell antigen receptor (BCR) and its co-receptor CD19, as well as on spleen tyrosine kinase (Syk) activity. The resulting release of intracellular Ca2+ was followed by an increase in the cell surface abundance of the activation marker CD86, augmented cytokine secretion, and subsequent cell death, replicating all of the events that are observed in vitro upon canonical and antigen-mediated B cell activation. Moreover, injection of BambL in mice resulted in a substantial, BCR-independent loss of B cells in the bone marrow with simultaneous, transient enlargement of the spleen (splenomegaly), as well as an increase in the numbers of splenic B cells and myeloid cells. Together, these data suggest that bacterial lectins can initiate polyclonal activation of B cells through their sole capacity to bind to fucose.


Assuntos
Linfócitos B/imunologia , Proteínas de Bactérias/imunologia , Burkholderia/imunologia , Carboidratos/imunologia , Lectinas/imunologia , Ativação Linfocitária , Pseudomonas aeruginosa/imunologia , Transdução de Sinais/imunologia , Animais , Antígenos CD19/genética , Antígenos CD19/imunologia , Antígeno B7-2/genética , Antígeno B7-2/imunologia , Proteínas de Bactérias/genética , Carboidratos/genética , Camundongos , Camundongos Knockout , Transdução de Sinais/genética , Quinase Syk/genética , Quinase Syk/imunologia
15.
ACS Infect Dis ; 3(10): 736-743, 2017 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-28707874

RESUMO

Structure-based epitope prediction drives the design of diagnostic peptidic probes to reveal specific antibodies elicited in response to infections. We previously identified a highly immunoreactive epitope from the peptidoglycan-associated lipoprotein (Pal) antigen from Burkholderia pseudomallei, which could also diagnose Burkholderia cepacia infections. Here, considering the high phylogenetic conservation within Burkholderia species, we ask whether cross-reactivity can be reciprocally displayed by the synthetic epitope from B. cenocepacia. We perform comparative analyses of the conformational preferences and diagnostic performances of the corresponding epitopes from the two Burkholderia species when presented in the context of the full-length proteins or as isolated peptides. The effects of conformation on the diagnostic potential and cross-reactivity of Pal peptide epitopes are rationalized on the basis of the 1.8 Å crystal structure of B. cenocepacia Pal and through computational analyses. Our results are discussed in the context of designing new diagnostic molecules for the early detection of infectious diseases.


Assuntos
Infecções por Burkholderia/diagnóstico , Burkholderia/imunologia , Mapeamento de Epitopos/métodos , Epitopos/metabolismo , Imunoensaio/métodos , Anticorpos/imunologia , Anticorpos/fisiologia , Proteínas de Bactérias , Infecções por Burkholderia/microbiologia , Clonagem Molecular , Simulação por Computador , Regulação Bacteriana da Expressão Gênica , Humanos , Modelos Moleculares , Conformação Proteica , Proteínas Recombinantes
16.
Artigo em Inglês | MEDLINE | ID: mdl-28638804

RESUMO

Pathogenic Burkholderia rely on host factors for efficient intracellular replication and are highly refractory to antibiotic treatment. To identify host genes that are required by Burkholderia spp. during infection, we performed a RNA interference (RNAi) screen of the human kinome and identified 35 host kinases that facilitated Burkholderia thailandensis intracellular survival in human monocytic THP-1 cells. We validated a selection of host kinases using imaging flow cytometry to assess efficiency of B. thailandensis survival in the host upon siRNA-mediated knockdown. We focused on the role of the novel protein kinase C isoform, PKC-η, in Burkholderia infection and characterized PKC-η/MARCKS signaling as a key event that promotes the survival of unopsonized B. thailandensis CDC2721121 within host cells. While infection of lung epithelial cells with unopsonized Gram-negative bacteria stimulated phosphorylation of Ser175/160 in the MARCKS effector domain, siRNA-mediated knockdown of PKC-η expression reduced the levels of phosphorylated MARCKS by >3-fold in response to infection with Bt CDC2721121. We compared the effect of the conventional PKC-α and novel PKC-η isoforms on the growth of B. thailandensis CDC2721121 within monocytic THP-1 cells and found that ≥75% knock-down of PRKCH transcript levels reduced intracellular bacterial load 100% more efficiently when compared to growth in cells siRNA-depleted of the classical PKC-α, suggesting that the PKC-η isoform can specifically mediate Burkholderia intracellular survival. Based on imaging studies of intracellular B. thailandensis, we found that PKC-η function stimulates phagocytic pathways that promote B. thailandensis escape into the cytoplasm leading to activation of autophagosome flux. Identification of host kinases that are targeted by Burkholderia during infection provides valuable molecular insights in understanding Burkholderia pathogenesis, and ultimately, in designing effective host-targeted therapies against infectious disease caused by intracellular pathogens.


Assuntos
Infecções por Burkholderia/imunologia , Burkholderia/imunologia , Burkholderia/patogenicidade , Citoplasma/imunologia , Interações Hospedeiro-Parasita/imunologia , Substrato Quinase C Rico em Alanina Miristoilada/metabolismo , Proteína Quinase C/metabolismo , Transdução de Sinais , Autofagossomos , Carga Bacteriana , Burkholderia/crescimento & desenvolvimento , Infecções por Burkholderia/microbiologia , Citoplasma/microbiologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Regulação da Expressão Gênica/imunologia , Técnicas de Silenciamento de Genes , Humanos , Pulmão/microbiologia , Fosforilação , Isoformas de Proteínas/metabolismo , Proteína Quinase C/química , Interferência de RNA , RNA Interferente Pequeno/genética , Células THP-1
17.
Infect Immun ; 85(6)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28348057

RESUMO

Burkholderia dolosa caused an outbreak in the cystic fibrosis (CF) clinic at Boston Children's Hospital from 1998 to 2005 and led to the infection of over 40 patients, many of whom died due to complications from infection by this organism. To assess whether B. dolosa significantly contributes to disease or is recognized by the host immune response, mice were infected with a sequenced outbreak B. dolosa strain, AU0158, and responses were compared to those to the well-studied CF pathogen Pseudomonas aeruginosa In parallel, mice were also infected with a polar flagellin mutant of B. dolosa to examine the role of flagella in B. dolosa lung colonization. The results showed a higher persistence in the host by B. dolosa strains, and yet, neutrophil recruitment and cytokine production were lower than those with P. aeruginosa The ability of host immune cells to recognize B. dolosa was then assessed, B. dolosa induced a robust cytokine response in cultured cells, and this effect was dependent on the flagella only when bacteria were dead. Together, these results suggest that B. dolosa can be recognized by host cells in vitro but may avoid or suppress the host immune response in vivo through unknown mechanisms. B. dolosa was then compared to other Burkholderia species and found to induce similar levels of cytokine production despite being internalized by macrophages more than Burkholderia cenocepacia strains. These data suggest that B. dolosa AU0158 may act differently with host cells and is recognized differently by immune systems than are other Burkholderia strains or species.


Assuntos
Infecções por Burkholderia/imunologia , Fibrose Cística/complicações , Citocinas/imunologia , Flagelos/imunologia , Flagelina/genética , Animais , Lavagem Broncoalveolar , Burkholderia/genética , Burkholderia/imunologia , Infecções por Burkholderia/microbiologia , Linhagem Celular , Fibrose Cística/microbiologia , Modelos Animais de Doenças , Epidemias , Feminino , Flagelos/genética , Humanos , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/imunologia
18.
Dev Comp Immunol ; 67: 97-106, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27825951

RESUMO

Riptortus pedestris, a common pest in soybean fields, harbors a symbiont Burkholderia in a specialized posterior midgut region of insects. Every generation of second nymphs acquires new Burkholderia cells from the environment. We compared in vitro cultured Burkholderia with newly in vivo colonized Burkholderia in the host midgut using biochemical approaches. The bacterial cell envelope of in vitro cultured and in vivo Burkholderia differed in structure, as in vivo bacteria lacked lipopolysaccharide (LPS) O-antigen. The LPS O-antigen deficient bacteria had a reduced colonization rate in the host midgut compared with that of the wild-type Burkholderia. To determine why LPS O-antigen-deficient bacteria are less able to colonize the host midgut, we examined in vitro survival rates of three LPS O-antigen-deficient Burkholderia mutants and lysates of five different midgut regions. The LPS O-antigen-deficient mutants were highly susceptible when cultured with the lysate of a specific first midgut region (M1), indicating that the M1 lysate contains unidentified substance(s) capable of killing LPS O-antigen-deficient mutants. We identified a 17 kDa protein from the M1 lysate, which was enriched in the active fractions. The N-terminal sequence of the protein was determined to be a soybean Kunitz-type trypsin inhibitor. These data suggest that the 17 kDa protein, which was originated from a main soybean source of the R. pedestris host, has antibacterial activity against the LPS O-antigen deficient (rough-type) Burkholderia.


Assuntos
Antibacterianos/imunologia , Infecções por Burkholderia/imunologia , Burkholderia/imunologia , Extratos Celulares/imunologia , Trato Gastrointestinal/imunologia , Heterópteros/imunologia , Proteínas de Insetos/imunologia , Inibidores da Tripsina/imunologia , Animais , Burkholderia/genética , Infecções por Burkholderia/genética , Células Cultivadas , Interações Hospedeiro-Patógeno , Lipopolissacarídeos/imunologia , Mutação/genética , Antígenos O/genética , Simbiose
19.
Microbiol Immunol ; 60(11): 725-739, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27862204

RESUMO

Lipopolysaccharide is one of the virulence factors of the soil-borne pathogens Burkholderia pseudomallei, B. thailandensis, B. cenocepacia and B. multivorans, which cause septic melioidosis (often in B. pseudomallei infections but rarely in B. thailandensis infections) or cepacia syndromes (commonly in B. cenocepacia infections but rarely in B. multivorans infections). The inflammatory responses in Burkholderia LPS-induced endotoxemia were evaluated in this study. Prior to induction, the conserved structures and functions of each purified LPS were determined using electrophoretic phenotypes, the ratios of 3-hydroxytetradecanoic to 3-hydroxyhexadecanoic acid and endotoxin units. In an in vitro assay, cytokine expression of myeloid differentiation primary response gene 88 and Toll/IL-1 receptor domain containing adapter-inducing INF-ß-dependent signaling-dependent signaling differed when stimulated by different LPS. Endotoxemia was induced in mice by s.c. injection as evidenced by increasing serum concentrations of 3-hydroxytetradecanoic acid and the septic prognostic markers CD62E and ICAM-1. During endotoxemia, splenic CD11b+ I-A+ , CD11b+ CD80+ , CD11b+ CD86+ and CD11b+ CD11c+ subpopulations increased. After induction with B. pseudomallei LPS, there were significant increases in splenic CD49b NK cells and CD14 macrophages. The inflamed CD11b+ CCR2+ , CD11b+ CD31+ , CD11b+ CD14+ , resident CD11b+ CX3 CR1+ and progenitor CD11b+ CD34+ cells showed delayed increases in bone marrow. B. multivorans LPS was the most potent inducer of serum cytokines and chemokines, whereas B. cenocepacia LPS induced relatively low concentrations of the chemokines MIP-1α and MIP-1ß. Endotoxin activities did not correlate with the virulence of Burkholderia strains. Thus factors other than LPS and/or other mechanisms of low activity LPS must mediate the pathogenicity of highly virulent Burkholderia strains.


Assuntos
Infecções por Burkholderia/imunologia , Burkholderia/imunologia , Endotoxemia/imunologia , Lipopolissacarídeos/imunologia , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Biomarcadores , Medula Óssea/imunologia , Medula Óssea/metabolismo , Infecções por Burkholderia/sangue , Burkholderia pseudomallei/imunologia , Citocinas/biossíntese , Citocinas/sangue , Modelos Animais de Doenças , Endotoxemia/sangue , Endotoxinas/sangue , Feminino , Imunofenotipagem , Lipopolissacarídeos/administração & dosagem , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Baço/citologia , Baço/imunologia , Baço/metabolismo
20.
Dev Comp Immunol ; 64: 75-81, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26774501

RESUMO

Valuable insect models have tremendously contributed to our understanding of innate immunity and symbiosis. Bean bug, Riptortus pedestris, is a useful insect symbiosis model due to harboring cultivable monospecific gut symbiont, genus Burkholderia. Bean bug is a hemimetabolous insect whose immunity is not well-understood. However, we recently identified three major antimicrobial peptides of Riptortus and examined the relationship between gut symbiosis and host immunity. We found that the presence of Burkholderia gut symbiont positively affects Riptortus immunity. From studying host regulation mechanisms of symbiont population, we revealed that the symbiotic Burkholderia cells are much more susceptible to Riptortus immune responses than the cultured cells. We further elucidated that the immune-susceptibility of the Burkholderia gut symbionts is due to the drastic change of bacterial cell envelope. Finally, we show that the immune-susceptible Burkholderia symbionts are able to prosper in host owing to the suppression of immune responses of the symbiotic midgut.


Assuntos
Infecções Bacterianas/imunologia , Burkholderia/imunologia , Hemípteros/imunologia , Imunidade Inata , Intestinos/imunologia , Microbiota , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Infecções Bacterianas/microbiologia , Parede Celular/metabolismo , Suscetibilidade a Doenças , Intestinos/microbiologia , Modelos Animais , Simbiose
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...