Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Nat Prod Rep ; 35(12): 1251-1293, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30023998

ABSTRACT

Covering: up to 2018 Burkholderia species are a vast group of human pathogenic, phytopathogenic, and plant- or environment-associated bacteria. B. pseudomallei, B. mallei, and B. cepacia complex are the causative agents of melioidosis, glanders, and cystic fibrosis-related infections, respectively, which are fatal diseases in humans and animals. Due to their high resistance to antibiotics, high mortality rates, and increased infectivity via the respiratory tract, B. pseudomallei and B. mallei have been listed as potential bioterrorism agents by the Centers for Disease Control and Prevention. Burkholderia species are able to produce a large network of surface-exposed polysaccharides, i.e., lipopolysaccharides, capsular polysaccharides, and exopolysaccharides, which are virulence factors, immunomodulators, major biofilm components, and protective antigens, and have crucial implications in the pathogenicity of Burkholderia-associated diseases. This review provides a comprehensive and up-to-date account regarding the structural elucidation and biological activities of surface polysaccharides produced by Burkholderia species. The chemical synthesis of oligosaccharides mimicking Burkholderia polysaccharides is described in detail. Emphasis is placed on the recent research efforts toward the development of glycoconjugate vaccines against melioidosis and glanders based on synthetic or native Burkholderia oligo/polysaccharides.


Subject(s)
Bacterial Vaccines/pharmacology , Burkholderia/chemistry , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/immunology , Animals , Bacterial Vaccines/immunology , Burkholderia/metabolism , Burkholderia/pathogenicity , Glanders/immunology , Glanders/prevention & control , Glycoconjugates/chemical synthesis , Glycoconjugates/chemistry , Humans , Melioidosis/immunology , Melioidosis/prevention & control , Molecular Mimicry , Plants/microbiology , Polysaccharides, Bacterial/genetics
2.
Infect Immun ; 85(8)2017 08.
Article in English | MEDLINE | ID: mdl-28507073

ABSTRACT

Burkholderia mallei, a facultative intracellular bacterium and tier 1 biothreat, causes the fatal zoonotic disease glanders. The organism possesses multiple genes encoding autotransporter proteins, which represent important virulence factors and targets for developing countermeasures in pathogenic Gram-negative bacteria. In the present study, we investigated one of these autotransporters, BatA, and demonstrate that it displays lipolytic activity, aids in intracellular survival, is expressed in vivo, elicits production of antibodies during infection, and contributes to pathogenicity in a mouse aerosol challenge model. A mutation in the batA gene of wild-type strain ATCC 23344 was found to be particularly attenuating, as BALB/c mice infected with the equivalent of 80 median lethal doses cleared the organism. This finding prompted us to test the hypothesis that vaccination with the batA mutant strain elicits protective immunity against subsequent infection with wild-type bacteria. We discovered that not only does vaccination provide high levels of protection against lethal aerosol challenge with B. mallei ATCC 23344, it also protects against infection with multiple isolates of the closely related organism and causative agent of melioidosis, Burkholderia pseudomallei Passive-transfer experiments also revealed that the protective immunity afforded by vaccination with the batA mutant strain is predominantly mediated by IgG antibodies binding to antigens expressed exclusively in vivo Collectively, our data demonstrate that BatA is a target for developing medical countermeasures and that vaccination with a mutant lacking expression of the protein provides a platform to gain insights regarding mechanisms of protective immunity against B. mallei and B. pseudomallei, including antigen discovery.


Subject(s)
Antibodies, Bacterial/immunology , Burkholderia mallei/immunology , Burkholderia pseudomallei/immunology , Melioidosis/prevention & control , Animals , Bacterial Proteins/genetics , Burkholderia mallei/genetics , Burkholderia mallei/growth & development , Burkholderia mallei/pathogenicity , Burkholderia pseudomallei/pathogenicity , Disease Models, Animal , Glanders/immunology , Glanders/microbiology , Glanders/prevention & control , Immunoglobulin G/immunology , Melioidosis/immunology , Melioidosis/microbiology , Mice , Mice, Inbred BALB C , Mutation , Vaccination , Virulence Factors/genetics
3.
Curr Opin Infect Dis ; 30(3): 297-302, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28177960

ABSTRACT

PURPOSE OF REVIEW: Burkholderia mallei is a facultative intracellular pathogen that causes the highly contagious and often the fatal disease, glanders. With its high rate of infectivity via aerosol and recalcitrance toward antibiotics, this pathogen is considered a potential biological threat agent. This review focuses on the most recent literature highlighting host innate immune response to B. mallei. RECENT FINDINGS: Recent studies focused on elucidating host innate immune responses to the novel mechanisms and virulence factors employed by B. mallei for survival. Studies suggest that pathogen proteins manipulate various cellular processes, including host ubiquitination pathways, phagosomal escape, and actin-cytoskeleton rearrangement. Immune-signaling molecules such as Toll-like receptors, nucleotode-binding oligomerization domain, myeloid differentiation primary response protein 88, and proinflammatory cytokines such as interferon-gamma and tumor necrosis factor-α, play key roles in the induction of innate immune responses. Modifications in B. mallei lipopolysaccharide, in particular, the lipid A acyl groups, stimulate immune responses via Toll-like receptor4 activation that may contribute to persistent infection. SUMMARY: Mortality is high because of septicemia and immune pathogenesis with B. mallei exposure. An effective innate immune response is critical to controlling the acute phase of the infection. Both vaccination and therapeutic approaches are necessary for complete protection against B. mallei.


Subject(s)
Burkholderia mallei/immunology , Glanders/immunology , Immunity, Innate , Animals , Burkholderia mallei/pathogenicity , Cytokines/immunology , Glanders/therapy , Humans , Lipopolysaccharides/immunology , Toll-Like Receptors/immunology , Virulence Factors/immunology
4.
Infect Immun ; 84(8): 2345-54, 2016 08.
Article in English | MEDLINE | ID: mdl-27271739

ABSTRACT

Burkholderia mallei is the causative agent of glanders, an incapacitating disease with high mortality rates in respiratory cases. Its endemicity and ineffective treatment options emphasize its public health threat and highlight the need for a vaccine. Live attenuated vaccines are considered the most viable vaccine strategy for Burkholderia, but single-gene-deletion mutants have not provided complete protection. In this study, we constructed the select-agent-excluded B. mallei ΔtonB Δhcp1 (CLH001) vaccine strain and investigated its ability to protect against acute respiratory glanders. Here we show that CLH001 is attenuated, safe, and effective at protecting against lethal B. mallei challenge. Intranasal administration of CLH001 to BALB/c and NOD SCID gamma (NSG) mice resulted in complete survival without detectable colonization or abnormal organ histopathology. Additionally, BALB/c mice intranasally immunized with CLH001 in a prime/boost regimen were fully protected against lethal challenge with the B. mallei lux (CSM001) wild-type strain.


Subject(s)
Bacterial Vaccines/immunology , Burkholderia mallei/immunology , Glanders/immunology , Vaccines, Attenuated/immunology , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Vaccines/genetics , Burkholderia mallei/genetics , Disease Models, Animal , Female , Glanders/mortality , Glanders/prevention & control , Immunization , Immunization, Secondary , Immunocompromised Host , Immunoglobulin G/immunology , Mice , Mutation , Vaccines, Attenuated/genetics
5.
Microb Pathog ; 78: 20-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25450887

ABSTRACT

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.Ā mallei in human and nonhuman primates has not been characterized. In this study, we examined the primary cellular immune response to B.Ā 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.Ā mallei elicited strong primary pro-inflammatory cytokines (IFN-ƎĀ³, TNF-α, IL-1Ɵ, and IL-6) equivalent to the levels of B.Ā pseudomallei in primary PBMC cultures of NHPs and humans. When we examined IL-1Ɵ and other cytokine responses by comparison to Escherichia coli LPS, African Green Monkeys appears to be most responsive to B.Ā 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.Ā mallei and B.Ā pseudomallei induced pro-inflammatory responses. Notably, the induced reporter activity with B.Ā mallei, B.Ā 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.Ā mallei infection, MyD88 targeted therapeutic intervention may be a successful strategy for therapy.


Subject(s)
Burkholderia mallei/immunology , Glanders/immunology , Immunity, Innate , Leukocytes, Mononuclear/immunology , Animals , Burkholderia mallei/physiology , Chlorocebus aethiops , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Glanders/genetics , Glanders/microbiology , Humans , Immunity, Cellular , Leukocytes, Mononuclear/microbiology , Macaca fascicularis , Macaca mulatta
6.
Nanomedicine ; 11(2): 447-56, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25194998

ABSTRACT

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


Subject(s)
Bacterial Vaccines/administration & dosage , Burkholderia mallei/drug effects , Glanders/drug therapy , Metal Nanoparticles/administration & dosage , Administration, Inhalation , Animals , Bacterial Vaccines/chemistry , Burkholderia mallei/pathogenicity , Disease Models, Animal , Glanders/immunology , Glanders/microbiology , Glycoconjugates/administration & dosage , Glycoconjugates/chemistry , Gold/chemistry , Humans , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Metal Nanoparticles/chemistry , Mice
7.
Infect Immun ; 80(1): 110-20, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22025508

ABSTRACT

The Gram-negative bacterium Burkholderia mallei causes rapidly fatal illness in equines and humans when contracted by inhalation and also has the potential to be used as a bioweapon. However, little is known regarding the early innate immune responses and signaling mechanisms required to generate protection from pneumonic B. mallei infection. We showed previously that monocyte chemoattractant protein 1 (MCP-1) was a critical chemokine required for protection from pneumonic B. mallei infection. We have now extended those studies to identify key Toll-like receptor (TLR) signaling pathways, effector cells, and cytokines required for protection from respiratory B. mallei infection. We found that MyD88-/- mice were highly susceptible to pulmonary challenge with B. mallei and had significantly short survival times, increased bacterial burdens, and severe organ pathology compared to wild-type mice. Notably, MyD88-/- mice had significantly fewer monocytes and dendritic cells (DCs) in lung tissues and airways than infected wild-type mice despite markedly higher bacterial burdens. The MyD88-/- mice were also completely unable to produce gamma interferon (IFN-ƎĀ³) at any time points following infection. In wild-type mice, NK cells were the primary cells producing IFN-ƎĀ³ in the lungs following B. mallei infection, while DCs and monocytes were the primary cellular sources of interleukin-12 (IL-12) production. Treatment with recombinant IFN-ƎĀ³ (rIFN-ƎĀ³) was able to significantly restore protective immunity in MyD88-/- mice. Thus, we conclude that the MyD88-dependent recruitment of inflammatory monocytes and DCs to the lungs and the local production of IL-12, followed by NK cell production of IFN-ƎĀ³, are the key initial cellular responses required for early protection from B. mallei infection.


Subject(s)
Burkholderia mallei/immunology , Dendritic Cells/immunology , Glanders/immunology , Monocytes/immunology , Myeloid Differentiation Factor 88/metabolism , Pneumonia, Bacterial/immunology , Animals , Bacterial Load , Disease Models, Animal , Female , Interferon-gamma/metabolism , Interleukin-12/metabolism , Lung/microbiology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Pneumonia, Bacterial/microbiology , Survival Analysis
8.
Eur J Immunol ; 41(1): 107-15, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21182082

ABSTRACT

Burkholderia pseudomallei causes melioidosis, a disease with a wide range of possible outcomes, from seroconversion and dormancy to sepsis and death. This spectrum of host-pathogen interactions poses challenging questions about the heterogeneity in immunity to B. pseudomallei. Models show protection to be dependent on CD4(+) cells and IFN-ƎĀ³, but little is known about specific target antigens. Having previously implicated the ABC transporter, LolC, in protective immunity, we here use epitope prediction, HLA-binding studies, HLA-transgenic models and studies of T cells from seropositive individuals to characterize HLA-restricted LolC responses. Immunized mice showed long-lasting memory to the protein, whereas predictive algorithms identified epitopes within LolC that subsequently demonstrated strong HLA class II binding. Immunization of HLA-DR transgenics with LolC stimulated T-cell responses to four of these epitopes. Furthermore, the responsiveness of HLA transgenics to LolC revealed a hierarchy supportive of HLA polymorphism-determined differential susceptibility. Seropositive human donors of diverse HLA class II types showed T-cell responses to LolC epitopes, which are conserved among Burkholderia species including Burkholderia cenocepacia, associated with life-threatening cepacia complex in cystic fibrosis patients and Burkholderia mallei, which causes glanders. These findings suggest a role for LolC epitopes in multiepitope vaccine design for melioidosis and related diseases.


Subject(s)
ATP-Binding Cassette Transporters/immunology , Burkholderia pseudomallei/immunology , CD4-Positive T-Lymphocytes/immunology , Melioidosis/immunology , Animals , Burkholderia cenocepacia/immunology , Burkholderia mallei/immunology , Female , Glanders/immunology , HLA Antigens/genetics , HLA Antigens/immunology , Histocompatibility Antigens/immunology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Polymorphism, Genetic/immunology
9.
Article in Russian | MEDLINE | ID: mdl-22442972

ABSTRACT

AIM: Isolation and composition comparison of extracellular antigens (ECA) of pathogenic burkholderiae in SDS-PAGE electrophoresis and their use for differentiation of these microorganisms by immunodiffusion methods. MATERIALS AND METHODS: 60 Burkholderia pseudomallei strains, 14 B. mallei strains, 5 B. thailandensis strains, 4 B. cepacia strains were studied. ECA was obtained by Liu technique on F-agar covered with cellophane. SDS-PAGE electrophoresis was performed in 10% gel by Laemmli, immunodiffusion reaction (IDR) in 1% agarose gel, IDR with live cultures, immunoelectrophoresis (IEPH) was performed by the standard techniques. Sera was obtained by immunizing rabbits with a mixture of ECA and incomplete Freund adjuvant. RESULTS: ECA spectra of typical strains of the studied burkholderiae strains after the electrophoresis in SDS-PAGE stained by silver have 8 - 9 major fractions. ECA electrophoregrams of B. pseudomallei and B. thailandensis had a high similarity. ECA analysis by IDR with antisera against ECA revealed maximum number of cross-reactive ECA (3) between B. pseudomallei B. thailandensis. These strains had only a single crossreactive ECA to B. mallei strain. IDR with live culture and antisera to B. thailandensis ECA revealed ECA in all the B. pseudomallei, B. thailandensis strains and did not reveal those in B. mallei strains. Analysis of electrophoregram obtained with IEPH method of pathogenic burkholderiae ECA with antisera to ECA revealed differences of the composition sufficient for their differentiation. CONCLUSION: The differences of ECA composition revealed by immunodiffusion methods allowed to develop additional approaches of differentiation ofglanders and melioidosis pathogenic agents.


Subject(s)
Antigens, Bacterial/analysis , Bacterial Typing Techniques/methods , Burkholderia mallei/isolation & purification , Burkholderia pseudomallei/isolation & purification , Burkholderia/isolation & purification , Glanders/diagnosis , Melioidosis/diagnosis , Animals , Antigens, Bacterial/immunology , Burkholderia/classification , Burkholderia/immunology , Burkholderia/pathogenicity , Burkholderia mallei/immunology , Burkholderia mallei/pathogenicity , Burkholderia pseudomallei/immunology , Culture Media, Conditioned/chemistry , Electrophoresis, Polyacrylamide Gel , Freund's Adjuvant/immunology , Gels , Glanders/immunology , Glanders/microbiology , Horses , Immune Sera/immunology , Immunoelectrophoresis , Melioidosis/immunology , Melioidosis/microbiology , Rabbits , Sepharose
10.
Infect Immun ; 77(4): 1579-88, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19179415

ABSTRACT

Burkholderia mallei and B. pseudomallei are important human pathogens and cause the diseases glanders and melioidosis, respectively. Both organisms are highly infectious when inhaled and are inherently resistant to many antimicrobials, thus making it difficult to treat pneumonic Burkholderia infections. We investigated whether it was possible to achieve rapid protection against inhaled Burkholderia infection by using inhaled immunotherapy. For this purpose, cationic liposome DNA complexes (CLDC), which are potent activators of innate immunity, were used to elicit the activation of pulmonary innate immune responses. We found that mucosal CLDC administration before or shortly after bacterial challenge could generate complete or nearly complete protection from inhalational challenge with 100% lethal doses of B. mallei and B. pseudomallei. Protection was found to be dependent on the CLDC-mediated induction of gamma interferon responses in lung tissues and was partially dependent on the activation of NK cells. However, CLDC-mediated protection was not dependent on the induction of inducible nitric oxide synthase, as assessed by depletion studies. We concluded that the potent local activation of innate immune responses in the lung could be used to elicit rapid and nonspecific protection from aerosol exposure to both B. mallei and B. pseudomallei.


Subject(s)
Burkholderia Infections , Burkholderia mallei/pathogenicity , Burkholderia pseudomallei/pathogenicity , Immunotherapy/methods , Lung Diseases , Administration, Inhalation , Animals , Burkholderia Infections/immunology , Burkholderia Infections/microbiology , Burkholderia Infections/prevention & control , Burkholderia Infections/therapy , Cations , Cell Line , DNA, Bacterial/administration & dosage , DNA, Bacterial/genetics , DNA, Bacterial/immunology , Escherichia coli/genetics , Glanders/immunology , Glanders/microbiology , Glanders/prevention & control , Glanders/therapy , Humans , Interferon-gamma/biosynthesis , Liposomes/administration & dosage , Liposomes/immunology , Lung Diseases/immunology , Lung Diseases/microbiology , Lung Diseases/prevention & control , Lung Diseases/therapy , Macrophages, Alveolar/microbiology , Melioidosis/immunology , Melioidosis/microbiology , Melioidosis/prevention & control , Melioidosis/therapy , Mice , Mice, Inbred BALB C , Plasmids/administration & dosage , Plasmids/genetics , Plasmids/immunology
11.
PLoS One ; 14(4): e0214963, 2019.
Article in English | MEDLINE | ID: mdl-30951554

ABSTRACT

Glanders is a zoonotic contagious disease of equids caused by Burkholderia (B.) mallei. Serodiagnosis of the disease is challenging because of false-positive and false-negative test results. The accuracy of the complement fixation test (CFT) which is prescribed for international trade by the World Organisation for Animal Health (OIE), five ELISAs and a Western blot (WB) were compared for serodiagnosis of glanders using sera from 3,000 glanders-free and 254 glanderous equids. Four ELISA tests are based on recombinant antigens (TssA, TssB, BimA and Hcp1), the IDVet ELISA is based on a semi-purified fraction of B. mallei and WB makes use of a purified LPS-containing B. mallei-antigen. Sensitivity and specificity of tests were estimated using cut-off values recommended by the test developers. The WB and all ELISAs, except BimA, were significantly more specific than the CFT. ELISAs based on TssA, TssB, and BimA antigens had significantly lower sensitivity compared to CFT while the sensitivities of the Hcp1-ELISA, the IDVet-ELISA and the WB did not differ significantly from that of the CFT. Given their comparable sensitivities and specificities, the CFT (98.0%, 96.4%), the WB (96.8%, 99.4%), the Hcp1-ELISA (95.3%, 99.6%) and the IDVet-ELISA (92.5%, 99.5%) should be further developed to meet OIE requirements.


Subject(s)
Antigens, Bacterial/blood , Blotting, Western , Burkholderia mallei , Complement Fixation Tests , Glanders/blood , Horses/blood , Animals , Antigens, Bacterial/immunology , Enzyme-Linked Immunosorbent Assay , Glanders/diagnosis , Glanders/immunology , Glanders/microbiology , Horses/immunology , Horses/microbiology
12.
PLoS Negl Trop Dis ; 13(7): e0007578, 2019 07.
Article in English | MEDLINE | ID: mdl-31306423

ABSTRACT

BACKGROUND: Glanders caused by Burkholderia mallei is a re-emerging zoonotic disease affecting solipeds and humans. Furthermore, B. mallei is genetically related to B. pseudomallei, which is the causative agent of melioidosis. Both facultative intracellular bacteria are classified as tier 1 select biothreat agents. Our previous study with a B. mallei ΔtonB Δhcp1 (CLH001) live-attenuated vaccine demonstrated that it is attenuated, safe and protective against B. mallei wild-type strains in the susceptible BALB/c mouse model. METHODOLOGY/PRINCIPAL FINDING: In our current work, we evaluated the protective efficacy of CLH001 against glanders and melioidosis in the more disease-resistant C57BL/6 mouse strain. The humoral as well as cellular immune responses were also examined. We found that CLH001-immunized mice showed 100% survival against intranasal and aerosol challenge with B. mallei ATCC 23344. Moreover, this vaccine also afforded significant cross-protection against B. pseudomallei K96243, with low level bacterial burden detected in organs. Immunization with a prime and boost regimen of CLH001 induced significantly greater levels of total and subclasses of IgG, and generated antigen-specific splenocyte production of IFN-ƎĀ³ and IL-17A. Interestingly, protection induced by CLH001 is primarily dependent on humoral immunity, while CD4+ and CD8+ T cells played a less critical protective role. CONCLUSIONS/SIGNIFICANCE: Our data indicate that CLH001 serves as an effective live attenuated vaccine to prevent glanders and melioidosis. The quantity and quality of antibody responses as well as improving cell-mediated immune responses following vaccination need to be further investigated prior to advancement to preclinical studies.


Subject(s)
Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Burkholderia mallei/immunology , Glanders/immunology , Immunization , Melioidosis/immunology , Membrane Proteins/immunology , Vaccines, Attenuated/immunology , Animals , Antibodies, Bacterial/immunology , Bacterial Proteins/genetics , Burkholderia mallei/genetics , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Glanders/microbiology , Glanders/prevention & control , Humans , Immunity, Humoral , Melioidosis/microbiology , Melioidosis/prevention & control , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Vaccination , Virulence Factors/genetics , Virulence Factors/immunology
13.
BMC Immunol ; 9: 55, 2008 Sep 29.
Article in English | MEDLINE | ID: mdl-18823549

ABSTRACT

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


Subject(s)
Bacterial Vaccines/immunology , Burkholderia mallei/immunology , Glanders/immunology , Immunity, Active , Vaccines, Inactivated/immunology , Animals , Antibodies, Blocking/immunology , B-Lymphocytes/immunology , Bacterial Vaccines/administration & dosage , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Models, Animal , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Vaccination , Vaccines, Inactivated/administration & dosage
14.
BMC Microbiol ; 7: 19, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17362501

ABSTRACT

BACKGROUND: The bacterial biothreat agents Burkholderia mallei and Burkholderia pseudomallei are the cause of glanders and melioidosis, respectively. Genomic and epidemiological studies have shown that B. mallei is a recently emerged, host restricted clone of B. pseudomallei. RESULTS: Using bacteriophage-mediated immunoscreening we identified genes expressed in vivo during experimental equine glanders infection. A family of immunodominant antigens were identified that share protein domain architectures with hemagglutinins and invasins. These have been designated Burkholderia Hep_Hag autotransporter (BuHA) proteins. A total of 110/207 positive clones (53%) of a B. mallei expression library screened with sera from two infected horses belonged to this family. This contrasted with 6/189 positive clones (3%) of a B. pseudomallei expression library screened with serum from 21 patients with culture-proven melioidosis. CONCLUSION: Members of the BuHA proteins are found in other Gram-negative bacteria and have been shown to have important roles related to virulence. Compared with other bacterial species, the genomes of both B. mallei and B. pseudomallei contain a relative abundance of this family of proteins. The domain structures of these proteins suggest that they function as multimeric surface proteins that modulate interactions of the cell with the host and environment. Their effect on the cellular immune response to B. mallei and their potential as diagnostics for glanders requires further study.


Subject(s)
Antibodies, Bacterial/biosynthesis , Bacterial Proteins/immunology , Burkholderia mallei/immunology , Burkholderia pseudomallei/immunology , Glanders/immunology , Melioidosis/immunology , Adolescent , Adult , Aged , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/biosynthesis , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/biosynthesis , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Base Sequence , Burkholderia mallei/genetics , Burkholderia pseudomallei/genetics , Child , Female , Gene Library , Glanders/diagnosis , Glanders/microbiology , Hemagglutinins/immunology , Horses , Humans , Male , Melioidosis/blood , Melioidosis/microbiology , Middle Aged , Molecular Sequence Data , Virulence Factors/biosynthesis , Virulence Factors/genetics , Virulence Factors/immunology
15.
Diagn Microbiol Infect Dis ; 59(2): 137-47, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17908615

ABSTRACT

We examined, by enzyme-linked immunosorbent assay and Western blot analysis, the host immune response to 2 heat-shock proteins (hsps) in a patient and mice previously infected with Burkholderia mallei. The patient was the first reported human glanders case in 50 years in the United States. The expression of the groEL and dnaK operons appeared to be dependent upon a sigma(32) RNA polymerase as suggested by conserved heat-shock promoter sequences, and the groESL operon may be negatively regulated by a controlling invert repeat of chaperone expression (CIRCE) site. In the antisera, the GroEL protein was found to be more immunoreactive than the DnaK protein in both a human patient and mice previously infected with B. mallei. Examination of the supernatant of a growing culture of B. mallei showed that more GroEL protein than DnaK protein was released from the cell. This may occur similarly within an infected host causing an elevated host immune response to the B. mallei hsps.


Subject(s)
Antibodies, Bacterial/blood , Burkholderia mallei/immunology , Chaperonin 60/immunology , Glanders/immunology , HSP70 Heat-Shock Proteins/immunology , Immunoglobulin G/blood , Animals , Burkholderia mallei/genetics , Burkholderia mallei/metabolism , Chaperonin 60/genetics , Chaperonin 60/metabolism , Gene Expression Regulation, Bacterial , Glanders/microbiology , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Operon , Sequence Analysis, DNA
16.
FEMS Microbiol Lett ; 277(2): 115-22, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18031330

ABSTRACT

Burkholderia mallei, the etiologic agent of the disease known as glanders, is primarily a disease affecting horses and is transmitted to humans by direct contact with infected animals. The use of B. mallei as a biological weapon has been reported and currently, there is no vaccine available for either humans or animals. Despite the history and highly infective nature of B. mallei, as well as its potential use as a bio-weapon, B. mallei research to understand the pathogenesis and the host responses to infection remains limited. Therefore, this minireview will focus on current efforts to elucidate B. mallei virulence, the associated host immune responses elicited during infection and discuss the feasibility of vaccine development.


Subject(s)
Burkholderia mallei/immunology , Burkholderia mallei/physiology , Glanders/immunology , Glanders/microbiology , Animals , Biological Warfare Agents , Horses , Humans , Virulence
17.
Exp Biol Med (Maywood) ; 232(4): 550-6, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17392491

ABSTRACT

Much effort has been devoted to the development of mouse monoclonal antibodies that react specifically with Burkholderia mallei and Burkholderia pseudomallei for diagnostic and/or therapeutic purposes. Our present study focused on the screening of a phage-displayed nonimmune human single-chain Fv (scFv) antibody library against heat-killed B. mallei and B. pseudomallei for the generation of human scFv antibodies specific to the two pathogenic species of bacteria. Using two different panning procedures, we obtained seven different scFv phage antibodies that interacted with the heat-killed whole bacterial cells of B. mallei and B. pseudomallei. Our results demonstrate that panning of a human scFv antibody library against heat-killed whole bacterial cells may provide a valuable strategy for developing human monoclonal antibodies against the highly pathogenic bacteria.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Burkholderia mallei/immunology , Burkholderia pseudomallei/immunology , Immunoglobulin Variable Region/immunology , Animals , Antibodies, Bacterial/genetics , Antibodies, Monoclonal/genetics , Cloning, Molecular , Glanders/diagnosis , Glanders/immunology , Humans , Immunoglobulin Variable Region/genetics , Melioidosis/diagnosis , Melioidosis/immunology , Mice
18.
Article in Russian | MEDLINE | ID: mdl-17672140

ABSTRACT

Test-system using index of phagocytosis of noncapsulated mutant loaded by one of the several capsular antigenic complexes was developed and used for screening for both immunogenic and protective capsular antigens of B. mallei. Direct correlation between index of phagocytosis, level of delayed-type hypersensivity, and protective effect of capsular antigens has been shown on the model of experimental melioidosis in susceptible white mice, guinea pigs and white rats. Obtained results let to use the developed test-system for initial selection of B. mallei protective capsular antigens and their further study as potential components of preparations for specific prophylaxis of glanders and melioidosis.


Subject(s)
Bacterial Capsules/immunology , Burkholderia mallei/immunology , Glanders/immunology , Glanders/prevention & control , Immunization , Animals , Bacterial Capsules/administration & dosage , Bacterial Capsules/isolation & purification , Bioterrorism , Guinea Pigs , Hypersensitivity, Delayed , Injections, Intramuscular , Injections, Subcutaneous , Macrophages, Peritoneal/immunology , Mice , Phagocytosis , Rats
19.
Vaccine ; 35(44): 5981-5989, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28336210

ABSTRACT

B. pseudomallei is the cause of melioidosis, a serious an often fatal disease of humans and animals. The closely related bacterium B. mallei, which cases glanders, is considered to be a clonal derivative of B. pseudomallei. Both B. pseudomallei and B. mallei were evaluated by the United States and the former USSR as potential bioweapons. Much of the effort to devise biodefence vaccines in the past decade has been directed towards the identification and formulation of sub-unit vaccines which could protect against both melioidosis and glanders. A wide range of proteins and polysaccharides have been identified which protective immunity in mice. In this review we highlight the significant progress that has been made in developing glycoconjugates as sub-unit vaccines. We also consider some of the important the criteria for licensing, including the suitability of the "animal rule" for assessing vaccine efficacy, the protection required from a vaccine and the how correlates of protection will be identified. Vaccines developed for biodefence purposes could also be used in regions of the world where naturally occurring disease is endemic.


Subject(s)
Bacterial Vaccines/immunology , Burkholderia mallei/immunology , Burkholderia pseudomallei/immunology , Glanders/immunology , Glanders/prevention & control , Melioidosis/immunology , Melioidosis/prevention & control , Animals , Clinical Trials as Topic , Humans
20.
J Med Biogr ; 25(2): 98-105, 2017 May.
Article in English | MEDLINE | ID: mdl-26307410

ABSTRACT

The anti-globulin test was described in 1945, and ever since has been synonymous with the lead author, Robin Coombs, a young veterinary surgeon, at that time embarking on a career in immunological research. This was marked by a number of important contributions in the field, including the description and categorisation of hypersensitivity reactions, co-authored with Philip Gell. Together they wrote the classical text, Clinical Aspects of Immunology, which has been updated and republished over the ensuing 50 years. Although Robin Coombs is best remembered for his contributions to medical immunology, he made a number of significant early advances in the field of veterinary immunology.


Subject(s)
Coombs Test/history , Globulins , Veterinary Medicine/history , Allergy and Immunology/history , Animals , Glanders/immunology , History, 20th Century , Horses , Humans , Surgeons , Writing
SELECTION OF CITATIONS
SEARCH DETAIL