Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Microbes Infect ; 9(8): 997-1002, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17556003

ABSTRACT

Yersinia pestis and the enteropathogenic Yersinia pseudotuberculosis and Yersinia enterocolitica share the virulence-antigen LcrV. Previously, using reverse genetics we have proven that LcrV contributes to the virulence of Y. enterocolitica serotype O:8 by inducing IL-10 via Toll-like receptor 2 (TLR2). However, both the ability of Y. pestis LcrV to activate TLR2 and a possible role of TLR2-dependent IL-10 induction by LcrV in Y. pestis are not yet known. To eliminate interference from additional protein sequences, we produced LcrVs without affinity tags from Y. pestis and from Y. enterocolitica O:8 (LcrVO:8). LcrVO:8 was much more potent in TLR2-activity than Y. pestis LcrV. To analyse the role of TLR2 in plague, we infected both wild-type and TLR2-/- mice subcutaneously with Y. pestis GB. While TLR2-/- mice exhibited lower blood levels of IL-10 (day 2 post-infection) and of the pro-inflammatory cytokines TNF-alpha, IFN-gamma and MCP-1 (day 4) than wild-type mice, there was no significant difference in survival. The low TLR2-activity of Y. pestis LcrV and associated cytokine expression might explain why - in contrast to Y. enterocolitica O:8 infection - TLR2-deficient mice are not more resistant than wild-type mice in a bubonic plague model.


Subject(s)
Antigens, Bacterial/metabolism , Macrophages/immunology , Pore Forming Cytotoxic Proteins/metabolism , Toll-Like Receptor 2/metabolism , Yersinia pestis/pathogenicity , Animals , Antigens, Bacterial/genetics , Cell Line , Cytokines/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Plague/immunology , Plague/microbiology , Plague/mortality , Plague/physiopathology , Pore Forming Cytotoxic Proteins/genetics , Specific Pathogen-Free Organisms , Toll-Like Receptor 2/genetics , Virulence , Yersinia pestis/genetics , Yersinia pestis/metabolism
2.
Infect Immun ; 74(6): 3687-91, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16714605

ABSTRACT

The identification of Yersinia pestis as a potential bioterrorism agent and the emergence of antibiotic-resistant strains have highlighted the need for improved vaccines and treatments for plague. The aim of this study was to evaluate the potential for ATP-binding cassette (ABC) transporter proteins to be exploited as novel vaccines against plague. Western blotting of ABC transporter proteins using sera from rabbits immunized with killed whole Y. pestis cells or human convalescent-phase sera identified four immunologically reactive proteins: OppA, PstS, YrbD, and PiuA. Mice immunized with these proteins developed antibody to the immunogen. When the immunized mice were challenged with Y. pestis, the OppA-immunized mice showed an increased time to death compared to other groups, and protection appeared to correlate with the level of immunoglobulin G antibody to OppA.


Subject(s)
Bacterial Proteins/immunology , Carrier Proteins/immunology , Lipoproteins/immunology , Plague Vaccine/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Bacterial/blood , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Immune Sera/immunology , Immunization , Immunoglobulin G/classification , Mice , Mice, Inbred BALB C , Plague/prevention & control , Yersinia pestis/immunology
3.
Infect Immun ; 74(5): 3068-70, 2006 May.
Article in English | MEDLINE | ID: mdl-16622253

ABSTRACT

Intratracheal delivery of aerosolized monoclonal antibodies with specificity for Yersinia pestis LcrV and F1 antigens protected mice in a model of pneumonic plague. These data support the utility of inhaled antibodies as a fast-acting postexposure treatment for plague.


Subject(s)
Antibodies, Bacterial/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Plague/prevention & control , Yersinia pestis/immunology , Aerosols , Animals , Female , Mice , Mice, Inbred BALB C , Pore Forming Cytotoxic Proteins
4.
Vaccine ; 24(20): 4433-9, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16546306

ABSTRACT

Protection against virulent plague challenge by the parenteral and aerosol routes was afforded by a single administration of microencapsulated Caf1 and LcrV antigens from Yersinia pestis in BALB/c mice. Recombinant Caf1 and LcrV were individually encapsulated in polymeric microspheres, to the surface of which additional antigen was adsorbed. The microspheres containing either Caf1 or LcrV were blended and used to immunise mice on a single occasion, by either the intra-nasal or intra-muscular route. Both routes of immunisation induced systemic and local immune responses, with high levels of serum IgG being developed in response to both vaccine antigens. In Elispot assays, secretion of cytokines by spleen and draining lymph node cells was demonstrated, revealing activation of both Th1 and Th2 associated cytokines; and spleen cells from animals immunised by either route were found to proliferate in vitro in response to both vaccine antigens. Virulent challenge experiments demonstrated that non-invasive immunisation by intra-nasal instillation can provide strong systemic and local immune responses and protect against high level challenge. Microencapsulation of these vaccine antigens has the added advantage that controlled release of the antigens occurs in vivo, so that protective immunity can be induced after only a single immunising dose.


Subject(s)
Bacterial Vaccines/administration & dosage , Plague/prevention & control , Administration, Intranasal , Bacterial Vaccines/immunology , Bronchoalveolar Lavage Fluid , Cell Proliferation , Drug Compounding , Humans , Immunoglobulin G/blood , Injections, Intramuscular , Plague/immunology , Yersinia pestis/immunology
5.
Infect Immun ; 74(3): 1706-11, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16495542

ABSTRACT

Burkholderia pseudomallei, the causative agent of melioidosis, is a gram-negative bacterium which can cause either chronic infections or acute lethal sepsis in infected individuals. The disease is endemic in Southeast Asia and northern Australia, but little is known about the mechanisms of protective immunity to the bacterium. In this study, we have developed a procedure to utilize dendritic cells in combination with CpG oligodeoxynucleotides as a vaccine delivery vector to induce protective immune responses to various strains of B. pseudomallei. Our results show that strong cell-mediated immune responses were generated, while antibody responses, although low, were detectable. Upon virulent challenge with B. pseudomallei strain K96243, NCTC 4845, or 576, animals immunized with dendritic cells that were pulsed with heat-killed K96243 and matured in the presence of CpG 1826 showed significant levels of protection. These results show that a vaccine strategy that actively targets dendritic cells can evoke protective immune responses.


Subject(s)
Antibodies, Bacterial/biosynthesis , Burkholderia pseudomallei , DNA/administration & dosage , Dendritic Cells/immunology , Melioidosis/prevention & control , Animals , Burkholderia pseudomallei/immunology , Cohort Studies , DNA/immunology , Immunization , Melioidosis/immunology , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides
6.
Vaccine ; 24(11): 1736-43, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16278038

ABSTRACT

Certain toll-like receptor (TLR) agonists, e.g. CpG DNA, can be used as potent vaccine 'adjuvants'. It is known that some sequences of single stranded (ss) RNA stimulate proinflammatory and antiviral responses following interaction with TLR 7 and 8. We have encapsulated ovalbumin (OVA) in the presence and absence of polyuridylic acid (poly-U) inside polylactide microparticles. In comparison to microparticles containing only OVA, bulk cultures of bone marrow-derived plasmacytoid and myeloid dendritic cells produced more (P<0.05) IL-12 and interferon (IFN)-alpha when stimulated with microparticles containing OVA and poly-U. Subcutaneous injection of comicroencapsulated OVA and poly-U resulted in statistically elevated levels of serum anti-OVA IgG1 (P<0.05 versus naïve mice). Conversely, anti-OVA IgG1 levels in C57 BL6 mice immunised with OVA loaded microparticles (without RNA) were statistically indifferent to naïve animals. Furthermore, injection of coencapsulated OVA and poly-U resulted in (P<0.05) greater numbers of OVA specific IFN-gamma secreting T-cells as compared with mice injected with OVA loaded microparticles. A similar trend was seen in mice immunised with OVA loaded microparticles decorated with CpG or solutions of admixed OVA and CpG (P<0.05). These data demonstrate, for the first time, that appropriately formulated ssRNA can act as a potent adjuvant and modulator of adaptive immunological responses.


Subject(s)
Adjuvants, Immunologic , Ovalbumin/immunology , Poly U/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Antibodies/blood , Dendritic Cells/immunology , Drug Compounding , Female , Immunoglobulin G/blood , Interferon-alpha/biosynthesis , Interleukin-12/biosynthesis , Mice , Mice, Inbred C57BL , Microspheres , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Ovalbumin/administration & dosage , Poly U/administration & dosage , T-Lymphocytes/immunology , Toll-Like Receptors
7.
Infect Immun ; 73(9): 5945-51, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16113315

ABSTRACT

Burkholderia pseudomallei, the causative agent of melioidosis, is a gram-negative bacillus endemic to areas of southeast Asia and northern Australia. Presently, there is no licensed vaccine for B. pseudomallei and the organism is refractive to antibiotic therapy. The bacterium is known to survive and multiply inside both phagocytic and nonphagocytic host cells and may be able to spread directly from cell to cell. Current vaccine delivery systems are unlikely to induce the correct immune effectors to stimulate a protective response to the organism. In this study, we have developed a procedure to utilize dendritic cells as a vaccine delivery vector to induce cell-mediated immune responses to B. pseudomallei. Dendritic cells were produced by culturing murine bone marrow progenitor cells in medium containing granulocyte-macrophage colony-stimulating factor and tumor necrosis factor alpha. Purified dendritic cells were pulsed with heat-killed whole-cell B. pseudomallei and used to immunize syngeneic mice. Strong cellular immune responses were elicited by this immunization method, although antibody responses were low. Booster immunizations of either a second dose of dendritic cells or heat-killed B. pseudomallei were administered to increase the immune response. Immunized animals were challenged with fully virulent B. pseudomallei, and protection was demonstrated in those with strong humoral and cell-mediated immunity. These results indicate the importance of both cell-mediated and humoral immune mechanisms in protection against intracellular pathogens.


Subject(s)
Antibodies, Bacterial/biosynthesis , Burkholderia pseudomallei/immunology , Dendritic Cells/immunology , Melioidosis/immunology , Melioidosis/microbiology , Adoptive Transfer , Animals , Antigen Presentation/immunology , Cells, Cultured , Dendritic Cells/transplantation , Immunity, Cellular/immunology , Melioidosis/prevention & control , Mice , Mice, Inbred BALB C , Peptide Fragments/immunology
8.
Infect Immun ; 73(4): 2005-11, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15784541

ABSTRACT

Live, attenuated bacteria are effective vectors for heterologous antigen delivery. However, loss of heterologous gene-bearing plasmids is problematic, and antibiotics and their resistance genes are not desirable for in vivo DNA vaccine delivery due to biosafety and regulatory concerns. To solve this problem, we engineered the first vaccine delivery strain that has no requirement for antibiotics or other selectable marker genes to maintain the recombinant plasmid. This model strain of Salmonella enterica serovar Typhimurium, SLDAPD, uses operator-repressor titration (ORT) technology, which requires only the short, nonexpressed lacO sequence for selection and maintenance. SLDAPD, recovered from the spleens and Peyer's patches of mice following oral inoculation, was shown to maintain a plasmid that, in contrast, was lost from parental strain SL3261. We also demonstrated successful application of this technology to vaccine development, since SLDAPD carrying a plasmid without an antibiotic resistance gene that expressed the Yersinia pestis F1 antigen was as efficacious in protecting vaccinated mice against plague as the parental SL3261 strain carrying an antibiotic-selected version of this plasmid. Protection of mice against plague by immunization with Salmonella expressing F1 has previously required two or more doses; here we demonstrated for the first time protective immunity after a single oral immunization. This technology can easily be used to convert any suitable attenuated strain to an antibiotic-free ORT strain for recombinant protein vaccine delivery in humans.


Subject(s)
Drug Resistance/genetics , Plague Vaccine/immunology , Plasmids , Salmonella typhimurium/genetics , Vaccines, Synthetic/immunology , Animals , Female , Mice , Mice, Inbred BALB C , Salmonella typhimurium/immunology
9.
Microb Pathog ; 37(4): 177-84, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15458778

ABSTRACT

The Caf1 and LcrV sub-unit vaccine for plague has been shown to be highly protective against challenge with virulent Yersinia pestis in a mouse model. Production of large amounts of IgG1 in response to the vaccine correlates with protection against aerosol and parenteral infection. In this study the effect of genetic mutation in the immune system on protection was addressed. Stat 6(-/-) mice which are unable to utilise the type 2 cytokines IL-4 and IL-13 and so should have reduced IgG1 responses were utilised in order to determine whether an immune system biased towards the type 1 axis could mount an effective response to the vaccine. Conversely in the Stat 4(-/-) mouse model, IL-12 and interferon-gamma-mediated immune mechanisms are inactive and the immune response should be biased towards the type 2 axis. Serum antibody responses to vaccination in both the knockout strains and their wild type controls revealed little difference in levels of IgG and isotype profiles. Elispot analysis of cytokine production at the single cell level did however reveal a functional defect in the Stat 4(-/-) mice which had low levels of IFN-gamma producing cells. Following virulent challenge, the Stat 6(-/-) mice showed high levels of protection, while the Stat 4(-/-) mice were poorly protected, indicating a fundamental defect in their immune systems which could not be overcome even by the passive transfer of CD4(+) cells from immunised BALB/c donors. It appears therefore that type 1 immune mechanisms, activated following Stat 4 phosphorylation, are essential in protection against plague.


Subject(s)
Antibodies, Bacterial/biosynthesis , DNA-Binding Proteins/metabolism , Plague Vaccine/immunology , Plague/immunology , Trans-Activators/metabolism , Yersinia pestis/immunology , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , Bacterial Proteins/administration & dosage , Bacterial Proteins/immunology , DNA-Binding Proteins/genetics , Immunoglobulin G/blood , Immunoglobulin Isotypes/blood , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Lymphocyte Activation , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/administration & dosage , Molecular Chaperones/immunology , Plague Vaccine/administration & dosage , Pore Forming Cytotoxic Proteins , STAT4 Transcription Factor , STAT6 Transcription Factor , Trans-Activators/genetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
10.
Nature ; 430(6998): 417, 2004 Jul 22.
Article in English | MEDLINE | ID: mdl-15272490

ABSTRACT

Mecsas and colleagues suggest that a deficiency in the chemokine receptor CCR5 in humans is unlikely to confer protection against plague, based on their study of Yersinia pestis infection in Ccr5-deficient mice. They were testing the hypothesis that a mutation in the CCR5 gene, frequently found in Caucasians, may have been selected for in the past because it provided protection against (bubonic) plague; the mutation, called CCR5Delta32, is characterized by a 32-base-pair deletion. We have also tested this hypothesis by using Y. pestis infection in mice and, in addition, we have done phagocytosis experiments with macrophages from wild-type and Ccr5-deficient mice. Although, like Mecsas et al., we did not see any difference in the survival of the two groups of mice, we did find that there was a significantly reduced uptake of Y. pestis by Ccr5-deficient macrophages in vitro. Our results indicate that the role of Ccr5 in Y. pestis infection may therefore be more complex than previously thought.


Subject(s)
Evolution, Molecular , Plague/genetics , Plague/metabolism , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Animals , Humans , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , Phagocytosis , Plague/microbiology , Receptors, CCR5/deficiency , Sequence Deletion/genetics , Survival Rate , Virulence , White People/genetics , Yersinia pestis/pathogenicity , Yersinia pestis/physiology
11.
J Med Virol ; 67(3): 384-93, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12116032

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) replicates in lymphoid tissues following peripheral inoculation and a high titre viraemia develops. Encephalitis develops after the virus enters the central nervous system from the blood, with the earliest neuronal involvement being via the olfactory nerve. Following aerosol challenge with virulent VEEV, the virus is thought to replicate in the nasal mucosa and there could be direct entry into the olfactory nerve via infected neuroepithelial cells. Protection from VEEV infection is believed to be primarily mediated by virus specific antibody. The correlation between protection and neutralising serum antibody titres is, however, inconsistent when the virulent virus is administered by the airborne route. This study demonstrates a link between antibody in serum and the nasal mucosa and protection by means of passive immunisation studies. Intra-nasal administration of antibody increased protection against airborne virus in Balb/c mice. Vaccination of mu MT strain mice that do not have functional B cells and cannot produce antibody revealed normal proliferation of spleen cells in vitro and robust cytokine production. Aerosol challenge of mu MT mice demonstrated that complete protection was only achieved when passive immunisation with antibody was supplemented with active immunisation with the TC-83 vaccine strain of the virus. This implies that cell-mediated immune functions are required for protection against airborne challenge with virulent VEEV.


Subject(s)
Antibodies, Viral/blood , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/prevention & control , Nasal Mucosa/immunology , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Antibody Specificity , Cell Line , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/virology , Immunity, Cellular , Immunity, Mucosal , Immunization, Passive , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/cytology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...