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










Publication year range
1.
mBio ; 15(3): e0003724, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38334423

ABSTRACT

We examined the relationship between the association of a vaccine antigen with immune cells in secondary lymphoid organs shortly after immunization and the resulting neutralizing antibody response induced by that antigen using three antigenic forms of anthrax protective antigen (PA) that induce qualitatively different antibody responses. The three PA forms used were wild-type PA, which binds to anthrax toxin receptors and elicits a robust antibody response that includes both neutralizing and non-neutralizing antibodies; a receptor-binding-deficient (RBD) mutant form of PA, which does not bind cellular receptors and elicits only barely detectable antibody responses; and an engineered chimeric form of PA, which binds cholera toxin receptors and elicits a robust total antibody response but a poor neutralizing antibody response. We found that both wild-type PA and the PA chimera associated with immune cells in secondary lymphoid organs after immunization, but the RBD mutant PA exhibited minimal association, revealing a relationship between antigen binding to toxin receptors on immune cells after immunization and subsequent antibody responses. A portion of wild-type PA that bound to immune cells was cell surface-associated and maintained its native conformation. Much lower amounts of conformationally intact PA chimera were associated with immune cells after immunization, correlating with the lower neutralizing antibody response elicited by the PA chimera. Thus, binding of an antigen to receptors on immune cells in secondary lymphoid organs after immunization and maintenance of conformational integrity of the cell-associated antigen help dictate the magnitude of the resulting neutralizing antibody response, but not necessarily the total antibody response.IMPORTANCEMany vaccines protect by the induction of antibodies that neutralize the action of the pathogen. Here, we followed the fate of three antigenic forms of a vaccine antigen in secondary lymphoid organs after immunization to investigate events leading to a robust neutralizing antibody response. We found that the magnitude of the neutralizing antibody response, but not the total antibody response, correlates with the levels of conformationally intact antigen associated with immune cells in secondary lymphoid organs after primary immunization. We believe that these results provide important insights into the genesis of neutralizing antibody responses induced by vaccine antigens and may have implications for vaccine design.


Subject(s)
Anthrax Vaccines , Bacillus anthracis , Antibodies, Neutralizing , Antibody Formation , Antigens, Bacterial/metabolism , Vaccination , Immunization , Antibodies, Bacterial , Bacillus anthracis/metabolism
2.
Vaccine ; 40(32): 4318-4321, 2022 07 30.
Article in English | MEDLINE | ID: mdl-35710508

ABSTRACT

Anthrax protective antigen (PA), the receptor-binding component of anthrax toxin, elicits toxin-neutralizing antibodies which provide protection against anthrax disease. PA binds to two mammalian receptors, capillary morphogenesis protein-2 (CMG2) and tumor endothelial marker-8 (TEM8). We previously observed that binding of PA to its receptors plays a role in eliciting a strong toxin-neutralizing antibody response. In this study, we examined the roles that individual receptors play in mediating the toxin-neutralizing antibody response. Mice immunized with PA that binds preferentially to CMG2 elicited a toxin-neutralizing antibody response similar to that elicited by wild-type PA, whereas the antibody response elicited by PA that binds preferentially to TEM8 was significantly lower. Also, the toxin-neutralizing antibody response elicited by wild-type PA in CMG2-null mice was found to be significantly lower than that induced in CMG2-sufficient mice, further supporting a predominant role for the CMG2 receptor in mediating a protective antibody response to PA.


Subject(s)
Anthrax , Receptors, Peptide , Animals , Antibodies, Neutralizing , Antigens, Bacterial , Bacterial Toxins , Mammals/metabolism , Mice , Morphogenesis , Receptors, Peptide/genetics , Receptors, Peptide/metabolism
3.
PLoS One ; 13(3): e0195342, 2018.
Article in English | MEDLINE | ID: mdl-29596507

ABSTRACT

Staphylococcus aureus bacteria are able to grow in a planktonic state that is associated with acute infections and in biofilms that are associated with chronic infections. Acute infections, such as skin infections, are often self-limiting. However, chronic infections, such as implant infections, can be difficult to clear and may require surgical intervention. The host immune response may contribute to the different outcomes often associated with these two disease types. We used proteomic arrays and two murine models for an initial, descriptive characterization of the contribution of the host immune response to outcomes of acute versus chronic S. aureus disease. We compared the immune responses between a model of self-limiting skin and soft tissue infection caused by the planktonic form of S. aureus versus a model of surgical mesh implant infection, which we show to be caused by a bacterial biofilm. The significantly altered host cytokines and chemokines were largely different in the two models, with responses diminished by 21 days post-implantation in surgical mesh infection. Because bacterial levels remained constant during the 21 days that the surgical mesh infection was followed, those cytokines that are significantly increased during chronic infection are not likely effective in eradicating biofilm. Comparison of the levels of cytokines and chemokines in acute versus chronic S. aureus infection can provide a starting point for evaluation of the role of specific immune factors that are present in one disease manifestation but not the other.


Subject(s)
Biofilms/growth & development , Disease Models, Animal , Soft Tissue Infections/immunology , Staphylococcal Infections/immunology , Staphylococcal Skin Infections/immunology , Staphylococcus aureus/immunology , Acute Disease , Animals , Chronic Disease , Female , Mice , Mice, Inbred C57BL , Soft Tissue Infections/microbiology , Staphylococcal Infections/microbiology , Staphylococcal Skin Infections/microbiology
4.
Clin Vaccine Immunol ; 21(5): 622-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24574539

ABSTRACT

Due to the emergence of highly virulent community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) infections, S. aureus has become a major threat to public health. A majority of CA-MRSA skin and soft tissue infections in the United States are caused by S. aureus USA300 strains that are known to produce high levels of alpha hemolysin (Hla). Therefore, vaccines that contain inactivated forms of this toxin are currently being developed. In this study, we sought to determine the immune mechanisms of protection for this antigen using a vaccine composed of a genetically inactivated form of Hla (HlaH35L). Using a murine model of skin and soft tissue infections (SSTI), we found that BALB/c mice were protected by vaccination with HlaH35L; however, Jh mice, which are deficient in mature B lymphocytes and lack IgM and IgG in their serum, were not protected. Passive immunization with anti-HlaH35L antibodies conferred protection against bacterial colonization. Moreover, we found a positive correlation between the total antibody concentration induced by active vaccination and reduced bacterial levels. Animals that developed detectable neutralizing antibody titers after active vaccination were significantly protected from infection. These data demonstrate that antibodies to Hla represent the major mechanism of protection afforded by active vaccination with inactivated Hla in this murine model of SSTI, and in this disease model, antibody levels correlate with protection. These results provide important information for the future development and evaluation of S. aureus vaccines.


Subject(s)
Antibodies, Bacterial/blood , Bacterial Toxins/immunology , Hemolysin Proteins/immunology , Soft Tissue Infections/prevention & control , Staphylococcal Skin Infections/prevention & control , Staphylococcal Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Bacterial Toxins/genetics , Disease Models, Animal , Female , Hemolysin Proteins/genetics , Male , Mice, Inbred BALB C , Mutant Proteins/genetics , Mutant Proteins/immunology , Soft Tissue Infections/immunology , Staphylococcal Skin Infections/immunology , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
5.
Clin Vaccine Immunol ; 20(8): 1338-40, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23803901

ABSTRACT

The staphylococcal adhesin clumping factor A (ClfA) has a variant amino acid sequence, generating the potential for alterations in epitope structure and immunogenicity of this vaccine candidate. We demonstrated for two recombinant ClfA(40-531) (a slightly truncated version of the fibrinogen-binding domain of ClfA containing amino acids 40 to 531) genetic variants that strain-specific epitopes are immunodominant. This work indicates that immune responses elicited by ClfA may, at least in part, be dependent on the strain of origin of the ClfA.


Subject(s)
Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Coagulase/genetics , Coagulase/immunology , Immunodominant Epitopes/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Animals , Antigens, Bacterial/chemistry , Coagulase/chemistry , Genetic Variation , Mice, Inbred BALB C , Models, Molecular , Protein Conformation
6.
PLoS One ; 8(4): e63040, 2013.
Article in English | MEDLINE | ID: mdl-23658662

ABSTRACT

Staphylococcus aureus is a major human pathogen and a leading cause of nosocomial and community-acquired infections. Development of a vaccine against this pathogen is an important goal. While S. aureus protective antigens have been identified in the literature, the majority have only been tested in a single animal model of disease. We wished to evaluate the ability of one S. aureus vaccine antigen to protect in multiple mouse models, thus assessing whether protection in one model translates to protection in other models encompassing the full breadth of infections the pathogen can cause. We chose to focus on genetically inactivated alpha toxin mutant HlaH35L. We evaluated the protection afforded by this antigen in three models of infection using the same vaccine dose, regimen, route of immunization, adjuvant, and challenge strain. When mice were immunized with HlaH35L and challenged via a skin and soft tissue infection model, HlaH35L immunization led to a less severe infection and decreased S. aureus levels at the challenge site when compared to controls. Challenge of HlaH35L-immunized mice using a systemic infection model resulted in a limited, but statistically significant decrease in bacterial colonization as compared to that observed with control mice. In contrast, in a prosthetic implant model of chronic biofilm infection, there was no significant difference in bacterial levels when compared to controls. These results demonstrate that vaccines may confer protection against one form of S. aureus disease without conferring protection against other disease presentations and thus underscore a significant challenge in S. aureus vaccine development.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Soft Tissue Infections/prevention & control , Staphylococcal Infections/prevention & control , Staphylococcal Skin Infections/prevention & control , Staphylococcal Vaccines/immunology , Adjuvants, Immunologic , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Bacterial Toxins/administration & dosage , Bacterial Toxins/genetics , Biofilms/drug effects , Biofilms/growth & development , Colony Count, Microbial , Disease Models, Animal , Female , Immunization , Immunoglobulin G/immunology , Mice , Soft Tissue Infections/immunology , Soft Tissue Infections/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus aureus/immunology , Vaccines, Attenuated
7.
PLoS One ; 8(3): e59232, 2013.
Article in English | MEDLINE | ID: mdl-23555002

ABSTRACT

In vivo bioluminescent imaging permits the visualization of bacteria in live animals, allowing researchers to monitor, both temporally and spatially, the progression of infection in each animal. We sought to engineer stably luminescent clinical strains of Staphylococcus aureus, with the goal of using such strains in mouse models. The gram-positive shuttle vector pMAD was used as the backbone for an integration plasmid. A chloramphenicol resistance gene, a modified lux operon from Photorhabdus luminescens, and approximately 650 bp of homology to the chromosome of the USA300 S. aureus strain NRS384 were added, generating plasmid pRP1195. Electroporation into strain RN4220 followed by temperature shift led to integration of pRP1195 into the chromosome. The integrated plasmid was transferred to clinical strains by phage transduction. Luminescent strains displayed no in vitro growth defects. Moreover, luminescence was stable in vitro after three rounds of subculture over 48 hours of growth in the absence of antibiotics. Mice were infected with a luminescent strain of NRS384 in skin and intravenous models. In a mouse skin model, luminescent bacteria were present in lesions that formed and cleared over the course of several days, and in an intravenous model, bacteria inoculated in the mouse tail vein were observed spreading to multiple tissues. No statistically significant difference in virulence was observed between NRS384 and the luminescent strain in either infection model. These preliminary data suggest that this luminescent USA300 strain is suitable for use in mouse models. Similar strains were engineered using other sequenced clinical strains. Because these strains are stably luminescent, they should prove useful in animal models of infection.


Subject(s)
Chromosomes, Bacterial/genetics , Founder Effect , Gene Transfer Techniques , Plasmids , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Animals , Bacteriophages/genetics , Electroporation , Injections, Intravenous , Luminescence , Luminescent Measurements , Mice , Operon , Photorhabdus/chemistry , Photorhabdus/genetics , Staphylococcal Infections/mortality , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/ultrastructure , Survival Analysis
8.
Carbohydr Res ; 345(11): 1600-8, 2010 Jul 19.
Article in English | MEDLINE | ID: mdl-20542498

ABSTRACT

There is no licensed vaccine for the prevention of shigellosis. Our approach to the development of a Shigella vaccines is based on inducing serum IgG antibodies to the O-specific polysaccharide (O-SP) domain of their lipopolysaccharides (LPS). We have shown that low molecular mass O-SP-core (O-SPC) fragments isolated from Shigella sonnei LPS conjugated to proteins induced significantly higher antibody levels in mice than the full length O-SP conjugates. This finding is now extended to the O-SPC of Shigella flexneri 2a and 6, and Shigella dysenteriae type 1. The structures of O-SPC, containing core plus 1-4 O-SP repeat units (RUs), were analyzed by NMR and mass spectroscopy. The first RUs attached to the cores of S. flexneri 2a and 6 LPS were different from the following RUs in their O-acetylation and/or glucosylation. Conjugates of core plus more than 1 RU were necessary to induce LPS antibodies in mice. The resulting antibody levels were comparable to those induced by the full length O-SP conjugates. In S. dysenteriae type 1, the first RU was identical to the following RUs, with the exception that the GlcNAc was bound to the core in the beta-configuration, while in all other RUs the GlcNAc was present in the alpha-configuration. In spite of this difference, conjugates of S. dysenteriae type 1 core with 1, 2, or 3 RUs induced LPS antibodies in mice with levels statistically higher than those of the full size O-SP conjugates. O-SPC conjugates are easy to prepare, characterize, and standardize, and their clinical evaluation is planned.


Subject(s)
Bacterial Vaccines/immunology , Glycoproteins/immunology , O Antigens/chemistry , O Antigens/immunology , Shigella dysenteriae/chemistry , Shigella flexneri/chemistry , Animals , Bacterial Vaccines/chemistry , Carbohydrate Sequence , Cattle , Glycoproteins/chemistry , Immunochemistry , Mice , Molecular Sequence Data , Shigella dysenteriae/immunology , Shigella flexneri/immunology
9.
Proc Natl Acad Sci U S A ; 107(3): 1172-7, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20080619

ABSTRACT

There is yet no licensed vaccine against malaria, a serious human disease affecting mostly children, with an annual death rate of about one million. Plasmodia, the malaria-causing parasites, have two obligatory hosts: mammals or birds, in which they multiply asexually, and mosquitoes with sexual multiplication. The most common and serious type of malaria is caused by Plasmodium falciparum. The circumsporozoite protein (CSP), a major surface antigen of sporozoites, is a protective antigen. A unique feature of P. falciparum CSP is its large central domain composed of over 30 tetrapeptide repeats of Asn-Ala-Asn-Pro (NANP). Several NANP peptide-protein conjugates were tested clinically but elicited a low level of CSP antibodies for a short duration. To provide a CSP-based candidate vaccine, we investigated recombinant CSP and NANP conjugates of various peptide lengths, with different N-terminal amino acids, bound at different ratios to various carrier proteins. Injected into mice, CSP alone and CSP or NANP conjugates induced antibodies with booster responses and were positive by the sporozoite immunofluorescent assay. The use of the mosquito stage P. falciparum ookinete surface protein, Pfs25, cross-linked onto itself as a carrier for NANP, induced in mice high levels of uniquely long-lasting antibodies to both vaccine components with secondary biological activities, that will provide immunity to liver infection by sporozoites and block transmission by mosquitoes.


Subject(s)
Malaria Vaccines/immunology , Peptides/chemistry , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Animals , Antibodies, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Female , Fluorescent Antibody Technique , Gas Chromatography-Mass Spectrometry , Malaria Vaccines/chemistry , Mice , Peptides/immunology , Protozoan Proteins/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , T-Lymphocytes/immunology
10.
Proc Natl Acad Sci U S A ; 106(19): 7974-8, 2009 May 12.
Article in English | MEDLINE | ID: mdl-19346477

ABSTRACT

Shigellosis, an enteric disease, is on the World Health Organization's priority prevention list. In one study, the Shigella sonnei O-specific polysaccharide (O-SP)-protein conjugate showed 72% protection against disease in Israeli army recruits exposed to high rates (8-14%) of infection. The protection was related to vaccine-induced IgG anti-O-SP levels. Synthetic oligosaccharides of Shigella dysenteriae type 1, bound by their reducing ends to a carrier protein ("sun"-type configuration), induced significantly higher antibody levels than the native O-SP bound to protein by multiple-point attachments ("lattice"-type configuration). Attempts to synthesize the S. sonnei O-SP based oligosaccharides were not successful. Here, we describe the isolation, characterization, and conjugation of low-molecular-mass O-SP-core (O-SPC) fragments. The O-SPC fragments were bound by their reducing ends similar to the preparation of the synthetic S. dysenteriae type 1 conjugates. The O-SPC conjugates used oxime linkages between the terminal Kdo residues at the reducing ends of the S. sonnei saccharides and aminooxy linkers bound to BSA or a recombinant diphtheria toxin. The coupling reaction was carried out at a neutral pH and room temperature. IgG antibody levels induced in young outbred mice by the S. sonnei O-SPC conjugates were significantly higher then those elicited by the O-SP conjugates. Accordingly, we propose to evaluate clinically these conjugates.


Subject(s)
Bacterial Vaccines/chemical synthesis , Dysentery, Bacillary/prevention & control , O Antigens/chemistry , Shigella sonnei/immunology , Animals , Antibodies/chemistry , Bacterial Vaccines/chemistry , Dysentery, Bacillary/immunology , Enzyme-Linked Immunosorbent Assay , Female , Glycoconjugates/chemistry , Hydrogen-Ion Concentration , Immunoglobulin G/chemistry , Lipopolysaccharides/chemistry , Magnetic Resonance Spectroscopy , Mice , Spectrometry, Mass, Electrospray Ionization
11.
Infect Immun ; 74(8): 4744-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16861662

ABSTRACT

The capsule of Bacillus anthracis, composed of poly-gamma-d-glutamic acid (gammaDPGA), is an essential virulence factor of B. anthracis. The capsule inhibits innate host defense through its antiphagocytic action. gammaDPGA is a poor immunogen, but when covalently bound to a carrier protein, it elicits serum antibodies. To identify the optimal construct for clinical use, synthetic gammaDPGAs of different lengths were bound to carrier proteins at different densities. The advantages of the synthetic over the natural polypeptide are the homogeneous chain length and end groups, allowing conjugates to be accurately characterized and standardized and their chemical compositions to be related to their immunogenicities. In the present study, we evaluated, in addition to methods reported by us, hydrazone, oxime, and thioether linkages between gammaDPGA and several proteins, including bovine serum albumin, recombinant Pseudomonas aeruginosa exotoxin A, recombinant B. anthracis protective antigen (rPA), and tetanus toxoid (TT). The effects of the dosage and formulation on the immunogenicities of the conjugates were evaluated in mice. All conjugates were immunogenic. The optimal gammaDPGA chain length of 10 to 15 amino acids and the density, an average of 15 mol gammaDPGA per mol of protein, were confirmed. The thioether bond was the optimal linkage type, and TT and rPA were the best carriers. The optimal dosage was 1.2 to 2.5 microg of gammaDPGA per mouse, and adsorption of the conjugates onto aluminum hydroxide significantly increased the antibody response to the protein with a lesser effect on anti-gammaDPGA levels.


Subject(s)
Anthrax Vaccines/immunology , Antibodies, Bacterial/blood , Bacillus anthracis/immunology , Polyglutamic Acid/chemistry , Vaccines, Conjugate/chemistry , Vaccines, Conjugate/immunology , Animals , Anthrax/immunology , Anthrax/prevention & control , Anthrax Vaccines/administration & dosage , Anthrax Vaccines/chemistry , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Bacterial Toxins/administration & dosage , Bacterial Toxins/chemistry , Bacterial Toxins/immunology , Female , Immunization , Mice , Polyglutamic Acid/administration & dosage , Polyglutamic Acid/immunology , Sulfides/chemistry , Tetanus Toxoid/administration & dosage , Tetanus Toxoid/chemistry , Tetanus Toxoid/immunology , Vaccines, Conjugate/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...