Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 328
Filter
1.
Immunol Cell Biol ; 102(5): 365-380, 2024.
Article in English | MEDLINE | ID: mdl-38572664

ABSTRACT

Staphylococcus aureus is a significant bacterial pathogen in both community and hospital settings, and the escalation of antimicrobial-resistant strains is of immense global concern. Vaccination is an inviting long-term strategy to curb staphylococcal disease, but identification of an effective vaccine has proved to be challenging. Three well-characterized, ubiquitous, secreted immune evasion factors from the staphylococcal superantigen-like (SSL) protein family were selected for the development of a vaccine. Wild-type SSL3, 7 and 11, which inhibit signaling through Toll-like receptor 2, cleavage of complement component 5 and neutrophil function, respectively, were successfully combined into a stable, active fusion protein (PolySSL7311). Vaccination of mice with an attenuated form of the PolySSL7311 protein stimulated significantly elevated specific immunoglobulin G and splenocyte proliferation responses to each component relative to adjuvant-only controls. Vaccination with PolySSL7311, but not a mixture of the individual proteins, led to a > 102 reduction in S. aureus tissue burden compared with controls after peritoneal challenge. Comparable antibody responses were elicited after coadministration of the vaccine in either AddaVax (an analog of MF59) or an Alum-based adjuvant; but only AddaVax conferred a significant reduction in bacterial load, aligning with other studies that suggest both cellular and humoral immune responses are necessary for protective immunity to S. aureus. Anti-sera from mice immunized with PolySSL7311, but not individual proteins, partially neutralized the functional activities of SSL7. This study confirms the importance of these SSLs for the survival of S. aureus in vivo and suggests that PolySSL7311 is a promising vaccine candidate.


Subject(s)
Bacterial Proteins , Staphylococcal Infections , Staphylococcal Vaccines , Staphylococcus aureus , Superantigens , Animals , Staphylococcus aureus/immunology , Staphylococcal Vaccines/immunology , Superantigens/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Mice , Bacterial Proteins/immunology , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Female , Recombinant Fusion Proteins/immunology , Immunoglobulin G/immunology , Immunoglobulin G/blood , Feasibility Studies , Vaccination , Antigens, Bacterial/immunology , Mice, Inbred BALB C , Adjuvants, Immunologic
2.
Nat Commun ; 15(1): 3420, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658531

ABSTRACT

Poly-ß-(1-6)-N-acetylglucosamine (PNAG) is an important vaccine target, expressed on many pathogens. A critical hurdle in developing PNAG based vaccine is that the impacts of the number and the position of free amine vs N-acetylation on its antigenicity are not well understood. In this work, a divergent strategy is developed to synthesize a comprehensive library of 32 PNAG pentasaccharides. This library enables the identification of PNAG sequences with specific patterns of free amines as epitopes for vaccines against Staphylococcus aureus (S. aureus), an important human pathogen. Active vaccination with the conjugate of discovered PNAG epitope with mutant bacteriophage Qß as a vaccine carrier as well as passive vaccination with diluted rabbit antisera provides mice with near complete protection against infections by S. aureus including methicillin-resistant S. aureus (MRSA). Thus, the comprehensive PNAG pentasaccharide library is an exciting tool to empower the design of next generation vaccines.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Animals , Staphylococcal Infections/prevention & control , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Mice , Staphylococcus aureus/immunology , Rabbits , Staphylococcal Vaccines/immunology , Staphylococcal Vaccines/administration & dosage , Female , Methicillin-Resistant Staphylococcus aureus/immunology , Acetylglucosamine/immunology , Humans , Epitopes/immunology , Mice, Inbred BALB C
3.
Int J Mol Sci ; 23(2)2022 Jan 16.
Article in English | MEDLINE | ID: mdl-35055134

ABSTRACT

The main purpose of this review is to present justification for the urgent need to implement specific prophylaxis of invasive Staphylococcus aureus infections. We emphasize the difficulties in achieving this goal due to numerous S. aureus virulence factors important for the process of infection and the remarkable ability of these bacteria to avoid host defense mechanisms. We precede these considerations with a brief overview of the global necessitiy to intensify the use of vaccines against other pathogens as well, particularly in light of an impasse in antibiotic therapy. Finally, we point out global trends in research into modern technologies used in the field of molecular microbiology to develop new vaccines. We focus on the vaccines designed to fight the infections caused by S. aureus, which are often resistant to the majority of available therapeutic options.


Subject(s)
Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/therapeutic use , Staphylococcus aureus/immunology , Drug Resistance, Bacterial/drug effects , Humans , Staphylococcal Infections/immunology , Staphylococcal Vaccines/immunology , Staphylococcal Vaccines/pharmacology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Vaccine Development , Virulence Factors/genetics , Virulence Factors/immunology
4.
Infect Genet Evol ; 96: 105149, 2021 12.
Article in English | MEDLINE | ID: mdl-34801756

ABSTRACT

BACKGROUND: The emergence of methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant S. aureus (VRSA) strains is a significant public health concern. Considering the high morbidity and mortality of invasive S. aureus infections and multi-drug resistant strains, there is an urgent need for non-antibiotic immune-based approaches to cure these infections. Despite all efforts, vaccine candidates targeting S. aureus failed in human clinical trials, and no approved vaccine is available against this pathogen. Therefore, this study aimed to introduce suitable candidates for immunization against S. aureus using a comprehensive reverse vaccinology approach. METHODS: In this study, we retrieved putative immunogenic targets from three different levels (literature review, automated reverse vaccinology, and manual reverse vaccinology) and evaluated them using several immunoinformatics analyses including antigenicity, allergenicity, PSI-BLAST to human proteome, physiochemical properties, B-cell, and T-cell epitopes. In the next step, the quartile method scoring was used to the shortlisted proteins. Finally, the molecular docking and immune simulation of immunogenic targets were performed. RESULTS: This study presents 12 vaccine candidates, including three enzymatic proteins (WP_000222271.1, WP_001170274, and WP_000827736.1), three cell wall-associated proteins (WP_001125631.1, WP_000731642, and WP_000751265.1), two hemolysins (WP_000594517.1, and WP_000916697.1), one secretion involved protein (WP_000725226.1), one heme­iron binding protein (WP_001041573.1), one superantigen like protein (WP_000668994.1) and one hypothetical proteins (WP_000737711.1). CONCLUSION: Through quartile scoring method, immune simulation and molecular docking, four promising targets including lytic transglycosylase IsaA, HlgA, secretory antigen precursor SsaA, and heme uptake protein IsdB were selected as the shortlisted proteins. It seems that a polarized immunization (Th1/Th17) response is needed for protection against this bacterium. An optimized formulation based on these putative immunogenic proteins and a wisely adjuvant selection may drive the immune system toward a full protection.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Vaccinology/methods , Humans , Molecular Docking Simulation , Vaccines, Subunit/immunology
5.
Biologicals ; 73: 8-15, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34376341

ABSTRACT

Staphylococcus aureus is an important human opportunistic pathogen that can have a major influence on public health. Here, we aimed to evaluate different aspects of the immune response to a novel multi-epitope fusion protein (HMS) based on HlaH35L, MntC, and SACOL0723 proteins in comparison to the individual antigens. For this purpose, specific total IgG, IgG1, and IgG2a isotypes and the cytokines related to Th1, Th2, and Th17 were assessed. The Bio-efficiency of the fusion protein was evaluated by opsonic killing activity. The HMS fusion protein elicited a high specific IgG level and also induced a higher level of Th1, Th2, and Th17-related cytokines which were more polarized towards the Th1 and Th17 compared to individual antigens. The HMS-specific antisera also significantly promoted phagocytosis of S. aureus COL strain by mouse macrophages. In conclusion, the fusion protein might be an effective vaccine for potential protective immunity against a lethal infection of S. aureus in mice.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Animals , Antibodies, Bacterial/immunology , Cytokines/immunology , Epitopes/immunology , Immunoglobulin G/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Mice , Mice, Inbred BALB C , Staphylococcal Infections/immunology , T-Lymphocytes/immunology
6.
Vet Immunol Immunopathol ; 238: 110290, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34217108

ABSTRACT

Staphylococcus aureus is a leading cause of bovine intramammary infections (IMI). Standard antibiotic treatments are not very effective and currently available vaccines lack tangible efficacy. Developing a vaccine formulation for S. aureus mastitis is challenging and selection of target antigens is critical. The gene products of six S. aureus genes that are highly expressed during IMI were selected as antigens for this study. The vaccine contained six recombinant proteins formulated with Emulsigen®-D, a CpG oligodeoxynucleotide and indolicidin. Nine cows in mid-lactation received the vaccine while ten received saline (placebo). Two immunizations were performed 10 weeks apart. All the antigens induced an immune response. A balanced immune response (IgG2/IgG1 ratio of 1) was observed for antigen SACOL0442 while a predominant Th2 response was observed for the other antigens (IgG2/IgG1 ratio <1). Immunizations induced CD4+ cell proliferation in response to SACOL0442, SACOL0029, SACOL0720 and SACOL1912 while a CD8+ cell proliferation was induced by SACOL0720. Four weeks after the second immunization, three quarters per animal were experimentally infused with ∼60 CFU of S. aureus. Although no difference in S. aureus counts was observed between the two groups after this robust infectious challenge, a sustained reduction in milk somatic cells counts (SCC) was observed in vaccinated cows. A correlation between SCC and S. aureus counts in milk was also observed. Altogether, this indicates that the collective immune responses induced by the antigens certainly contribute to the observed benefits of the whole vaccine. More work is needed to understand how different antigens stimulate a different response using the same adjuvant.


Subject(s)
Bacterial Proteins/immunology , Mastitis, Bovine/prevention & control , Staphylococcal Vaccines/immunology , Staphylococcus/classification , Adjuvants, Immunologic , Animals , Antibodies, Bacterial/blood , Cattle , Female , Immunity, Cellular , Immunity, Humoral , Immunoglobulin G/blood , Mastitis, Bovine/microbiology , Vaccination
7.
Front Immunol ; 12: 705360, 2021.
Article in English | MEDLINE | ID: mdl-34305945

ABSTRACT

Staphylococcus aureus is one of the most important human pathogens worldwide. Its high antibiotic resistance profile reinforces the need for new interventions like vaccines in addition to new antibiotics. Vaccine development efforts against S. aureus have failed so far however, the findings from these human clinical and non-clinical studies provide potential insight for such failures. Currently, research is focusing on identifying novel vaccine formulations able to elicit potent humoral and cellular immune responses. Translational science studies are attempting to discover correlates of protection using animal models as well as in vitro and ex vivo models assessing efficacy of vaccine candidates. Several new vaccine candidates are being tested in human clinical trials in a variety of target populations. In addition to vaccines, bacteriophages, monoclonal antibodies, centyrins and new classes of antibiotics are being developed. Some of these have been tested in humans with encouraging results. The complexity of the diseases and the range of the target populations affected by this pathogen will require a multipronged approach using different interventions, which will be discussed in this review.


Subject(s)
Staphylococcal Infections/prevention & control , Staphylococcal Vaccines , Staphylococcus aureus/immunology , Vaccine Development , Adjuvants, Immunologic , Animals , Antigens, Bacterial/immunology , Clinical Trials as Topic , Extracellular Vesicles/immunology , Glycoconjugates/immunology , Gram-Negative Bacteria/immunology , Host-Pathogen Interactions , Humans , Immunity, Cellular , Immunity, Humoral , Immunogenicity, Vaccine , In Vitro Techniques , Mice , Models, Animal , Nucleic Acid-Based Vaccines/immunology , Periplasm/immunology , Recombinant Proteins/immunology , Staphylococcal Vaccines/immunology , Staphylococcal Vaccines/therapeutic use , Translational Science, Biomedical , Vaccines, Attenuated/immunology , Vaccines, Synthetic/immunology
8.
Front Immunol ; 12: 684823, 2021.
Article in English | MEDLINE | ID: mdl-34122448

ABSTRACT

HI, a fusion protein that consists of the alpha-toxin (Hla) and the N2 domain of iron surface determinant B (IsdB), is one of the antigens in the previously reported S. aureus vaccine rFSAV and has already entered phase II clinical trials. Previous studies revealed that HI is highly immunogenic in both mice and healthy volunteers, and the humoral immune response plays key roles in HI-mediated protection. In this study, we further investigated the protective efficacy of immunization with HI plus four different adjuvants in a mouse bacteremia model. Results showed that HI-mediated protection was altered in response to different adjuvants. Using antisera from immunized mice, we identified seven B-cell immunodominant epitopes on Hla and IsdB, including 6 novel epitopes (Hla1-18, Hla84-101, Hla186-203, IsdB342-359, IsdB366-383, and IsdB384-401). The immunodominance of B-cell epitopes, total IgG titers and the levels of IFN-γ and IL-17A from mice immunized with HI plus different adjuvants were different from each other, which may explain the difference in protective immunity observed in each immunized group. Thus, our results indicate that adjuvants largely affected the immunodominance of epitopes and the protective efficacy of HI, which may guide further adjuvant screening for vaccine development and optimization.


Subject(s)
Bacteremia/immunology , Bacterial Toxins/immunology , Cation Transport Proteins/immunology , Epitopes, B-Lymphocyte/immunology , Hemolysin Proteins/immunology , Immunodominant Epitopes/immunology , Staphylococcal Infections/prevention & control , Animals , Bacteremia/prevention & control , Disease Models, Animal , Female , Immunization, Passive , Immunotherapy, Adoptive , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice , Mice, Inbred BALB C , Staphylococcal Infections/immunology , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/immunology
9.
J Immunol Methods ; 494: 113055, 2021 07.
Article in English | MEDLINE | ID: mdl-33857474

ABSTRACT

To develop a suitable and effective vaccine against Staphylococcus aureus (S. aureus), we selected the Hla-MntC-SACOL0723 (HMS) recombinant protein with two different formulations of alum and Monophosphoryl lipid A (MPL) adjuvants. In this study, we aimed to evaluate the potentials of alum and MPL adjuvants in stimulating the immune response of HMS vaccine candidate against S. aureus. To evaluate the type of induced immune response, anti-HMS total IgG, IgG1, IgG2a, and IFN-γ, IL-2, IL-4, and IL-17 cytokines were determined after vaccination of mice with HMS-alum, HMS-MPL candidates. Mice were challenged with Methicillin-resistant Staphylococcus aureus (MRSA) was isolated from pressure sores and evaluated for bacterial load in the kidney homogenates and survival rate. It was observed that total IgG and isotypes (IgG1 and IgG2a), IL-4, and IL-17 were significantly increased in the group that received HMS-alum vaccine compared with the group that received HMS-MPL formulation. On the other hand, the levels of IFN-γ and IL-2 cytokines in the group that received HMS-MPL were higher than the group that received HMS-alum formulation. Bacterial load in the mice who received HMS protein formulated with alum adjuvant was reduced more than the mice who received HMS protein formulated with MPL adjuvant. Histopathological analysis showed more pathological changes in kidney tissues of the group received of HMS-MPL compared with the HMS-alum formulation. The survival rate was equal in both groups of immunized with HMS-alum and HMS-MPL formulations. Finally, it could be concluded that both adjuvants of alum and MPL are suitable immune response enhancers to HMS vaccine candidate.


Subject(s)
Kidney/pathology , Methicillin-Resistant Staphylococcus aureus/physiology , Periplasmic Binding Proteins/genetics , Sepsis/immunology , Staphylococcal Infections/immunology , Staphylococcal Vaccines/immunology , Staphylococcus aureus/physiology , Alum Compounds , Animals , Female , HLA Antigens/genetics , Immunoglobulin G/metabolism , Interleukin-17/metabolism , Interleukin-4/metabolism , Lipid A/analogs & derivatives , Lipid A/immunology , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Sepsis/prevention & control , Up-Regulation
10.
Front Immunol ; 12: 624310, 2021.
Article in English | MEDLINE | ID: mdl-33777005

ABSTRACT

Staphylococcus aureus causes a wide range of diseases from skin infections to life threatening invasive diseases such as bacteremia, endocarditis, pneumonia, surgical site infections, and osteomyelitis. Skin infections such as furuncles, carbuncles, folliculitis, erysipelas, and cellulitis constitute a large majority of infections caused by S. aureus (SA). These infections cause significant morbidity, healthcare costs, and represent a breeding ground for antimicrobial resistance. Furthermore, skin infection with SA is a major risk factor for invasive disease. Here we describe the pre-clinical efficacy of a multicomponent toxoid vaccine (IBT-V02) for prevention of S. aureus acute skin infections and recurrence. IBT-V02 targets six SA toxins including the pore-forming toxins alpha hemolysin (Hla), Panton-Valentine leukocidin (PVL), leukocidin AB (LukAB), and the superantigens toxic shock syndrome toxin-1 and staphylococcal enterotoxins A and B. Immunization of mice and rabbits with IBT-V02 generated antibodies with strong neutralizing activity against toxins included in the vaccine, as well as cross-neutralizing activity against multiple related toxins, and protected against skin infections by several clinically relevant SA strains of USA100, USA300, and USA1000 clones. Efficacy of the vaccine was also shown in non-naïve mice pre-exposed to S. aureus. Furthermore, vaccination with IBT-V02 not only protected mice from a primary infection but also demonstrated lasting efficacy against a secondary infection, while prior challenge with the bacteria alone was unable to protect against recurrence. Serum transfer studies in a primary infection model showed that antibodies are primarily responsible for the protective response.


Subject(s)
Reinfection/prevention & control , Staphylococcal Skin Infections/prevention & control , Staphylococcal Vaccines/pharmacology , Staphylococcus aureus/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Disease Models, Animal , Female , Immunization , Immunogenicity, Vaccine , Male , Mice, Inbred BALB C , Rabbits , Reinfection/immunology , Reinfection/microbiology , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Staphylococcal Vaccines/immunology
11.
Front Immunol ; 12: 621754, 2021.
Article in English | MEDLINE | ID: mdl-33717122

ABSTRACT

Staphylococcus aureus is a leading cause of significant morbidity and mortality and an enormous economic burden to public health worldwide. Infections caused by methicillin-resistant S. aureus (MRSA) pose a major threat as MRSA strains are becoming increasingly prevalent and multi-drug resistant. To this date, vaccines targeting surface-bound antigens demonstrated promising results in preclinical testing but have failed in clinical trials. S. aureus pathogenesis is in large part driven by immune destructive and immune modulating toxins and thus represent promising vaccine targets. Hence, the objective of this study was to evaluate the safety and immunogenicity of a staphylococcal 4-component vaccine targeting secreted bi-component pore-forming toxins (BCPFTs) and superantigens (SAgs) in non-human primates (NHPs). The 4-component vaccine proved to be safe, even when repeated vaccinations were given at a dose that is 5 to 10- fold higher than the proposed human dose. Vaccinated rhesus macaques did not exhibit clinical signs, weight loss, or changes in hematology or serum chemistry parameters related to the administration of the vaccine. No acute, vaccine-related elevation of serum cytokine levels was observed after vaccine administration, confirming the toxoid components lacked superantigenicity. Immunized animals demonstrated high level of toxin-specific total and neutralizing antibodies toward target antigens of the 4-component vaccine as well as cross-neutralizing activity toward staphylococcal BCPFTs and SAgs that are not direct targets of the vaccine. Cross-neutralization was also observed toward the heterologous streptococcal pyogenic exotoxin B. Ex vivo stimulation of PBMCs with individual vaccine components demonstrated an overall increase in several T cell cytokines measured in supernatants. Immunophenotyping of CD4 T cells ex vivo showed an increase in Ag-specific polyfunctional CD4 T cells in response to antigen stimulation. Taken together, we demonstrate that the 4-component vaccine is well-tolerated and immunogenic in NHPs generating both humoral and cellular immune responses. Targeting secreted toxin antigens could be the next-generation vaccine approach for staphylococcal vaccines if also proven to provide efficacy in humans.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/immunology , Staphylococcal Toxoid/immunology , Staphylococcal Vaccines/immunology , Animals , Antibodies, Bacterial/blood , Antibody Formation , Broadly Neutralizing Antibodies/blood , Immunity, Heterologous , Immunogenicity, Vaccine , Lymphocyte Activation , Macaca mulatta , Superantigens/immunology , Vaccination
12.
Front Immunol ; 12: 633672, 2021.
Article in English | MEDLINE | ID: mdl-33692805

ABSTRACT

Recurrent S. aureus infections are common, suggesting that natural immune responses are not protective. All candidate vaccines tested thus far have failed to protect against S. aureus infections, highlighting an urgent need to better understand the mechanisms by which the bacterium interacts with the host immune system to evade or prevent protective immunity. Although there is evidence in murine models that both cellular and humoral immune responses are important for protection against S. aureus, human studies suggest that T cells are critical in determining susceptibility to infection. This review will use an "anatomic" approach to systematically outline the steps necessary in generating a T cell-mediated immune response against S. aureus. Through the processes of bacterial uptake by antigen presenting cells, processing and presentation of antigens to T cells, and differentiation and proliferation of memory and effector T cell subsets, the ability of S. aureus to evade or inhibit each step of the T-cell mediated response will be reviewed. We hypothesize that these interactions result in the redirection of immune responses away from protective antigens, thereby precluding the establishment of "natural" memory and potentially inhibiting the efficacy of vaccination. It is anticipated that this approach will reveal important implications for future design of vaccines to prevent these infections.


Subject(s)
Drug Design , Immune Evasion , Immunologic Memory , Reinfection/prevention & control , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/therapeutic use , Staphylococcus aureus/immunology , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Antigens, Bacterial/immunology , Epitopes , Humans , Immunogenicity, Vaccine , Lymphocyte Activation , Reinfection/immunology , Reinfection/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Vaccines/adverse effects , Staphylococcal Vaccines/immunology , Staphylococcus aureus/pathogenicity , T-Lymphocytes/microbiology
13.
Toxins (Basel) ; 12(10)2020 10 02.
Article in English | MEDLINE | ID: mdl-33023185

ABSTRACT

Staphylococcal superantigen toxins lead to a devastating cytokine storm resulting in shock and multi-organ failure. We have previously assessed the safety and immunogenicity of a recombinant toxic shock syndrome toxin 1 variant vaccine (rTSST-1v) in clinical trials (NCT02971670 and NCT02340338). The current study assessed neutralizing antibody titers after repeated vaccination with escalating doses of rTSST-1v. At study entry, 23 out of 34 subjects (67.6%) had neutralizing antibody titers inhibiting T cell activation as determined by 3H-thymidine incorporation at a serum dilution of ≤1:100 with similar figures for inhibition of IL-2 activation (19 of 34 subjects, 55.9%) as assessed by quantitative PCR. After the first vaccination, numbers of subjects with neutralization titers inhibiting T cell activation (61.7% ≥ 1:1000) and inhibiting IL-2 gene induction (88.2% ≥ 1:1000) increased. The immune response was augmented after the second vaccination (inhibiting T cell activation: 78.8% ≥ 1:1000; inhibiting IL-2 induction: 93.9% ≥ 1:1000) corroborated with a third immunization months later in a small subgroup of subjects. Assessment of IFNγ, TNFα and IL-6 inhibition revealed similar results, whereas neutralization titers did not change in placebo participants. Antibody titer studies show that vaccination with rTSST-1v in subjects with no/low neutralizing antibodies can rapidly induce high titer neutralizing antibodies persisting over months.


Subject(s)
Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Bacterial Toxins/administration & dosage , Cytokine Release Syndrome/prevention & control , Enterotoxins/administration & dosage , Immunogenicity, Vaccine , Shock, Septic/prevention & control , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/administration & dosage , Staphylococcus aureus/drug effects , Superantigens/administration & dosage , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Cells, Cultured , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/microbiology , Cytokines/genetics , Cytokines/metabolism , Double-Blind Method , Enterotoxins/genetics , Enterotoxins/immunology , Humans , Lymphocyte Activation/drug effects , Prospective Studies , Shock, Septic/immunology , Shock, Septic/microbiology , Single-Blind Method , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Vaccines/genetics , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Superantigens/genetics , Superantigens/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Treatment Outcome , Vaccination , Vaccines, Synthetic/administration & dosage
14.
PLoS Pathog ; 16(8): e1008733, 2020 08.
Article in English | MEDLINE | ID: mdl-32817694

ABSTRACT

Staphylococcus aureus (S. aureus) is one of the most common bacterial infections worldwide, and antibiotic resistant strains such as Methicillin-Resistant S. aureus (MRSA) are a major threat and burden to public health. MRSA not only infects immunocompromised patients but also healthy individuals and has rapidly spread from the healthcare setting to the outside community. However, all vaccines tested in clinical trials to date have failed. Immunocompromised individuals such as patients with HIV or decreased levels of CD4+ T cells are highly susceptible to S. aureus infections, and they are also at increased risk of developing fungal infections. We therefore wondered whether stimulation of antifungal immunity might promote the type of immune responses needed for effective host defense against S. aureus. Here we show that vaccination of mice with a fungal ß-glucan particle (GP) loaded with S. aureus antigens provides protective immunity to S. aureus. We generated glucan particles loaded with the four S. aureus proteins ClfA, IsdA, MntC, and SdrE, creating the 4X-SA-GP vaccine. Vaccination of mice with three doses of 4X-SA-GP promoted protection in a systemic model of S. aureus infection with a significant reduction in the bacterial burden in the spleen and kidneys. 4X-SA-GP vaccination induced antigen-specific Th1 and Th17 CD4+ T cell and antibody responses and provided long-term protection. This work suggests that the GP vaccine system has potential as a novel approach to developing vaccines for S. aureus.


Subject(s)
Saccharomyces cerevisiae/immunology , Staphylococcal Infections/immunology , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Animals , Antibodies, Bacterial/immunology , Coagulase/administration & dosage , Coagulase/genetics , Coagulase/immunology , Female , Humans , Mice , Mice, Inbred C57BL , Saccharomyces cerevisiae/chemistry , Staphylococcal Infections/microbiology , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/genetics , Staphylococcus aureus/genetics , Th1 Cells/immunology , Th17 Cells/immunology , Vaccination , beta-Glucans/administration & dosage , beta-Glucans/immunology
15.
Theranostics ; 10(16): 7131-7149, 2020.
Article in English | MEDLINE | ID: mdl-32641983

ABSTRACT

Background: Vaccination provides an alternative to antibiotics in addressing drug-resistant Staphylococcus aureus (S. aureus) infection. However, vaccine potency is often limited by a lack of antigenic breadth and a demand on the generation of antibody responses alone. Methods: In this study, bacterial extracellular vesicles (EVs) coating indocyanine green (ICG)-loaded magnetic mesoporous silica nanoparticles (MSN) were constructed as multi-antigenic vaccines (EV/ICG/MSN) with the ability to modulate antigen presentation pathways in dendritic cells (DCs) to induce cellular immune responses. Results: Exposing the EV/ICG/MSNs to a laser could promote DC maturation and enhance the proteasome-dependent antigen presentation pathway by facilitating endolysosomal escape, improving proteasome activity, and elevating MHC-I expression. Immunization by EV/ICG/MSNs with laser irradiation in vivo triggered improved CD8+ T cell responses while maintaining CD4+ T cell responses and humoral immunity. In addition, in vivo tracking data revealed that the vaccine could be efficiently transported from the injection site into lymph nodes. Skin infection experiments showed that the vaccine not only prevented and treated superficial infection but also decreased bacterial invasiveness, thus strongly suggesting that EV/ICG/MSNs were effective in preventing complications resulting from the introduction of S. aureus infections. Conclusion: This multi-antigenic nanovaccine-based modulation of antigen presentation pathways provides an effective strategy against drug-resistant S. aureus infection.


Subject(s)
Drug Carriers/chemistry , Extracellular Vesicles/immunology , Staphylococcal Skin Infections/therapy , Staphylococcal Vaccines/administration & dosage , Staphylococcus aureus/immunology , Animals , Antigen Presentation , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Disease Models, Animal , Drug Resistance, Bacterial/immunology , Humans , Immunity, Cellular , Male , Mice , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Staphylococcal Vaccines/genetics , Staphylococcal Vaccines/immunology , Staphylococcus aureus/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
16.
Iran J Allergy Asthma Immunol ; 19(2): 159-171, 2020 Apr 16.
Article in English | MEDLINE | ID: mdl-32372629

ABSTRACT

Staphylococcal enterotoxin B (SEB), apotent superantigen, is responsible for many disorders caused by Staphylococcus aureus. With regard to the appearance of multidrug-resistant strains of the bacteria, there is a great need to develop an efficient vaccine against this pathogen. In the present study, the immunogenicity of recombinant SEB was evaluated following nasal administration to BABLB/c mice. Indeed, the rSEB protein was entrapped into chitosan nanoparticles and the immunogenicity of nano-formulation was investigated. SEB protein was expressed in E. coli BL21 (DE3) and purified by using a nickel column. Chitosan nanoparticles were synthesized in the presence of rSEB; using the ionic gelation technique. Synthesized NPs containing rSEB and bare rSEB were administered to mice nasally. Serum and stool IgG and IgA antibody showed that both formulations were able to evoke the mice's immune responses and there was no significant difference between them. Results of the toxin neutralization test on Vero cells indicated that the sera of the immunized mice had an inhibitory effect on the growth of these cells (p<0.001). Nasal administration of bare rSEB could efficiently simulate the mice's immune system and nano-delivery of this protein via nasal route had not a significant impact on its immunogenicity improvement.


Subject(s)
Enterotoxins/immunology , Recombinant Proteins/immunology , Staphylococcal Infections/immunology , Staphylococcal Vaccines/immunology , Staphylococcus aureus/physiology , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Chitosan/chemistry , Enterotoxins/chemistry , Humans , Immunity, Humoral , Immunoglobulin A/blood , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Recombinant Proteins/chemistry
17.
J Dairy Sci ; 103(5): 4588-4605, 2020 May.
Article in English | MEDLINE | ID: mdl-32113759

ABSTRACT

Staphylococcus aureus is one of the pathogens most frequently isolated from cases of mastitis worldwide. To decrease the effect of S. aureus mastitis in dairy farming, alternative strategies for controlling mastitis are needed that depend on a better knowledge of cow-to-cow variations in S. aureus antibody production. The present study sought to explore the diversity of S. aureus antibodies produced by dairy cows with a distinct mastitis history and vaccinated with a polyvalent mastitis vaccine. We obtained protein extracts from S. aureus isolates derived from persistent subclinical mastitis. Proteins were fractionated using 2-dimensional gel electrophoresis and Western blotting. Then, Western blotting membranes were exposed to sera from 24 dairy cows that had been divided into the following groups: vaccinated dairy cows that were infected with S. aureus, further subdivided according to whether they (a) remained infected by S. aureus or (b) recovered from the intramammary infection; unvaccinated dairy cows infected with S. aureus; and vaccinated healthy dairy cows with no history of S. aureus mastitis. Proteins found to be reactive by Western blot were identified by mass spectrometry (MALDI/TOF-TOF). Our most important finding was that F0F1 ATP synthase subunit α, succinyl-diaminopimelate desuccinylase, and cysteinyl-tRNA synthetase were potential candidate proteins for the prevention of S. aureus mastitis. This study strengthens the notion that variations among animals should not be ignored and shows that the heterogeneity of antibody production against anti-staphylococcal antigens in animals may enable the identification of new immunotherapy targets.


Subject(s)
Antibodies, Bacterial/blood , Mastitis, Bovine/immunology , Staphylococcal Infections/veterinary , Staphylococcal Vaccines/administration & dosage , Staphylococcus aureus/immunology , Animals , Antibodies, Bacterial/immunology , Cattle , Female , Humans , Mastitis, Bovine/microbiology , Mastitis, Bovine/prevention & control , Milk , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Vaccines/immunology
18.
Immunol Cell Biol ; 98(5): 369-381, 2020 05.
Article in English | MEDLINE | ID: mdl-32150301

ABSTRACT

Most pathogens initiate infection via the mucosa, therefore delivery of vaccines directly to the mucosa is likely to be advantageous for stimulating protective immunity at the site of entry. PilVax is a novel mucosal vaccine platform that harnesses Lactococcus lactis bacteria engineered to stably express multiple copies of vaccine peptide antigens within pili, hair-like structures which extend from the cell wall. This strategy elicited systemic and mucosal antibody responses to a model antigen after intranasal immunization, but has not been tested for its capacity to stimulate protective mucosal immunity. A well-characterized linear B-cell epitope, D3(22-33) , from the fibronectin-binding protein A of Staphylococcus aureus was successfully introduced into PilVax and delivered intranasally to mice. Specific antipeptide immunoglobulin (Ig) G and IgA antibodies were detected in the serum and respiratory mucosa of vaccinated mice. Responses to the major pilus backbone protein Spy0128 were also assessed; robust antibody responses to this antigen were generated both systemically and in the respiratory and intestinal mucosa. Mice were challenged intranasally with the mouse-adapted S. aureus JSNZ strain and the S. aureus load quantified 7 days after challenge. Unexpectedly, exposure to PilVax, irrespective of the presence of the peptide, resulted in a significant reduction in S. aureus load in both the intestine and nasal mucosa (both P < 0.05) when compared with unvaccinated control mice. The mechanism(s) of protection are unclear, but merit further investigation to determine whether PilVax is a suitable platform for delivery of vaccine candidate antigens to the mucosa.


Subject(s)
Immunity, Mucosal , Lactococcus lactis , Staphylococcal Vaccines/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Bacterial Load , Epitopes, B-Lymphocyte/immunology , Intestines/microbiology , Lactococcus lactis/immunology , Mice , Mice, Inbred BALB C , Nasal Mucosa/microbiology , Staphylococcus aureus
19.
Infect Immun ; 88(4)2020 03 23.
Article in English | MEDLINE | ID: mdl-32014894

ABSTRACT

Unbiased identification of individual immunogenic B-cell epitopes in major antigens of a pathogen remains a technology challenge for vaccine discovery. We therefore developed a platform for rapid phage display screening of deep recombinant libraries consisting of as few as one major pathogen antigen. Using the bicomponent pore-forming leukocidin (Luk) exotoxins of the major pathogen Staphylococcus aureus as a prototype, we randomly fragmented and separately ligated the hemolysin gamma A (HlgA) and LukS genes into a custom-built phage display system, termed pComb-Opti8. Deep sequence analysis of barcoded amplimers of the HlgA and LukS gene fragment libraries demonstrated that biopannng against a cross-reactive anti-Luk monoclonal antibody (MAb) recovered convergent molecular clones with short overlapping homologous sequences. We thereby identified an 11-amino-acid sequence that is highly conserved in four Luk toxin subunits and is ubiquitous in representation within S. aureus clinical isolates. The isolated 11-amino-acid peptide probe was predicted to retain the native three-dimensional (3D) conformation seen within the Luk holotoxin. Indeed, this peptide was recognized by the selecting anti-Luk MAb, and, using mutated peptides, we showed that a particular amino acid side chain was essential for these interactions. Furthermore, murine immunization with this peptide elicited IgG responses that were highly reactive with both the autologous synthetic peptide and the full-length Luk toxin homologues. Thus, using a gene fragment- and phage display-based pipeline, we have identified and validated immunogenic B-cell epitopes that are cross-reactive between members of the pore-forming leukocidin family. This approach could be harnessed to identify novel epitopes for a much-needed S. aureus-protective subunit vaccine.


Subject(s)
Bacterial Proteins/immunology , Epitope Mapping , Epitopes, B-Lymphocyte/immunology , Exotoxins/immunology , Staphylococcus aureus/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Immunoglobulin G/blood , Mice , Peptide Library , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
20.
J Clin Invest ; 130(3): 1122-1127, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31873074

ABSTRACT

Staphylococcus aureus remains a leading cause of human infection. These infections frequently recur when the skin is a primary site of infection, especially in infants and children. In contrast, invasive staphylococcal disease is less commonly associated with reinfection, suggesting that tissue-specific mechanisms govern the development of immunity. Knowledge of how S. aureus manipulates protective immunity has been hampered by a lack of antigen-specific models to interrogate the T cell response. Using a chicken egg OVA-expressing S. aureus strain to analyze OVA-specific T cell responses, we demonstrated that primary skin infection was associated with impaired development of T cell memory. Conversely, invasive infection induced antigen-specific memory and protected against reinfection. This defect in adaptive immunity following skin infection was associated with a loss of DCs, attributable to S. aureus α-toxin (Hla) expression. Gene- and immunization-based approaches to protect against Hla during skin infection restored the T cell response. Within the human population, exposure to α-toxin through skin infection may modulate the establishment of T cell-mediated immunity, adversely affecting long-term protection. These studies prompt consideration that vaccination targeting S. aureus may be most effective if delivered prior to initial contact with the organism.


Subject(s)
Bacterial Toxins/immunology , Hemolysin Proteins/immunology , Immunity, Cellular , Immunologic Memory , Staphylococcal Skin Infections/immunology , Staphylococcus aureus/immunology , T-Lymphocytes/immunology , Animals , Bacterial Toxins/genetics , Hemolysin Proteins/genetics , Humans , Mice , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/pathology , Staphylococcal Vaccines/genetics , Staphylococcal Vaccines/immunology , Staphylococcal Vaccines/pharmacology , Staphylococcus aureus/genetics , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...