Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 673
Filtrar
1.
JCI Insight ; 9(13)2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38973612

RESUMO

Staphylococcus aureus is a major human pathogen. An effective anti-S. aureus vaccine remains elusive as the correlates of protection are ill-defined. Targeting specific T cell populations is an important strategy for improving anti-S. aureus vaccine efficacy. Potential bottlenecks that remain are S. aureus-induced immunosuppression and the impact this might have on vaccine-induced immunity. S. aureus induces IL-10, which impedes effector T cell responses, facilitating persistence during both colonization and infection. Thus, it was hypothesized that transient targeting of IL-10 might represent an innovative way to improve vaccine efficacy. In this study, IL-10 expression was elevated in the nares of persistent carriers of S. aureus, and this was associated with reduced systemic S. aureus-specific Th1 responses. This suggests that systemic responses are remodeled because of commensal exposure to S. aureus, which negatively implicates vaccine function. To provide proof of concept that targeting immunosuppressive responses during immunization may be a useful approach to improve vaccine efficacy, we immunized mice with T cell-activating vaccines in combination with IL-10-neutralizing antibodies. Blocking IL-10 during vaccination enhanced effector T cell responses and improved bacterial clearance during subsequent systemic and subcutaneous infection. Taken together, these results reveal a potentially novel strategy for improving anti-S. aureus vaccine efficacy.


Assuntos
Interleucina-10 , Infecções Estafilocócicas , Vacinas Antiestafilocócicas , Staphylococcus aureus , Interleucina-10/metabolismo , Interleucina-10/imunologia , Animais , Infecções Estafilocócicas/prevenção & controle , Infecções Estafilocócicas/imunologia , Vacinas Antiestafilocócicas/imunologia , Camundongos , Staphylococcus aureus/imunologia , Feminino , Camundongos Endogâmicos C57BL , Células Th1/imunologia , Imunização/métodos , Humanos , Anticorpos Neutralizantes/imunologia , Eficácia de Vacinas , Vacinação/métodos
2.
Hum Vaccin Immunother ; 20(1): 2360338, 2024 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-38857905

RESUMO

Staphylococcal Enterotoxin B (SEB), produced by Staphylococcus aureus (S. aureus), is a powerful superantigen that induces severe immune disruption and toxic shock syndrome (TSS) upon binding to MHC-II and TCR. Despite its significant impact on the pathogenesis of S. aureus, there are currently no specific therapeutic interventions available to counteract the mechanism of action exerted by this toxin. In this study, we have identified a human monoclonal antibody, named Hm0487, that specifically targets SEB by single-cell sequencing using PBMCs isolated from volunteers enrolled in a phase I clinical trial of the five-antigen S. aureus vaccine. X-ray crystallography studies revealed that Hm0487 exhibits high affinity for a linear B cell epitope in SEB (SEB138-147), which is located distantly from the site involved in the formation of the MHC-SEB-TCR ternary complex. Furthermore, in vitro studies demonstrated that Hm0487 significantly impacts the interaction of SEB with both receptors and the binding to immune cells, probably due to an allosteric effect on SEB rather than competing with receptors for binding sites. Moreover, both in vitro and in vivo studies validated that Hm0487 displayed efficient neutralizing efficacy in models of lethal shock and sepsis induced by either SEB or bacterial challenge. Our findings unveil an alternative mechanism for neutralizing the pathogenesis of SEB by Hm0487, and this antibody provides a novel strategy for mitigating both SEB-induced toxicity and S. aureus infection.


Assuntos
Anticorpos Monoclonais , Anticorpos Neutralizantes , Enterotoxinas , Enterotoxinas/imunologia , Enterotoxinas/antagonistas & inibidores , Humanos , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Animais , Cristalografia por Raios X , Staphylococcus aureus/imunologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/prevenção & controle , Epitopos de Linfócito B/imunologia , Camundongos , Choque Séptico/imunologia , Choque Séptico/prevenção & controle , Feminino , Leucócitos Mononucleares/imunologia , Vacinas Antiestafilocócicas/imunologia , Anticorpos Antibacterianos/imunologia , Superantígenos/imunologia
3.
Front Immunol ; 15: 1417220, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38868766

RESUMO

Staphylococcus aureus bacteremia causes significant morbidity and mortality. Treatment of staphylococcal infections is hindered by widespread antibiotic resistance, and attempts to develop an S. aureus vaccine have failed. Improved S. aureus treatment and infection prevention options require a deeper understanding of the correlates of protective immunity. CD4+ T cells have been identified as key orchestrators in the defense against S. aureus, but uncertainties persist regarding the subset, polarity, and breadth of the memory CD4+ T-cell pool required for protection. Here, using a mouse model of systemic S. aureus infection, we discovered that the breadth of bacterium-specific memory CD4+ T-cell pool is a critical factor for protective immunity against invasive S. aureus infections. Seeding mice with a monoclonal bacterium-specific circulating memory CD4+ T-cell population failed to protect against systemic S. aureus infection; however, the introduction of a polyclonal and polyfunctional memory CD4+ T-cell pool significantly reduced the bacterial burden. Our findings support the development of a multi-epitope T-cell-based S. aureus vaccine, as a strategy to mitigate the severity of S. aureus bacteremia.


Assuntos
Bacteriemia , Linfócitos T CD4-Positivos , Infecções Estafilocócicas , Staphylococcus aureus , Animais , Bacteriemia/imunologia , Bacteriemia/microbiologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia , Camundongos , Linfócitos T CD4-Positivos/imunologia , Células T de Memória/imunologia , Memória Imunológica , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Feminino , Vacinas Antiestafilocócicas/imunologia , Índice de Gravidade de Doença
4.
Immunol Cell Biol ; 102(5): 365-380, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38572664

RESUMO

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.


Assuntos
Proteínas de Bactérias , Infecções Estafilocócicas , Vacinas Antiestafilocócicas , Staphylococcus aureus , Superantígenos , Animais , Staphylococcus aureus/imunologia , Vacinas Antiestafilocócicas/imunologia , Superantígenos/imunologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/prevenção & controle , Camundongos , Proteínas de Bactérias/imunologia , Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/sangue , Feminino , Proteínas Recombinantes de Fusão/imunologia , Imunoglobulina G/imunologia , Imunoglobulina G/sangue , Estudos de Viabilidade , Vacinação , Antígenos de Bactérias/imunologia , Camundongos Endogâmicos BALB C , Adjuvantes Imunológicos
5.
Nat Commun ; 15(1): 3420, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38658531

RESUMO

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.


Assuntos
Infecções Estafilocócicas , Staphylococcus aureus , Animais , Infecções Estafilocócicas/prevenção & controle , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Camundongos , Staphylococcus aureus/imunologia , Coelhos , Vacinas Antiestafilocócicas/imunologia , Vacinas Antiestafilocócicas/administração & dosagem , Feminino , Staphylococcus aureus Resistente à Meticilina/imunologia , Acetilglucosamina/imunologia , Humanos , Epitopos/imunologia , Camundongos Endogâmicos BALB C
6.
Immunol Res ; 72(3): 490-502, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38383811

RESUMO

To increase the effectiveness of methicillin-resistant Staphylococcus aureus vaccines (MRSA), a new generation of immune system stimulating adjuvants is necessary, along with other adjuvants. In some vaccines, monophosphoryl lipid A (MPLA) as a toll-like receptor 4 agonist is currently used as an adjuvant or co-adjuvant. MPLA could increase the immune response and vaccine immunogenicity. The current investigation assessed the immunogenicity and anti-MRSA efficacy of recombinant autolysin formulated in MPLA and Alum as co-adjuvant/adjuvant. r-Autolysin was expressed and purified by Ni-NTA affinity chromatography and characterized by SDS-PAGE. Then, the vaccine candidate formulation in MPLAs and Alum was prepared. To investigate the immunogenic responses, total IgG, isotype (IgG1 and IgG2a) levels, and cytokines (IL-4, IL-12, TNF-α, and IFN-γ) profiles were evaluated by ELISA. Also, the bacterial burden in internal organs, opsonophagocytosis, survival rate, and pathobiology changes was compared among the groups. Results demonstrated that mice immunized with the r-Autolysin + Alum + MPLA Synthetic and r-Autolysin + Alum + MPLA Biologic led to increased levels of opsonic antibodies, IgG1, IgG2a isotype as well as increased levels of cytokines profiles, as compared with other experimental groups. More importantly, mice immunized with MPLA and r-Autolysin exhibited a decrease in mortality and bacterial burden, as compared with the control group. The highest level of survival was seen in the r-Autolysin + Alum + MPLA Synthetic group. We concluded that both MPLA forms, synthetic and biological, are reliable candidates for immune response improvement against MRSA infection.


Assuntos
Adjuvantes Imunológicos , Anticorpos Antibacterianos , Modelos Animais de Doenças , Imunoglobulina G , Lipídeo A , Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Vacinas Antiestafilocócicas , Animais , Lipídeo A/análogos & derivados , Lipídeo A/imunologia , Lipídeo A/administração & dosagem , Lipídeo A/farmacologia , Camundongos , Staphylococcus aureus Resistente à Meticilina/imunologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas/imunologia , Vacinas Antiestafilocócicas/administração & dosagem , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Adjuvantes Imunológicos/administração & dosagem , Feminino , Citocinas/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/imunologia , Desenvolvimento de Vacinas , Compostos de Alúmen/administração & dosagem , Camundongos Endogâmicos BALB C , Adjuvantes de Vacinas , Humanos
7.
Cell Rep Med ; 5(1): 101360, 2024 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-38232694

RESUMO

The failure of the Staphylococcus aureus (SA) IsdB vaccine trial can be explained by the recall of non-protective immune imprints from prior SA exposure. Here, we investigate natural human SA humoral imprints to understand their broader impact on SA immunizations. We show that antibody responses against SA cell-wall-associated antigens (CWAs) are non-opsonic, while antibodies against SA toxins are neutralizing. Importantly, the protective characteristics of the antibody imprints accurately predict the failure of corresponding vaccines against CWAs and support vaccination against toxins. In passive immunization platforms, natural anti-SA human antibodies reduce the efficacy of the human monoclonal antibodies suvratoxumab and tefibazumab, consistent with the results of their respective clinical trials. Strikingly, in the absence of specific humoral memory responses, active immunizations are efficacious in both naive and SA-experienced mice. Overall, our study points to a practical and predictive approach to evaluate and develop SA vaccines based on pre-existing humoral imprint characteristics.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Vacinas Antiestafilocócicas , Vacinas , Animais , Humanos , Camundongos , Imunização , Staphylococcus aureus , Ensaios Clínicos como Assunto
8.
Vaccine ; 42(6): 1247-1258, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38281900

RESUMO

Mastitis is an inflammation of the mammary gland commonly caused by bacteria or fungi. Staphylococcus aureus is a major bacterium that causes mastitis in dairy cows. Non-aureus staphylococci are also increasingly reported, with Staphylococcus chromogenes being the most common species. Current staphylococcal mastitis control programs are not fully effective, and treatment with antibiotics is not sustainable. Non-antibiotic sustainable control tools, such as effective vaccines, are critically needed. We previously developed S. aureus surface-associated proteins (SASP) and S. chromogenes surface-associated proteins (SCSP) vaccines that conferred partial protective effects. We hypothesized that vaccination with SASP or SCSP would reduce the incidence of S. aureus mastitis throughout the lactation period. The objective of this study was to evaluate the efficacy of SASP and SCSP vaccines against S. aureus and non-aureus staphylococcal mastitis under natural exposure over 300 days of lactation. Pregnant Holstein dairy cows (n = 45) were enrolled and assigned to receive SASP (n = 15) or SCSP (n = 16) vaccines or unvaccinated control (n = 14). Cows were vaccinated with 1.2 mg of SASP or SCSP with Emulsigen-D adjuvant. Control cows were injected with phosphate-buffered saline with Emulsigen-D adjuvant. Three vaccine injections were given subcutaneously at 60, 40, and 20 days before the expected calving. Booster vaccinations were given at 120 and 240 days in milk. Cows were monitored for mastitis at quarter and cow levels, staphylococcal mastitis incidence, changes in serum and milk anti-SASP and anti-SCSP antibody titers, bacterial counts in milk, adverse reactions, milk yield and milk somatic cells count over 300 days of lactation. The SCSP vaccine conferred a significant reduction in the incidence of staphylococcal mastitis. Milk and serum anti-SASP and anti-SCSP antibody titers were increased in the vaccinated cows compared to unvaccinated control cows. Anti-SASP and anti-SCSP antibody titers decreased at about 120 days in milk, indicating the duration of immunity of about four months. In conclusion, the SASP and SCSP vaccines conferred partial protection from natural infection.


Assuntos
Mastite Bovina , Infecções Estafilocócicas , Vacinas Antiestafilocócicas , Vacinas , Feminino , Gravidez , Bovinos , Animais , Humanos , Staphylococcus aureus , Infecções Estafilocócicas/prevenção & controle , Infecções Estafilocócicas/veterinária , Leite , Lactação , Proteínas de Membrana
9.
Microbiol Res ; 271: 127362, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36958134

RESUMO

Staphylococcus aureus (S. aureus) is a leading and crucial infectious threat to global public health due to the widespread emergence of antibiotic-resistant strains such as Methicillin-Resistant S. aureus (MRSA). MRSA infects immunocompromised patients and healthy individuals and has rapidly spread from the healthcare setting to the outside community. The development of flawless vaccines become a medical need worldwide against multi-drug resistant S. aureus. Therefore, protection by an immune-based strategy may provide valuable measures to contain the spread of invasive S. aureus infections. Several vaccine candidates have been prepared which are either in the preclinical phase or in the early clinical phase, whereas several candidates have failed to show a protective efficacy in human subjects. Currently, research is focusing on identifying novel vaccine formulations able to elicit potent humoral and cellular immune responses. Several approaches have also been made to the development of monoclonal or polyclonal antibodies for passive immunization to protect against S. aureus infections. In recent years, a multi-epitope vaccine has emerged as a novel platform for subunit vaccine design by using computational approaches. Therefore, in this review, we have summarized and discussed the mechanistic overview of different strategies used to develop potential vaccine candidates and passive interventions which are in different stages of clinical trials to fight multi-drug resistant S. aureus infections.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Vacinas Antiestafilocócicas , Humanos , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus , Desenvolvimento de Vacinas
11.
Cell Host Microbe ; 30(8): 1070-1071, 2022 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-35952643

RESUMO

In this issue of Cell Host & Microbe, Tsai et al. (2022) provide evidence that Staphylococcus aureus should be numbered among the pathogens for which original antigenic sin governs subsequent immune responses. Their approach may restore the utility of murine models in staphylococcal vaccine development.


Assuntos
Infecções Estafilocócicas , Vacinas Antiestafilocócicas , Vacinas , Animais , Humanos , Camundongos , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus
12.
Int J Mol Sci ; 23(2)2022 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-35055134

RESUMO

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.


Assuntos
Infecções Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas/uso terapêutico , Staphylococcus aureus/imunologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Humanos , Infecções Estafilocócicas/imunologia , Vacinas Antiestafilocócicas/imunologia , Vacinas Antiestafilocócicas/farmacologia , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidade , Desenvolvimento de Vacinas , Fatores de Virulência/genética , Fatores de Virulência/imunologia
13.
J Infect Dis ; 225(8): 1460-1470, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-33895843

RESUMO

Vaccines against Staphylococcus aureus have eluded researchers for >3 decades while the burden of staphylococcal diseases has increased. Early vaccine attempts mainly used rodents to characterize preclinical efficacy, and all subsequently failed in human clinical efficacy trials. More recently, leukocidin AB (LukAB) has gained interest as a vaccine antigen. We developed a minipig deep surgical wound infection model offering 3 independent efficacy readouts: bacterial load at the superficial and at the deep-seated surgical site, and dissemination of bacteria. Due to similarities with humans, minipigs are an attractive option to study novel vaccine candidates. With this model, we characterized the efficacy of a LukAB toxoid as vaccine candidate. Compared to control animals, a 3-log reduction of bacteria at the deep-seated surgical site was observed in LukAB-treated minipigs and dissemination of bacteria was dramatically reduced. Therefore, LukAB toxoids may be a useful addition to S. aureus vaccines and warrant further study.


Assuntos
Infecções Estafilocócicas , Vacinas Antiestafilocócicas , Animais , Carga Bacteriana , Proteínas de Bactérias , Leucocidinas , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus , Infecção da Ferida Cirúrgica/prevenção & controle , Suínos , Porco Miniatura , Vacinação
14.
J Appl Microbiol ; 132(2): 1422-1434, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34487603

RESUMO

AIMS: Staphylococcus aureus is an opportunistic pathogen of humans. No commercial vaccine is available to combat S. aureus infections. In this study, we have investigated the protective immune response generated by S. aureus non-covalently associated cell wall surface protein N-acetylmuramoyl-L-alanine amidase (AM) in combination with Alum (Al) and heat-killed S. aureus (hkSA) using murine models. METHODS AND RESULTS: BALB/c mice were immunized with increasing concentrations of AM antigen or hkSA to determine their optimum concentration for vaccination. Fifty micrograms of AM and hkSA each were found to generate maximum anti-AM IgG antibody production. BALB/c mice were immunized next with 50 µg of AM, 50 µg of hKSA and 1 mg Al vaccine formulation. Vaccine efficacy was validated by challenging immunized BALB/c mice with S. aureus Newman and three clinical methicillin-resistant S. aureus strains. AM-hkSA-Al-immunized mice generated high anti-AM IgG antibody response with IgG1 and IgG2b as the predominant immunoglobulin subtypes. Increased survival (60%-90%) with decreased clinical disease symptoms was observed in the vaccinated BALB/c mice group. A significantly lower bacterial load and decreased kidney abscess formation was observed following the challenge with S. aureus in the vaccinated BALB/c mice group. Furthermore, the efficacy of AM-hkSA-Al vaccine was also validated using C57 BL/6 and Swiss albino mice. CONCLUSIONS: Using murine infection models, we have demonstrated that AM-hkSA-Al vaccine would be effective in preventing S. aureus infections. SIGNIFICANCE AND IMPACT OF STUDY: AM-hkSA-Al vaccine elicited strong immune response and may be considered for future vaccine design against S. aureus infections.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Vacinas Antiestafilocócicas , Vacinas , Compostos de Alúmen , Amidoidrolases , Animais , Anticorpos Antibacterianos , Camundongos , Camundongos Endogâmicos BALB C , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus , Eficácia de Vacinas
15.
Infect Genet Evol ; 96: 105149, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34801756

RESUMO

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.


Assuntos
Staphylococcus aureus Resistente à Meticilina/fisiologia , Infecções Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas/imunologia , Vacinologia/métodos , Humanos , Simulação de Acoplamento Molecular , Vacinas de Subunidades Antigênicas/imunologia
16.
Biologicals ; 73: 8-15, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34376341

RESUMO

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.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Citocinas/imunologia , Epitopos/imunologia , Imunoglobulina G/imunologia , Staphylococcus aureus Resistente à Meticilina/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções Estafilocócicas/imunologia , Linfócitos T/imunologia
17.
Front Immunol ; 12: 705360, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34305945

RESUMO

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.


Assuntos
Infecções Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas , Staphylococcus aureus/imunologia , Desenvolvimento de Vacinas , Adjuvantes Imunológicos , Animais , Antígenos de Bactérias/imunologia , Ensaios Clínicos como Assunto , Vesículas Extracelulares/imunologia , Glicoconjugados/imunologia , Bactérias Gram-Negativas/imunologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Celular , Imunidade Humoral , Imunogenicidade da Vacina , Técnicas In Vitro , Camundongos , Modelos Animais , Vacinas Baseadas em Ácido Nucleico/imunologia , Periplasma/imunologia , Proteínas Recombinantes/imunologia , Vacinas Antiestafilocócicas/imunologia , Vacinas Antiestafilocócicas/uso terapêutico , Ciência Translacional Biomédica , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/imunologia
18.
Vet Immunol Immunopathol ; 238: 110290, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34217108

RESUMO

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.


Assuntos
Proteínas de Bactérias/imunologia , Mastite Bovina/prevenção & controle , Vacinas Antiestafilocócicas/imunologia , Staphylococcus/classificação , Adjuvantes Imunológicos , Animais , Anticorpos Antibacterianos/sangue , Bovinos , Feminino , Imunidade Celular , Imunidade Humoral , Imunoglobulina G/sangue , Mastite Bovina/microbiologia , Vacinação
19.
Front Immunol ; 12: 684823, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34122448

RESUMO

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.


Assuntos
Bacteriemia/imunologia , Toxinas Bacterianas/imunologia , Proteínas de Transporte de Cátions/imunologia , Epitopos de Linfócito B/imunologia , Proteínas Hemolisinas/imunologia , Epitopos Imunodominantes/imunologia , Infecções Estafilocócicas/prevenção & controle , Animais , Bacteriemia/prevenção & controle , Modelos Animais de Doenças , Feminino , Imunização Passiva , Imunoterapia Adotiva , Interferon gama/metabolismo , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Infecções Estafilocócicas/imunologia , Vacinas Antiestafilocócicas/administração & dosagem , Vacinas Antiestafilocócicas/imunologia
20.
Vaccine ; 39(23): 3179-3188, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-33962841

RESUMO

BACKGROUND: Military trainees are at increased risk for Staphylococcus aureus colonization and infection. Disease prevention strategies are needed, but a S. aureus vaccine does not currently exist. METHODS: We enrolled US Army Infantry trainees (Fort Benning, GA) in a phase 2, randomized, double-blind, placebo-controlled trial of NDV-3A, a vaccine containing a recombinant adhesin/invasion protein of Candida albicans that has structural similarity to the S. aureus protein clumping factor A. Study participants received one intramuscular dose of NDV-3A or placebo (adjuvant alone) within 72 h of arrival on base. Longitudinal nasal and oral (throat) swabs were collected throughout the 14-week Infantry training cycle. Safety, immunogenicity, and efficacy of NDV-3A against S. aureus nasal / oral acquisition were the endpoints. RESULTS: The NDV-3A candidate had minimal reactogenicity and elicited robust antigen-specific B- and T-cell responses. During the 56-day post-vaccination period, there was no difference in the incidence of S. aureus nasal acquisition between those who were randomized to receive NDV-3A vs. placebo (25.6% vs. 29.1%; vaccine efficacy [VE]: 12.1%; p = 0.31). In time-to-event analysis, there was no difference between study groups with respect to the S. aureus colonization-free interval (VE: 13%; p = 0.29). Similarly, the efficacy of NDV-3A against S. aureus oral acquisition was poor (VE: 2.4%; p = 0.52). CONCLUSIONS: A single dose of NDV-3A did not prevent nasal nor oral acquisition of S. aureus in a population of military trainees at high risk for colonization.


Assuntos
Militares , Infecções Estafilocócicas , Vacinas Antiestafilocócicas , Vacinas , Humanos , Imunogenicidade da Vacina , Infecções Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas/efeitos adversos , Staphylococcus aureus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA