Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22
Filtrer
1.
Front Immunol ; 15: 1373367, 2024.
Article de Anglais | MEDLINE | ID: mdl-38633244

RÉSUMÉ

Staphylococcus aureus is the leading cause of skin and soft tissue infections (SSTIs) in the U.S. as well as more serious invasive diseases, including bacteremia, sepsis, endocarditis, surgical site infections, osteomyelitis, and pneumonia. These infections are exacerbated by the emergence of antibiotic-resistant clinical isolates such as methicillin-resistant S. aureus (MRSA), highlighting the need for alternatives to antibiotics to treat bacterial infections. We have previously developed a multi-component toxoid vaccine (IBT-V02) in a liquid formulation with efficacy against multiple strains of Staphylococcus aureus prevalent in the industrialized world. However, liquid vaccine formulations are not compatible with the paucity of cold chain storage infrastructure in many low-to-middle income countries (LMICs). Furthermore, whether our IBT-V02 vaccine formulations are protective against S. aureus isolates from LMICs is unknown. To overcome these limitations, we developed lyophilized and spray freeze-dried formulations of IBT-V02 vaccine and demonstrated that both formulations had comparable biophysical attributes as the liquid formulation, including similar levels of toxin neutralizing antibodies and protective efficacy against MRSA infections in murine and rabbit models. To enhance the relevancy of our findings, we then performed a multi-dimensional screen of 83 S. aureus clinical isolates from LMICs (e.g., Democratic Republic of Congo, Palestine, and Cambodia) to rationally down-select strains to test in our in vivo models based on broad expression of IBT-V02 targets (i.e., pore-forming toxins and superantigens). IBT-V02 polyclonal antisera effectively neutralized toxins produced by the S. aureus clinical isolates from LMICs. Notably, the lyophilized IBT-V02 formulation exhibited significant in vivo efficacy in various preclinical infection models against the S. aureus clinical isolates from LMICs, which was comparable to our liquid formulation. Collectively, our findings suggested that lyophilization is an effective alternative to liquid vaccine formulations of our IBT-V02 vaccine against S. aureus infections, which has important implications for protection from S. aureus isolates from LMICs.


Sujet(s)
Staphylococcus aureus résistant à la méticilline , Infections à staphylocoques , Animaux , Souris , Lapins , Staphylococcus aureus , Pays en voie de développement , Antibactériens , Vaccins antibactériens , Toxoïdes
2.
Vaccine ; 41(45): 6625-6629, 2023 10 26.
Article de Anglais | MEDLINE | ID: mdl-37793976

RÉSUMÉ

AIM: Pneumococcal conjugate vaccines (PCV13, PCV15, PCV20) effectively target the capsular polysaccharides of the most common disease-causing Streptococcus pneumoniae serotypes. In this short communication, we analyzed healthy participants who received PCV13 and PCV15 vaccines as part of a recently concluded exploratory clinical trial and report antibody responses to the 13 shared serotypes (1, 3, 4, 5, 6A, 6B, 7F, 9V, 14, 18C, 19A, 19F, and 23F) as well as functional OPA responses to serotype 3. METHODS: Sera from 87 adult participants (18 through 49 years of age) randomized to receive either PCV13 or PCV15 were collected (n = 46 or n = 41, respectively), from 17 study centers in the US. IgG concentrations of the 13 shared serotypes and serotype 3-specific OPA titers were analyzed before and 1 month after vaccination using internally validated assays. RESULTS: At 1 month after vaccination, IgG GMCs of the 13 shared serotypes in PCV13 were similar to those for PCV15. Specifically, serotype 3 OPA GMTs and 95% CIs were similar 1 month after vaccination for PCV13 (62.9 [48.9, 80.9]) and PCV15 (71.1 [50.9, 99.2]). CONCLUSION: In healthy participants who received either PCV13 or PCV15, similar serotype-specific responses were observed between all shared serotypes when a uniform validated internal assay was used. Of note, data from this study suggest that both vaccines induce similar functional antibody responses against pneumococcal serotype 3.


Sujet(s)
Infections à pneumocoques , Streptococcus pneumoniae , Adulte , Humains , Anticorps antibactériens , Immunoglobuline G , Infections à pneumocoques/prévention et contrôle , Vaccins antipneumococciques , Sérogroupe , Vaccins conjugués , Adolescent , Jeune adulte , Adulte d'âge moyen
3.
Front Immunol ; 12: 624310, 2021.
Article de Anglais | MEDLINE | ID: mdl-33777005

RÉSUMÉ

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.


Sujet(s)
Réinfection/prévention et contrôle , Infections cutanées à staphylocoques/prévention et contrôle , Vaccins antistaphylococciques/pharmacologie , Staphylococcus aureus/immunologie , Animaux , Anticorps antibactériens/sang , Anticorps neutralisants/sang , Modèles animaux de maladie humaine , Femelle , Immunisation , Immunogénicité des vaccins , Mâle , Souris de lignée BALB C , Lapins , Réinfection/immunologie , Réinfection/microbiologie , Infections cutanées à staphylocoques/immunologie , Infections cutanées à staphylocoques/microbiologie , Vaccins antistaphylococciques/immunologie
4.
Front Immunol ; 12: 621754, 2021.
Article de Anglais | MEDLINE | ID: mdl-33717122

RÉSUMÉ

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.


Sujet(s)
Lymphocytes T CD4+/immunologie , Staphylococcus aureus résistant à la méticilline/physiologie , Infections à staphylocoques/immunologie , Anatoxine staphylococcique/immunologie , Vaccins antistaphylococciques/immunologie , Animaux , Anticorps antibactériens/sang , Production d'anticorps , Anticorps neutralisants à large spectre/sang , Immunité hétérologue , Immunogénicité des vaccins , Activation des lymphocytes , Macaca mulatta , Superantigènes/immunologie , Vaccination
5.
J Exp Med ; 217(12)2020 12 07.
Article de Anglais | MEDLINE | ID: mdl-32766723

RÉSUMÉ

The Nck-associated protein 1-like (NCKAP1L) gene, alternatively called hematopoietic protein 1 (HEM-1), encodes a hematopoietic lineage-specific regulator of the actin cytoskeleton. Nckap1l-deficient mice have anomalies in lymphocyte development, phagocytosis, and neutrophil migration. Here we report, for the first time, NCKAP1L deficiency cases in humans. In two unrelated patients of Middle Eastern origin, recessive mutations in NCKAP1L abolishing protein expression led to immunodeficiency, lymphoproliferation, and hyperinflammation with features of hemophagocytic lymphohistiocytosis. Immunophenotyping showed an inverted CD4/CD8 ratio with a major shift of both CD4+ and CD8+ cells toward memory compartments, in line with combined RNA-seq/proteomics analyses revealing a T cell exhaustion signature. Consistent with the core function of NCKAP1L in the reorganization of the actin cytoskeleton, patients' T cells displayed impaired early activation, immune synapse morphology, and leading edge formation. Moreover, knockdown of nckap1l in zebrafish led to defects in neutrophil migration. Hence, NCKAP1L mutations lead to broad immune dysregulation in humans, which could be classified within actinopathies.


Sujet(s)
Déficits immunitaires/complications , Inflammation/complications , Syndromes lymphoprolifératifs/complications , Protéines membranaires/métabolisme , Actines/métabolisme , Animaux , Dégranulation cellulaire , Prolifération cellulaire , Enfant , Cytotoxicité immunologique , Famille , Femelle , Homozygote , Humains , Déficits immunitaires/immunologie , Synapses immunologiques/métabolisme , Nourrisson , Inflammation/immunologie , Inflammation/anatomopathologie , Activation des lymphocytes/immunologie , Syndromes lymphoprolifératifs/immunologie , Mâle , Protéines membranaires/composition chimique , Protéines membranaires/déficit , Protéines membranaires/génétique , Mutation/génétique , Pedigree , Phénotype , Syndrome , Danio zébré
6.
ACS Appl Bio Mater ; 3(5): 3187-3195, 2020 May 18.
Article de Anglais | MEDLINE | ID: mdl-33880435

RÉSUMÉ

Resiquimod or R848 (RSQD) is a Toll-like receptor (TLR) 7/8 agonist which shows promise as vaccine adjuvant due to its potential to promote highly desirable cellular immunity. The development of this small molecule in the field to date has been largely impeded by its rapid in vivo clearance and lack of association with vaccine antigens. Here, we report a multimeric TLR 7/8 construct of nano-scale size, which results from a spontaneous self-assembly of RSQD with a water-soluble clinical-stage polymer - poly[di(carboxylatophenoxy)phosphazene] (PCPP). The formation of ionically paired construct (PCPP-R) and a ternary complex, which also includes Hepatitis C virus (HCV) antigen, has been demonstrated by dynamic lights scattering (DLS), turbidimetry, fluorescence spectroscopy, asymmetric flow field flow fractionation (AF4), and 1H NMR spectroscopy methods. The resulting supramolecular assembly PCPP-R enabled superior immunostimulation in cellular assays (mouse macrophage reporter cell line) and displayed improved in vitro hemocompatibility (human erythrocytes). In vivo studies demonstrated that PCPP-R adjuvanted HCV formulation induced higher serum neutralization titers in BALB/c mice and shifted the response towards desirable cellular immunity, as evaluated by antibody isotype ratio (IgG2a/IgG1) and ex vivo analysis of cytokine secreting splenocytes (higher levels of interferon gamma (IFN-γ) single and tumor necrosis factor alpha (TNF-α)/IFN-γ double producing cells). The non-covalent multimerization approach stands in contrast to previously suggested RSQD delivery methods, which involve covalent conjugation or encapsulation, and offers a flexible methodology that can be potentially integrated with other parenterally administered drugs.

7.
Toxins (Basel) ; 11(6)2019 06 14.
Article de Anglais | MEDLINE | ID: mdl-31207937

RÉSUMÉ

Staphylococcus aureus (SA) infections cause high mortality and morbidity in humans. Being central to its pathogenesis, S. aureus thwarts the host defense by secreting a myriad of virulence factors, including bicomponent, pore-forming leukotoxins. While all vaccine development efforts that aimed at achieving opsonophagocytic killing have failed, targeting virulence by toxoid vaccines represents a novel approach to preventing mortality and morbidity that are caused by SA. The recently discovered leukotoxin LukAB kills human phagocytes and monocytes and it is present in all known S. aureus clinical isolates. While using a structure-guided approach, we generated a library of mutations that targeted functional domains within the LukAB heterodimer to identify attenuated toxoids as potential vaccine candidates. The mutants were evaluated based on expression, solubility, yield, biophysical properties, cytotoxicity, and immunogenicity, and several fully attenuated LukAB toxoids that were capable of eliciting high neutralizing antibody titers were identified. Rabbit polyclonal antibodies against the lead toxoid candidate provided potent neutralization of LukAB. While the neutralization of LukAB alone was not sufficient to fully suppress leukotoxicity in supernatants of S. aureus USA300 isolates, a combination of antibodies against LukAB, α-toxin, and Panton-Valentine leukocidin completely neutralized the cytotoxicity of these strains. These data strongly support the inclusion of LukAB toxoids in a multivalent toxoid vaccine for the prevention of S. aureus disease.


Sujet(s)
Protéines bactériennes/immunologie , Vaccins antibactériens , Leucocidine/immunologie , Infections à staphylocoques/prévention et contrôle , Toxoïdes/immunologie , Animaux , Protéines bactériennes/génétique , Survie cellulaire , Escherichia coli/génétique , Femelle , Cellules HL-60 , Humains , Leucocidine/génétique , Souris de lignée ICR , Monocytes , Cellules THP-1 , Toxoïdes/génétique
8.
J Mol Biol ; 431(21): 4354-4367, 2019 10 04.
Article de Anglais | MEDLINE | ID: mdl-30928493

RÉSUMÉ

To develop an antibody (Ab) therapeutic against staphylococcal enterotoxin B (SEB), a potential incapacitating bioterrorism agent and a major cause of food poisoning, we developed a "class T" anti-SEB neutralizing Ab (GC132) targeting an epitope on SEB distinct from that of previously developed "class M" Abs. A systematic engineering approach was applied to affinity-mature Ab GC132 to yield an optimized therapeutic candidate (GC132a) with sub-nanomolar binding affinity. Mapping of the binding interface by hydrogen-deuterium exchange coupled to mass spectrometry revealed that the class T epitope on SEB overlapped with the T-cell receptor binding site, whereas other evidence suggested that the class M epitope overlapped with the binding site for the major histocompatibility complex. In the IgG format, GC132a showed ∼50-fold more potent toxin-neutralizing efficacy than the best class M Ab in vitro, and fully protected mice from lethal challenge in a toxic shock post-exposure model. We also engineered bispecific Abs (bsAbs) that bound tetravalently by utilizing two class M binding sites and two class T binding sites. The bsAbs displayed enhanced toxin neutralization efficacy compared with the respective monospecific Ab subunits as well as a mixture of the two, indicating that enhanced efficacy was due to heterotypic tetravalent binding to two non-overlapping epitopes on SEB. Together, these results suggest that class T anti-SEB Ab GC132a is an excellent candidate for clinical development and for bsAb engineering.


Sujet(s)
Anticorps antibactériens/métabolisme , Anticorps neutralisants/métabolisme , Récepteurs aux antigènes des cellules T/métabolisme , Animaux , Anticorps bispécifiques/métabolisme , Techniques d'exposition à la surface cellulaire , Entérotoxines/métabolisme , Humains , Spectrométrie de masse , Modèles biologiques , Ingénierie des protéines/méthodes
9.
Sci Rep ; 9(1): 3279, 2019 03 01.
Article de Anglais | MEDLINE | ID: mdl-30824769

RÉSUMÉ

Superantigens (SAgs) play a major role in the pathogenesis of Staphylococcus aureus and are associated with several diseases, including food poisoning, bacterial arthritis, and toxic shock syndrome. Monoclonal antibodies to these SAgs, primarily TSST-1, SEB and SEA have been shown to provide protection in animal studies and to reduce clinical severity in bacteremic patients. Here we quantify the pre-existing antibodies against SAgs in many human plasma and IVIG samples and demonstrate that in a major portion of the population these antibody titers are suboptimal and IVIG therapy only incrementally elevates the anti-SAg titers. Our in vitro neutralization studies show that a combination of antibodies against SEA, SEB,and TSST-1 can provide broad neutralization of staphylococcal SAgs. We report a single fusion protein (TBA225) consisting of the toxoid versions of TSST-1, SEB and SEA and demonstrate its immunogenicity and protective efficacy in a mouse model of toxic shock. Antibodies raised against this fusion vaccine provide broad neutralization of purified SAgs and culture supernatants of multiple clinically relevant S. aureus strains. Our data strongly supports the use of this fusion protein as a component of an anti-virulence based multivalent toxoid vaccine against S. aureus disease.


Sujet(s)
Entérotoxines/toxicité , Protéines de fusion recombinantes/pharmacologie , Anatoxine staphylococcique/pharmacologie , Staphylococcus aureus , Superantigènes/toxicité , Animaux , Entérotoxines/composition chimique , Entérotoxines/génétique , Entérotoxines/immunologie , Femelle , Humains , Souris , Souris de lignée BALB C , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/immunologie , Anatoxine staphylococcique/composition chimique , Anatoxine staphylococcique/génétique , Anatoxine staphylococcique/immunologie , Staphylococcus aureus/composition chimique , Staphylococcus aureus/génétique , Staphylococcus aureus/immunologie , Superantigènes/composition chimique , Superantigènes/génétique , Superantigènes/immunologie
10.
Article de Anglais | MEDLINE | ID: mdl-30782986

RÉSUMÉ

Staphylococcal enterotoxin B (SEB) is a protein exotoxin found on the cell surface of Staphylococcus aureus that is the source for multiple pathologies in humans. When purified and concentrated in aerosol form, SEB can cause an acute and often fatal intoxication and thus is considered a biological threat agent. There are currently no vaccines or treatments approved for human use. Studies with rodent models of SEB intoxication show that antibody therapy may be a promising treatment strategy; however, many have used antibodies only prophylactically or well before any clinical signs of intoxication are apparent. We assessed and compared the protective efficacies of two monoclonal antibodies, Ig121 and c19F1, when administered after aerosol exposure in a uniformly lethal nonhuman primate model of SEB intoxication. Rhesus macaques were challenged using small-particle aerosols of SEB and then were infused intravenously with a single dose of either Ig121 or c19F1 (10 mg/kg of body weight) at either 0.5, 2, or 4 h postexposure. Onset of clinical signs and hematological and cytokine response in untreated controls confirmed the acute onset and potency of the toxin used in the challenge. All animals administered either Ig121 or c19F1 survived SEB challenge, whereas the untreated controls succumbed to SEB intoxication 30 to 48 h postexposure. These results represent the successful therapeutic in vivo protection by two investigational drugs against SEB in a severe nonhuman primate disease model and punctuate the therapeutic value of monoclonal antibodies when faced with treatment options for SEB-induced toxicity in a postexposure setting.


Sujet(s)
Aérosols/toxicité , Anticorps monoclonaux/usage thérapeutique , Entérotoxines/toxicité , Animaux , Test ELISA , Macaca mulatta
11.
FEMS Microbiol Lett ; 365(18)2018 09 01.
Article de Anglais | MEDLINE | ID: mdl-30084923

RÉSUMÉ

Spores of Bacillus subtilis are encased in a protein coat composed of ∼80 different proteins. Recently, we reconstituted the basement layer of the coat, composed of two structural proteins (SpoVM and SpoIVA) around spore-sized silica beads encased in a lipid bilayer, to create synthetic spore-like particles termed 'SSHELs'. We demonstrated that SSHELs could display thousands of copies of proteins and small molecules of interest covalently linked to SpoIVA. In this study, we investigated the efficacy of SSHELs in delivering vaccines. We show that intramuscular vaccination of mice with undecorated one micron-diameter SSHELs elicited an antibody response against SpoIVA. We further demonstrate that SSHELs covalently modified with a catalytically inactivated staphylococcal alpha toxin variant (HlaH35L), without an adjuvant, resulted in improved protection against Staphylococcus aureus infection in a bacteremia model as compared to vaccination with the antigen alone. Although vaccination with either HlaH35L or HlaH35L conjugated to SSHELs similarly elicited the production of neutralizing antibodies to Hla, we found that a subset of memory T cells was differentially activated when the antigen was delivered on SSHELs. We propose that the particulate nature of SSHELs elicits a more robust immune response to the vaccine that results in superior protection against subsequent S. aureus infection.


Sujet(s)
Toxines bactériennes/immunologie , Vecteurs de médicaments/administration et posologie , Hémolysines/immunologie , Infections à staphylocoques/prévention et contrôle , Vaccins antistaphylococciques/immunologie , Animaux , Anticorps antibactériens/sang , Anticorps neutralisants/sang , Bactériémie/prévention et contrôle , Protéines bactériennes/génétique , Toxines bactériennes/génétique , Modèles animaux de maladie humaine , Hémolysines/génétique , Injections musculaires , Souris , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/immunologie , Vaccins antistaphylococciques/administration et posologie , Vaccins antistaphylococciques/génétique , Sous-populations de lymphocytes T/immunologie , Résultat thérapeutique , Vaccins sous-unitaires/administration et posologie , Vaccins sous-unitaires/génétique , Vaccins sous-unitaires/immunologie , Vaccins synthétiques/administration et posologie , Vaccins synthétiques/génétique , Vaccins synthétiques/immunologie
12.
J Exp Med ; 215(4): 1079-1090, 2018 04 02.
Article de Anglais | MEDLINE | ID: mdl-29490936

RÉSUMÉ

Appropriate regulation of IL-17 production in the host can mean the difference between effective control of pathogens and uncontrolled inflammation that causes tissue damage. Investigation of conventional CD4+ T cells (Th17 cells) has yielded invaluable insights into IL-17 function and its regulation. More recently, we and others reported production of IL-17 from innate αß+ T cell populations, which was shown to occur primarily via IL-23R signaling through the transcription factor STAT-3. In our current study, we identify promyelocytic leukemia zinc finger (PLZF)-expressing iNKT, CD4-/CD8+, and CD4-/CD8- (DN) αß+T cells, which produce IL-17 in response to TCR and IL-1 receptor ligation independently of STAT-3 signaling. Notably, this noncanonical pathway of IL-17 production may be important in mucosal defense and is by itself sufficient to control pathogenic Staphylococcus aureus infection at the ocular surface.


Sujet(s)
Infections de l'oeil/immunologie , Infections de l'oeil/anatomopathologie , Immunité innée , Interleukine-17/biosynthèse , Récepteur lymphocytaire T antigène, alpha-bêta/métabolisme , Facteur de transcription STAT-3/métabolisme , Animaux , Mémoire immunologique , Interleukines/métabolisme , Souris de lignée C57BL , Souris knockout , Muqueuse/immunologie , Muqueuse/microbiologie , Membre-3 du groupe F de la sous-famille-1 de récepteurs nucléaires/métabolisme , Phosphorylation , Protéine à doigts de zinc de la leucémie promyélocytaire/métabolisme , Transduction du signal , Staphylococcus aureus/physiologie , Lymphocytes T/métabolisme , Cellules Th17/métabolisme , Thymus (glande)/métabolisme
13.
J Infect Dis ; 215(8): 1231-1239, 2017 04 15.
Article de Anglais | MEDLINE | ID: mdl-28329242

RÉSUMÉ

Multiple candidate vaccines against Staphylococcus aureus infections have failed in clinical trials. Analysis of a recent prematurely halted vaccine trial revealed increased mortality rates among vaccine recipients in whom postsurgical S. aureus infection developed, emphasizing the potential for induction of detrimental immune responses and the need to better understand the requirements for protective immunity against S. aureus. These failures of single-antigen vaccines have prompted ongoing development of multicomponent vaccines to target the multitude of S. aureus virulence factors. In the current study, we used lethally irradiated S. aureus as a model multicomponent vaccine and showed that vaccination of mice decreased survival in a bacteremia challenge model. These deleterious effects were due to a CD4 T-cell-dependent interferon γ response and could be prevented by inhibiting development of this response during vaccination. Our results identify the potential for vaccination to induce pathological immune responses, and they have implications for recent vaccine failures and the design of future staphylococcal vaccines.


Sujet(s)
Interféron gamma/immunologie , Infections à staphylocoques/prévention et contrôle , Vaccins antistaphylococciques/administration et posologie , Vaccins antistaphylococciques/effets indésirables , Lymphocytes auxiliaires Th1/immunologie , Animaux , Bactériémie/prévention et contrôle , Femelle , Staphylococcus aureus résistant à la méticilline/effets des radiations , Souris , Souris de lignée C57BL , Souris knockout , Infections à staphylocoques/immunologie
14.
Curr Top Microbiol Immunol ; 409: 419-439, 2017.
Article de Anglais | MEDLINE | ID: mdl-26919865

RÉSUMÉ

A complex interplay between host and bacterial factors allows Staphylococcus aureus to occupy its niche as a human commensal and a major human pathogen. The role of neutrophils as a critical component of the innate immune response against S. aureus, particularly for control of systemic infection, has been established in both animal models and in humans with acquired and congenital neutrophil dysfunction. The role of the adaptive immune system is less clear. Although deficiencies in adaptive immunity do not result in the marked susceptibility to S. aureus infection that neutrophil dysfunction imparts, emerging evidence suggests both T cell- and B cell-mediated adaptive immunity can influence host susceptibility and control of S. aureus. The contribution of adaptive immunity depends on the context and site of infection and can be either beneficial or detrimental to the host. Furthermore, S. aureus has evolved mechanisms to manipulate adaptive immune responses to its advantage. In this chapter, we will review the evidence for the role of adaptive immunity during S. aureus infections. Further elucidation of this role will be important to understand how it influences susceptibility to infection and to appropriately design vaccines that elicit adaptive immune responses to protect against subsequent infections.


Sujet(s)
Staphylococcus aureus , Immunité acquise , Animaux , Humains , Immunité innée , Infections à staphylocoques
15.
PLoS Pathog ; 11(12): e1005293, 2015 Dec.
Article de Anglais | MEDLINE | ID: mdl-26679537

RÉSUMÉ

Candida is the most common human fungal pathogen and causes systemic infections that require neutrophils for effective host defense. Humans deficient in the C-type lectin pathway adaptor protein CARD9 develop spontaneous fungal disease that targets the central nervous system (CNS). However, how CARD9 promotes protective antifungal immunity in the CNS remains unclear. Here, we show that a patient with CARD9 deficiency had impaired neutrophil accumulation and induction of neutrophil-recruiting CXC chemokines in the cerebrospinal fluid despite uncontrolled CNS Candida infection. We phenocopied the human susceptibility in Card9-/- mice, which develop uncontrolled brain candidiasis with diminished neutrophil accumulation. The induction of neutrophil-recruiting CXC chemokines is significantly impaired in infected Card9-/- brains, from both myeloid and resident glial cellular sources, whereas cell-intrinsic neutrophil chemotaxis is Card9-independent. Taken together, our data highlight the critical role of CARD9-dependent neutrophil trafficking into the CNS and provide novel insight into the CNS fungal susceptibility of CARD9-deficient humans.


Sujet(s)
Protéines adaptatrices de signalisation CARD/immunologie , Candidose/immunologie , Infections du système nerveux central/immunologie , Déficits immunitaires/immunologie , Infiltration par les neutrophiles/immunologie , Animaux , Technique de Western , Protéines adaptatrices de signalisation CARD/déficit , Femelle , Cytométrie en flux , Humains , Déficits immunitaires/microbiologie , Souris , Souris knockout
16.
PLoS One ; 8(6): e65384, 2013.
Article de Anglais | MEDLINE | ID: mdl-23762356

RÉSUMÉ

Previous efforts towards S. aureus vaccine development have largely focused on cell surface antigens to induce opsonophagocytic killing aimed at providing sterile immunity, a concept successfully applied to other Gram-positive pathogens such as Streptococcus pneumoniae. However, these approaches have largely failed, possibly in part due to the remarkable diversity of the staphylococcal virulence factors such as secreted immunosuppressive and tissue destructive toxins. S. aureus produces several pore-forming toxins including the single subunit alpha hemolysin as well as bicomponent leukotoxins such as Panton-Valentine leukocidin (PVL), gamma hemolysins (Hlg), and LukED. Here we report the generation of highly attenuated mutants of PVL subunits LukS-PV and LukF-PV that were rationally designed, based on an octameric structural model of the toxin, to be deficient in oligomerization. The attenuated subunit vaccines were highly immunogenic and showed significant protection in a mouse model of S. aureus USA300 sepsis. Protection against sepsis was also demonstrated by passive transfer of rabbit immunoglobulin raised against LukS-PV. Antibodies to LukS-PV inhibited the homologous oligomerization of LukS-PV with LukF-PV as well heterologous oligomerization with HlgB. Importantly, immune sera from mice vaccinated with the LukS mutant not only inhibited the PMN lytic activity produced by the PVL-positive USA300 but also blocked PMN lysis induced by supernatants of PVL-negative strains suggesting a broad protective activity towards other bicomponent toxins. These findings strongly support the novel concept of an anti-virulence, toxin-based vaccine intended for prevention of clinical S. aureus invasive disease, rather than achieving sterile immunity. Such a multivalent vaccine may include attenuated leukotoxins, alpha hemolysin, and superantigens.


Sujet(s)
Bactériémie/immunologie , Bactériémie/prévention et contrôle , Protéines bactériennes/immunologie , Leucocidine/immunologie , Staphylococcus aureus/immunologie , Vaccins atténués/immunologie , Vaccins sous-unitaires/immunologie , Adjuvants immunologiques/pharmacologie , Acides aminés , Animaux , Anticorps neutralisants/pharmacologie , Bactériémie/microbiologie , Charge bactérienne/effets des médicaments et des substances chimiques , Protéines bactériennes/composition chimique , Toxines bactériennes/immunologie , Réactions croisées/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Conception de médicament , Exotoxines/immunologie , Immunisation , Leucocidine/composition chimique , Souris , Souris de lignée BALB C , Protéines mutantes/composition chimique , Protéines mutantes/métabolisme , Mutation/génétique , Multimérisation de protéines/effets des médicaments et des substances chimiques , Stabilité protéique/effets des médicaments et des substances chimiques , Dépliement des protéines/effets des médicaments et des substances chimiques , Similitude de séquences d'acides aminés , Staphylococcus aureus/effets des médicaments et des substances chimiques , Température , Vaccins atténués/composition chimique , Vaccins sous-unitaires/composition chimique
17.
J Infect Dis ; 206(6): 915-23, 2012 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-22807524

RÉSUMÉ

BACKGROUND: Staphylococcus aureus has numerous virulence factors, including exotoxins that may increase the severity of infection. This study was aimed at assessing whether preexisting antibodies to S. aureus toxins are associated with a lower risk of sepsis in adults with S. aureus infection complicated by bacteremia. METHODS: We prospectively identified adults with S. aureus infection from 4 hospitals in Baltimore, MD, in 2009­2011. We obtained serum samples from prior to or at presentation of S. aureus bacteremia to measure total immunoglobulin G (IgG) and IgG antibody levels to 11 S. aureus exotoxins. Bacterial isolates were tested for the genes encoding S. aureus exotoxins using polymerase chain reaction (PCR). RESULTS: One hundred eligible subjects were included and 27 of them developed sepsis. When adjusted for total IgG levels and stratified for the presence of toxin in the infecting isolate as appropriate, the risk of sepsis was significantly lower in those patients with higher levels of IgG against α-hemolysin (Hla), δ-hemolysin (Hld), Panton Valentine leukocidin (PVL), staphylococcal enterotoxin C-1 (SEC-1), and phenol-soluble modulin α3 (PSM-α3). CONCLUSIONS: Our results suggest that higher antibody levels against Hla, Hld, PVL, SEC-1, and PSM-α3 may protect against sepsis in patients with invasive S. aureus infections.


Sujet(s)
Anticorps antibactériens/sang , Exotoxines/immunologie , Sepsie/microbiologie , Infections à staphylocoques/immunologie , Staphylococcus aureus/immunologie , Adulte , Sujet âgé , Protéines bactériennes/immunologie , Études de cohortes , Femelle , Hospitalisation , Humains , Immunoglobuline G/sang , Mâle , Adulte d'âge moyen , Odds ratio , Facteurs de risque , Sepsie/immunologie , Infections à staphylocoques/microbiologie , Staphylococcus aureus/métabolisme
18.
PLoS One ; 7(6): e38567, 2012.
Article de Anglais | MEDLINE | ID: mdl-22701668

RÉSUMÉ

Staphylococcus aureus (S. aureus) is a human pathogen associated with skin and soft tissue infections (SSTI) and life threatening sepsis and pneumonia. Efforts to develop effective vaccines against S. aureus have been largely unsuccessful, in part due to the variety of virulence factors produced by this organism. S. aureus alpha-hemolysin (Hla) is a pore-forming toxin expressed by most S. aureus strains and reported to play a key role in the pathogenesis of SSTI and pneumonia. Here we report a novel recombinant subunit vaccine candidate for Hla, rationally designed based on the heptameric crystal structure. This vaccine candidate, denoted AT-62aa, was tested in pneumonia and bacteremia infection models using S. aureus strain Newman and the pandemic strain USA300 (LAC). Significant protection from lethal bacteremia/sepsis and pneumonia was observed upon vaccination with AT-62aa along with a Glucopyranosyl Lipid Adjuvant-Stable Emulsion (GLA-SE) that is currently in clinical trials. Passive transfer of rabbit immunoglobulin against AT-62aa (AT62-IgG) protected mice against intraperitoneal and intranasal challenge with USA300 and produced significant reduction in bacterial burden in blood, spleen, kidney, and lungs. Our Hla-based vaccine is the first to be reported to reduce bacterial dissemination and to provide protection in a sepsis model of S. aureus infection. AT62-IgG and sera from vaccinated mice effectively neutralized the toxin in vitro and AT62-IgG inhibited the formation of Hla heptamers, suggesting antibody-mediated neutralization as the primary mechanism of action. This remarkable efficacy makes this Hla-based vaccine a prime candidate for inclusion in future multivalent S. aureus vaccine. Furthermore, identification of protective epitopes within AT-62aa could lead to novel immunotherapy for S. aureus infection.


Sujet(s)
Bactériémie/prévention et contrôle , Toxines bactériennes/immunologie , Hémolysines/immunologie , Modèles moléculaires , Pneumopathie infectieuse/prévention et contrôle , Vaccins antistaphylococciques , Staphylococcus aureus/immunologie , Vaccins synthétiques , Animaux , Bactériémie/immunologie , Toxines bactériennes/composition chimique , Amorces ADN/génétique , Test ELISA , Femelle , Hémolysines/composition chimique , Immunoglobuline G/immunologie , Immunothérapie/méthodes , Souris , Souris de lignée BALB C , Tests de neutralisation , Plasmides/génétique , Pneumopathie infectieuse/immunologie , Lapins
19.
J Biol Chem ; 287(30): 25203-15, 2012 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-22645125

RÉSUMÉ

Staphylococcal enterotoxin B (SEB) is a potent toxin that can cause toxic shock syndrome and act as a lethal and incapacitating agent when used as a bioweapon. There are currently no vaccines or immunotherapeutics available against this toxin. Using phage display technology, human antigen-binding fragments (Fabs) were selected against SEB, and proteins were produced in Escherichia coli cells and characterized for their binding affinity and their toxin neutralizing activity in vitro and in vivo. Highly protective Fabs were converted into full-length IgGs and produced in mammalian cells. Additionally, the production of anti-SEB antibodies was explored in the Nicotiana benthamiana plant expression system. Affinity maturation was performed to produce optimized lead anti-SEB antibody candidates with subnanomolar affinities. IgGs produced in N. benthamiana showed characteristics comparable with those of counterparts produced in mammalian cells. IgGs were tested for their therapeutic efficacy in the mouse toxic shock model using different challenge doses of SEB and a treatment with 200 µg of IgGs 1 h after SEB challenge. The lead candidates displayed full protection from lethal challenge over a wide range of SEB challenge doses. Furthermore, mice that were treated with anti-SEB IgG had significantly lower IFNγ and IL-2 levels in serum compared with mock-treated mice. In summary, these anti-SEB monoclonal antibodies represent excellent therapeutic candidates for further preclinical and clinical development.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Entérotoxines/antagonistes et inhibiteurs , Fragments Fab d'immunoglobuline/pharmacologie , Choc septique/thérapie , Animaux , Anticorps monoclonaux/génétique , Anticorps monoclonaux/immunologie , Affinité des anticorps/génétique , Entérotoxines/immunologie , Entérotoxines/toxicité , Cellules HEK293 , Humains , Fragments Fab d'immunoglobuline/génétique , Fragments Fab d'immunoglobuline/immunologie , Interféron gamma/génétique , Interféron gamma/immunologie , Interleukine-2/génétique , Interleukine-2/immunologie , Souris , Souris de lignée BALB C , Protéines recombinantes/génétique , Protéines recombinantes/immunologie , Protéines recombinantes/pharmacologie , Choc septique/induit chimiquement , Choc septique/génétique , Choc septique/immunologie , Facteurs temps , Nicotiana/génétique
20.
J Biomol Struct Dyn ; 28(1): 1-12, 2010 Aug.
Article de Anglais | MEDLINE | ID: mdl-20476791

RÉSUMÉ

Panton-Valentine leukocidin (PVL) is a bipartite toxin that plays an important role in the pathogenesis of methicillin-resistant Staphylococcus aureus. Recent clinical data suggest a correlation between PVL and severe cases of S. aureus pneumonia. A clear understanding of the structure and function of PVL is critical to the development of novel, effective treatments. Here, we report an all-atom model of the macromolecular structure of Panton-Valentine leukocidin in its octameric, pre-pore conformation that confirms and extends our understanding of the toxin's mechanism of action.


Sujet(s)
Toxines bactériennes/composition chimique , Exotoxines/composition chimique , Leucocidine/composition chimique , Staphylococcus aureus résistant à la méticilline/composition chimique , Modèles moléculaires , Conformation des protéines , Toxines bactériennes/génétique , Cristallographie aux rayons X , Exotoxines/génétique , Humains , Liaison hydrogène , Leucocidine/génétique , Données de séquences moléculaires , Mutation
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...