Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 639
Filtrar
1.
Talanta ; 274: 126021, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38569370

RESUMO

α-hemolysin (Hla), a toxin secreted by Staphylococcus aureus (S. aureus), has been proved to be involved in the occurrence and aggravation of food poisoning. Hence, it is quite essential to establish its rapid detection methods to guarantee food safety. Sandwich ELISA based on nanobody is well known to be viable for toxins, but there is absence of nanobody against Hla, let alone a pair for it. Therefore, in this paper, we screened specific nanobodies by bio-panning and obtained the optimal nanobody pair for sandwich ELISA firstly. Then, RANbody, a novel nanobody owning both recognition and catalytic capability, is generated in a single step and at low cost through molecular recombination technology. Subsequently, sandwich ELISA was developed to detect Hla based on the nanobody and RANbody, that not only eliminated the use of secondary antibodies and animal-derived antibody, but also reduced detection time and cost, compared with traditional sandwich ELISA. Lastly, the performance has been evaluated, especially for specificity which showed no response to other hemolysins and a low limit of detection of 10 ng/mL. Besides, the proposed sandwich ELISA exhibits favorable feasibility and was successfully employed for the detection of Hla in milk and pork samples.


Assuntos
Ensaio de Imunoadsorção Enzimática , Proteínas Hemolisinas , Leite , Proteínas Hemolisinas/imunologia , Proteínas Hemolisinas/análise , Proteínas Hemolisinas/química , Ensaio de Imunoadsorção Enzimática/métodos , Animais , Leite/química , Anticorpos de Domínio Único/imunologia , Anticorpos de Domínio Único/química , Contaminação de Alimentos/análise , Toxinas Bacterianas/análise , Toxinas Bacterianas/imunologia , Suínos , Staphylococcus aureus/isolamento & purificação , Staphylococcus aureus/imunologia , Limite de Detecção , Análise de Alimentos/métodos
2.
Infect Immun ; 92(5): e0044023, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38591882

RESUMO

Extraintestinal pathogenic Escherichia coli (ExPEC) is a leading cause of worldwide morbidity and mortality, the top cause of antimicrobial-resistant (AMR) infections, and the most frequent cause of life-threatening sepsis and urinary tract infections (UTI) in adults. The development of an effective and universal vaccine is complicated by this pathogen's pan-genome, its ability to mix and match virulence factors and AMR genes via horizontal gene transfer, an inability to decipher commensal from pathogens, and its intimate association and co-evolution with mammals. Using a pan virulome analysis of >20,000 sequenced E. coli strains, we identified the secreted cytolysin α-hemolysin (HlyA) as a high priority target for vaccine exploration studies. We demonstrate that a catalytically inactive pure form of HlyA, expressed in an autologous host using its own secretion system, is highly immunogenic in a murine host, protects against several forms of ExPEC infection (including lethal bacteremia), and significantly lowers bacterial burdens in multiple organ systems. Interestingly, the combination of a previously reported autotransporter (SinH) with HlyA was notably effective, inducing near complete protection against lethal challenge, including commonly used infection strains ST73 (CFT073) and ST95 (UTI89), as well as a mixture of 10 of the most highly virulent sequence types and strains from our clinical collection. Both HlyA and HlyA-SinH combinations also afforded some protection against UTI89 colonization in a murine UTI model. These findings suggest recombinant, inactive hemolysin and/or its combination with SinH warrant investigation in the development of an E. coli vaccine against invasive disease.


Assuntos
Infecções por Escherichia coli , Proteínas de Escherichia coli , Vacinas contra Escherichia coli , Escherichia coli Extraintestinal Patogênica , Proteínas Hemolisinas , Animais , Escherichia coli Extraintestinal Patogênica/genética , Escherichia coli Extraintestinal Patogênica/imunologia , Infecções por Escherichia coli/prevenção & controle , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/imunologia , Camundongos , Proteínas Hemolisinas/imunologia , Proteínas Hemolisinas/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/imunologia , Vacinas contra Escherichia coli/imunologia , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/genética , Feminino , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Sistemas de Secreção Tipo V/imunologia , Sistemas de Secreção Tipo V/genética , Modelos Animais de Doenças , Humanos
3.
Science ; 376(6599): eabm6380, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35587511

RESUMO

The molecular basis of interindividual clinical variability upon infection with Staphylococcus aureus is unclear. We describe patients with haploinsufficiency for the linear deubiquitinase OTULIN, encoded by a gene on chromosome 5p. Patients suffer from episodes of life-threatening necrosis, typically triggered by S. aureus infection. The disorder is phenocopied in patients with the 5p- (Cri-du-Chat) chromosomal deletion syndrome. OTULIN haploinsufficiency causes an accumulation of linear ubiquitin in dermal fibroblasts, but tumor necrosis factor receptor-mediated nuclear factor κB signaling remains intact. Blood leukocyte subsets are unaffected. The OTULIN-dependent accumulation of caveolin-1 in dermal fibroblasts, but not leukocytes, facilitates the cytotoxic damage inflicted by the staphylococcal virulence factor α-toxin. Naturally elicited antibodies against α-toxin contribute to incomplete clinical penetrance. Human OTULIN haploinsufficiency underlies life-threatening staphylococcal disease by disrupting cell-intrinsic immunity to α-toxin in nonleukocytic cells.


Assuntos
Toxinas Bacterianas , Síndrome de Cri-du-Chat , Endopeptidases , Haploinsuficiência , Proteínas Hemolisinas , Infecções Estafilocócicas , Staphylococcus aureus , Toxinas Bacterianas/imunologia , Síndrome de Cri-du-Chat/genética , Síndrome de Cri-du-Chat/imunologia , Endopeptidases/genética , Haploinsuficiência/genética , Haploinsuficiência/imunologia , Proteínas Hemolisinas/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Celular/genética , Necrose , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/patologia
4.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-35055067

RESUMO

α-hemolysin (HlyA) of E. coli binds irreversibly to human erythrocytes and induces cell swelling, ultimately leading to hemolysis. We characterized the mechanism involved in water transport induced by HlyA and analyzed how swelling and hemolysis might be coupled. Osmotic water permeability (Pf) was assessed by stopped-flow light scattering. Preincubation with HlyA strongly reduced Pf in control- and aquaporin 1-null red blood cells, although the relative Pf decrease was similar in both cell types. The dynamics of cell volume and hemolysis on RBCs was assessed by electrical impedance, light dispersion and hemoglobin release. Results show that HlyA induced erythrocyte swelling, which is enhanced by purinergic signaling, and is coupled to osmotic hemolysis. We propose a mathematical model of HlyA activity where the kinetics of cell volume and hemolysis in human erythrocytes depend on the flux of osmolytes across the membrane, and on the maximum volume that these cells can tolerate. Our results provide new insights for understanding signaling and cytotoxicity mediated by HlyA in erythrocytes.


Assuntos
Tamanho Celular , Eritrócitos/citologia , Eritrócitos/fisiologia , Proteínas de Escherichia coli/farmacologia , Proteínas Hemolisinas/farmacologia , Modelos Biológicos , Trifosfato de Adenosina/metabolismo , Biomarcadores , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Relação Dose-Resposta a Droga , Proteínas de Escherichia coli/imunologia , Proteínas Hemolisinas/imunologia , Hemólise , Interações Hospedeiro-Patógeno , Humanos , Cinética , Permeabilidade
5.
J Infect Dis ; 225(1): 177-185, 2022 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-34145461

RESUMO

BACKGROUND: Staphylococcus aureus infections are common throughout the lifespan, with recurrent infections occurring in nearly half of infected children. There is no licensed vaccine, underscoring the need to better understand how S. aureus evades protective immunity. Despite much study, the relative contributions of antibodies and T cells to protection against S. aureus infections in humans are not fully understood. METHODS: We prospectively quantified S. aureus-specific antibody levels by ELISA and T-cell responses by ELISpot in S. aureus-infected and healthy children. RESULTS: S. aureus-specific antibody levels and T-cell responses increased with age in healthy children, suggesting a coordinated development of anti-staphylococcal immunity. Antibody levels against leukotoxin E (LukE) and Panton-Valentine leukocidin (LukS-PV), but not α-hemolysin (Hla), were higher in younger infected children, compared with healthy children; these differences disappeared in older children. We observed a striking impairment of global and S. aureus-specific T-cell function in children with invasive and noninvasive infection, suggesting that S. aureus-specific immune responses are dysregulated during childhood infection regardless of the infection phenotype. CONCLUSIONS: These findings identify a potential mechanism by which S. aureus infection actively evades adaptive immune responses, thereby preventing the development of protective immunity and maintaining susceptibility to recurrent infection.


Assuntos
Anticorpos Antibacterianos/sangue , Exotoxinas/imunologia , Leucocidinas/imunologia , Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas/epidemiologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus , Adolescente , Toxinas Bacterianas , Criança , Pré-Escolar , Ensaio de Imunoadsorção Enzimática , Feminino , Proteínas Hemolisinas/imunologia , Humanos , Lactente , Masculino , Estudos Prospectivos , Estudos Soroepidemiológicos , Linfócitos T , Adulto Jovem
6.
Infect Immun ; 89(10): e0034221, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34227839

RESUMO

In the Gram-positive pathogen Staphylococcus aureus, pore-forming toxins (PFTs), such as leukocidins and hemolysins, play prominent roles in staphylococcal pathogenesis by killing host immune cells and red blood cells (RBCs). However, it remains unknown which combination of toxin antigens would induce the broadest protective immune response against those toxins. In this study, by targeting six major staphylococcal PFTs (i.e., gamma-hemolysin AB [HlgAB], gamma-hemolysin CB [HlgCB], leukocidin AB [LukAB], leukocidin ED [LukED], Panton-Valentine leukocidin [LukSF-PV], and alpha-hemolysin [Hla]), we generated 10 recombinant toxins or toxin subunits, 3 toxoids, and their rabbit antibodies. Using the cytolytic assay for RBCs and polymorphonuclear cells (PMNs), we determined the best combination of toxin antibodies conferring the broadest protection against those staphylococcal PFTs. Although anti-HlgA IgG (HlgA-IgG) showed low cross-reactivity to other toxin components, it was essential to protect rabbit and human RBCs and human PMNs. For the protection of rabbit RBCs, HlaH35L toxoid-IgG was also required, whereas for human PMNs, LukS-IgG and LukAE323AB-IgG were essential too. When the toxin/toxoid antigens HlgA, LukS-PV, HlaH35L, and LukAE323AB were used to immunize rabbits, they increased rabbit survival; however, they did not block staphylococcal abscess formation in kidneys. Based on these results, we proposed that the combination of HlgA, LukS, HlaH35L, and LukAE323AB is the optimal vaccine component to protect human RBCs and PMNs from staphylococcal PFTs. We also concluded that a successful S. aureus vaccine requires not only those toxin antigens but also other antigens that can induce immune responses blocking staphylococcal colonization.


Assuntos
Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Vacinas Combinadas/imunologia , Animais , Proteínas de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Reações Cruzadas/imunologia , Eritrócitos/imunologia , Eritrócitos/microbiologia , Exotoxinas/imunologia , Proteínas Hemolisinas/imunologia , Humanos , Imunização/métodos , Leucocidinas/imunologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Coelhos , Infecções Estafilocócicas/microbiologia , Toxoides/imunologia
7.
PLoS Pathog ; 17(7): e1009752, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34288976

RESUMO

Highly immunogenic exotoxins are used as carrier proteins because they efficiently improve the immunogenicity of polysaccharides. However, their efficiency with protein antigens remains unclear. In the current study, the candidate antigen PA0833 from Pseudomonas aeruginosa was fused to the α-hemolysin mutant HlaH35A from Staphylococcus aureus to form a HlaH35A-PA0833 fusion protein (HPF). Immunization with HPF resulted in increased PA0833-specific antibody titers, higher protective efficacy, and decreased bacterial burden and pro-inflammatory cytokine secretion compared with PA0833 immunization alone. Using fluorescently labeled antigens to track antigen uptake and delivery, we found that HlaH35A fusion significantly improved antigen uptake in injected muscles and antigen delivery to draining lymph nodes. Both in vivo and in vitro studies demonstrated that the increased antigen uptake after immunization with HPF was mainly due to monocyte- and macrophage-dependent macropinocytosis, which was probably the result of HPF binding to ADAM10, the Hla host receptor. Furthermore, a transcriptome analysis showed that several immune signaling pathways were activated by HPF, shedding light on the mechanism whereby HlaH35A fusion improves immunogenicity. Finally, the improvement in immunogenicity by HlaH35A fusion was also confirmed with two other antigens, GlnH from Klebsiella pneumoniae and the model antigen OVA, indicating that HlaH35A could serve as a universal carrier protein to improve the immunogenicity of protein antigens.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas Hemolisinas/imunologia , Vacinas/imunologia , Células A549 , Animais , Exotoxinas/imunologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células RAW 264.7 , Proteínas Recombinantes de Fusão/imunologia
8.
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
9.
Fish Shellfish Immunol ; 115: 189-197, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34147613

RESUMO

The present study examines the effectiveness of DNA vaccine against Aeromonas hydrophila through oral route using chitosan-tripolyphosphate (Cs-TPP) nanoparticles encapsulation. The virulent gene of outer membrane protein (OMP) and hemolysin (hly) related to pathogenicity of A. hydrophila was used to construct a DNA vaccine using pVAX1, and the construct was named as pVAX-OMP and pVAX-hly DNA vaccines. The pVAX-OMP and pVAX-hly DNA vaccines were encapsulated by Cs-TPP nanoparticles and size measured by field emission scanning electron microscopy (FE-SEM). The encapsulation efficiency of Cs-TPP nanoparticles was found to be 79.6% for pVAX-OMP DNA and 82.3% for pVAX-hly DNA binding with Cs-TPP nanoparticles. The stability and invitro release profile of plasmid DNA was also determined after encapsulation using DNase and chitosanase. DNA vaccines distribution in tissues was investigated in fish fed with the pVAX-OMP, pVAX-hly and pVAX-OMP+pVAX-hly encapsulated in Cs-TPP nanoparticles and confirmed by PCR and multiplex PCR. The results suggest that Cs-TPP nanoparticles encapsulated DNA vaccine delivered into fish by feeding. After oral vaccination of Labeo rohita were challenged with A. hydrophila by intraperitoneal injection. Relatively, gene expression of c- and g-type lysozyme followed by pro- and anti-inflammatory cytokines (Interlukin-10 and Tumor Growth Factor ß) was up-regulated in heart and kidney for pVAX-OMP+pVAX-hly vaccinated group. Moreover, fish fed with pVAX-OMP+pVAX-hly encapsulated in Cs-TPP nanoparticles had a significantly higher survival rate (76.2%) against A. hydrophila. This study concludes that pVAX-OMP and pVAX-hly DNA vaccines can be delivered orally using Cs-TPP nanoparticles for protection against A. hydrophilainfection.


Assuntos
Vacinas Bacterianas/administração & dosagem , Quitosana/análogos & derivados , Cyprinidae , Doenças dos Peixes/prevenção & controle , Infecções por Bactérias Gram-Negativas/veterinária , Vacinação/veterinária , Vacinas de DNA/administração & dosagem , Administração Oral , Aeromonas hydrophila/fisiologia , Animais , Proteínas da Membrana Bacteriana Externa/imunologia , Quitosana/administração & dosagem , Quitosana/imunologia , Doenças dos Peixes/microbiologia , Infecções por Bactérias Gram-Negativas/microbiologia , Infecções por Bactérias Gram-Negativas/prevenção & controle , Proteínas Hemolisinas/imunologia , Nanopartículas/administração & dosagem
10.
Anal Biochem ; 625: 114222, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-33932355

RESUMO

The anti-idiotypic antibody is widely used in the field of immunology to simulate structural features or even induce the biological activity of antigens. In this study, we obtained seven anti-idiotypic single-chain variable fragments (scFv) antibodies of Cry2Aa toxin from a phage-displayed mutant library constructed using error-prone PCR technique. A mutant designated 2-12B showed the best binding ability amongst all anti-idiotypic scFv isolates to Plutella xylostella brush border membrane vesicles (BBMVs). 2-12B and Cry2Aa toxin shared a potential receptor of polycalin in P. xylostella BBMVs. Homology modeling and molecular docking demonstrated that 2-12B and Cry2Aa toxin have seven common binding amino acid residues in polycalin. Insect bioassay results suggested that 2-12 had insecticidal efficacy against P. xylostella larvae. These results indicated that the Cry2Aa anti-idiotypic scFv antibody 2-12B partially mimicked the structure and function of Cry2Aa toxin. The anti-idiotypic scFv antibody provides the basic material for the future study of surrogate molecules or new insecticidal materials.


Assuntos
Anticorpos Anti-Idiotípicos/química , Anticorpos Monoclonais/química , Toxinas de Bacillus thuringiensis/química , Endotoxinas/química , Proteínas Hemolisinas/química , Região Variável de Imunoglobulina/química , Anticorpos de Cadeia Única/química , Animais , Anticorpos Anti-Idiotípicos/genética , Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Anti-Idiotípicos/metabolismo , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Toxinas de Bacillus thuringiensis/imunologia , Toxinas de Bacillus thuringiensis/metabolismo , Membrana Celular/metabolismo , Endotoxinas/imunologia , Endotoxinas/metabolismo , Proteínas Hemolisinas/imunologia , Proteínas Hemolisinas/metabolismo , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Região Variável de Imunoglobulina/metabolismo , Simulação de Acoplamento Molecular , Mariposas , Mutação , Biblioteca de Peptídeos , Conformação Proteica , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/metabolismo
11.
Int J Biol Macromol ; 183: 1346-1351, 2021 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-34004200

RESUMO

Anti-idiotypic antibody technique is a new approach for the rapid development of insecticidal protein. In this study, anti-Cry1A polyclonal antibodies were used as antigen to screen the anti-idiotypic antibody that can simulate Cry1A toxins from a phage display human domain antibody (DAB) library. After four rounds of panning, five positive clones that have binding activities with anti-Cry1A polyclonal antibodies were obtained. Indirect competitive ELISA (IC-ELISA) results showed that the positive clone D6 showed significant inhibition for the binding of Cry1A toxins with anti-Cry1A polyclonal antibodies, and the inhibition ratio increased with the increase of D6 content. While, B3, F4, G5, C7 and the controls showed no obvious inhibition to Cry1A toxins. The results suggest that D6 is the "ß" subtype anti-idiotypic antibody, which can simulate Cry1A toxins and competitive binding with anti-Cry1A polyclonal antibodies. Meanwhile, D6 had certain binding activity with the brush border membrane vesicles (BBMV) of p. xylostella, which was the receptor of Cry1A toxins. The results of bioassay showed that D6 had certain insecticidal activity, and the lethal concentration of 50% (LC50) was 976 ng/cm2. This study provides basic materials and experience for the development of Cry toxin simulants.


Assuntos
Toxinas de Bacillus thuringiensis/imunologia , Endotoxinas/imunologia , Proteínas Hemolisinas/imunologia , Biblioteca de Peptídeos , Proteínas de Bactérias/imunologia , Ensaio de Imunoadsorção Enzimática , Humanos
12.
Mol Immunol ; 135: 45-52, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33873093

RESUMO

Staphylococcus aureus (SA), especially the methicillin-resistant variant (MRSA), is becoming a serious threat to human health in hospitals and communities, making the development of an effective vaccine urgent. Alpha-hemolysin (Hla) is a key virulence factor and also a good target for the development of SA vaccines. However, the epitopes in Hla recognized by human immunity are not characterized in detail, which hinders the design of epitope-based human vaccines against SA. In this study, we collected sera from volunteers in a phase 1b clinical trial of a novel recombinant five-antigen SA vaccine (NCT03966040). Using a Luminex-based assay, we characterized the human serologic response against Hla, and identified Hla121-138 as a neutralizing epitope. In addition, we successfully produced ferritin nanoparticles carrying the neutralizing Hla121-138 epitope (EpNP) in E. coli. EpNP presented as homogenous nanoparticles in aqueous solution. Immunization with EpNP elicited potent hemolysis-neutralizing antibodies and conferred significant protection in a mouse model of SA skin infection. Our data suggest that EpNP, carrying the neutralizing epitope Hla121-138, is a good candidate for a vaccine against SA.


Assuntos
Anticorpos Antibacterianos/imunologia , Anticorpos Neutralizantes/imunologia , Toxinas Bacterianas/imunologia , Proteínas Hemolisinas/imunologia , Staphylococcus aureus Resistente à Meticilina/imunologia , Infecções Estafilocócicas/prevenção & controle , Animais , Toxinas Bacterianas/antagonistas & inibidores , Vacinas Bacterianas/imunologia , Epitopos/imunologia , Feminino , Proteínas Hemolisinas/antagonistas & inibidores , Humanos , Nanopartículas Metálicas/química , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Camundongos , Camundongos Endogâmicos BALB C , Infecções Estafilocócicas/microbiologia , Vacinação , Fatores de Virulência
13.
Dev Comp Immunol ; 121: 104071, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33766585

RESUMO

The insecticidal Bacillus thuringiensis protein Cry1Ac is produced as a protoxin and becomes activated to a toxin when ingested by larvae. Both proteins are immunogenic and able to activate macrophages. The proposed mechanism of immunostimulation by Cry1Ac protoxin has been related to its capacity to activate antigen-presenting cells (APC), but its ability to activate dendritic cells (DC) has not been explored. Here we evaluated, in the popliteal lymph nodes (PLN), spleen and peritoneum, the activation of DC CD11c+ MHC-II+ following injection with single doses (50 µg) of Cry1Ac toxin or protoxin via the intradermal (i.d.) and intraperitoneal (i.p.) routes in C57BL/6 mice. In vivo stimulation with both Cry1Ac proteins induced activation of DC via upregulation of CD86, primarily in PLN 24 h after i. d. injection. Moreover, this activation was detected in DC, displaying CD103+, a typical marker of migratory DC, while upregulation of CD80 was uniquely induced by toxin. Tracking experiments showed that Cy5-labeled Cry1Ac proteins could rapidly reach the PLN and localize near DC, but some label remained in the footpad. When the capacity of Cry1Ac-activated DC to induce antigen presentation was examined, significant proliferation of naïve T lymphocytes was induced exclusively by the protoxin. The protoxin elicited a Th17-biased cytokine profile. Moreover, only the Cry1Ac toxin induced a pronounced proliferation of B cells from both untreated and Cry1Ac-injected mice, suggesting that it acts as a polyclonal activator. In conclusion, Cry1Ac protoxin and toxin show a distinctive capacity to activate APCs.


Assuntos
Linfócitos B/imunologia , Toxinas de Bacillus thuringiensis/imunologia , Bacillus thuringiensis/imunologia , Células Dendríticas/imunologia , Endotoxinas/imunologia , Proteínas Hemolisinas/imunologia , Animais , Apresentação de Antígeno , Linfócitos B/metabolismo , Toxinas de Bacillus thuringiensis/administração & dosagem , Células Dendríticas/metabolismo , Endotoxinas/administração & dosagem , Feminino , Proteínas Hemolisinas/administração & dosagem , Ativação Linfocitária , Camundongos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia
14.
J Invest Dermatol ; 141(4): 810-820.e8, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32946878

RESUMO

The staphylococcal α-hemolysin is critical for the pathogenesis of Staphylococcus aureus skin and soft tissue infection. Vaccine and infection-elicited α-hemolysin-specific antibodies protect against S. aureus‒induced dermonecrosis, a key feature of skin and soft tissue infection. Many interactions between α-hemolysin and host cells have been identified that promote tissue damage and modulate immune responses, but the mechanisms by which protective adaptive responses cross talk with innate responses at the tissue level are not clear. Using an established mouse model of skin and soft tissue infection and a newly developed histopathologic scoring system, we observed pathologic correlates early after infection, predicting protection against dermonecrosis by anti-α-hemolysin antibody. Protection was characterized by robust neutrophilic inflammation and compartmentalization of bacteria into discrete abscesses, which led to the attenuation of dermonecrosis and enhancement of bacterial clearance later in the infection. The ultimate outcome of infection was driven by the recruitment of neutrophils within the first day after infection but not later. Antibody-mediated protection was dependent on toxin neutralization rather than on enhanced opsonophagocytic killing by neutrophils or protection against toxin-mediated neutrophil lysis. Together, these findings advance our understanding of the mechanisms by which the early synergism between antibody-mediated toxin neutralization and tissue-specific neutrophilic inflammation preserve tissue integrity during infection.


Assuntos
Anticorpos Antibacterianos/metabolismo , Anticorpos Neutralizantes/metabolismo , Toxinas Bacterianas/imunologia , Proteínas Hemolisinas/imunologia , Neutrófilos/imunologia , Pele/patologia , Infecções Cutâneas Estafilocócicas/imunologia , Animais , Anticorpos Antibacterianos/administração & dosagem , Anticorpos Antibacterianos/imunologia , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/imunologia , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Feminino , Voluntários Saudáveis , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Humanos , Imunização Passiva/métodos , Camundongos , Necrose/imunologia , Necrose/microbiologia , Necrose/patologia , Infiltração de Neutrófilos , Cultura Primária de Células , Pele/imunologia , Pele/microbiologia , Infecções Cutâneas Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/patologia , Staphylococcus aureus/imunologia
16.
Front Immunol ; 11: 599439, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33193453

RESUMO

The Vibrio vulnificus (V. vulnificus) hemolysin (VVH) is a pore-forming cholesterol-dependent cytolysin (CDC). Although there has been some debate surrounding the in vivo virulence effects of the VVH, it is becoming increasingly clear that it drives different cellular outcomes and is involved in the pathogenesis of V. vulnificus. This minireview outlines recent advances in our understanding of the regulation of vvhA gene expression, the biological activity of the VVH and its role in pathogenesis. An in-depth examination of the role of the VVH in V. vulnificus pathogenesis will help reveal the potential targets for therapeutic and preventive interventions to treat fatal V. vulnificus septicemia in humans. Future directions in VVH research will also be discussed.


Assuntos
Regulação Bacteriana da Expressão Gênica , Proteínas Hemolisinas/imunologia , Vibrioses/imunologia , Vibrio vulnificus , Animais , Proteínas de Bactérias/imunologia , Humanos , Vibrio vulnificus/imunologia , Vibrio vulnificus/patogenicidade
17.
Sheng Wu Gong Cheng Xue Bao ; 36(7): 1378-1385, 2020 Jul 25.
Artigo em Chinês | MEDLINE | ID: mdl-32748595

RESUMO

Listeria monocytogenes (Lm) is zoonotic pathogen that can cause listeriosis, and vaccine is one of the effective methods to prevent this pathogen infection. In this study, we developed a novel vaccine that is a mixture of inactivated bacteria and Montanide™ ISA 61 VG, a mineral oil adjuvant, and evaluated the safety and immune response characteristics of this vaccine. The mice immunized with the ISA 61 VG adjuvant had high safety, and it could induce significantly higher titer of anti-listeriolysin O (LLO) antibody and higher value of IgG2a/IgG1 ratio compared with the group without the adjuvant. In particular, it could provide 100% immune protection against lethal doses of Lm challenge in mice. In summary, ISA 61VG adjuvant significantly enhanced the ability of inactivated listeria vaccine to induce humoral and cellular immune responses, thereby enhanced the protective immune response in the host, and it is a potential vaccine candidate for the prevention of Lm infection in humans and animals.


Assuntos
Adjuvantes Imunológicos , Proteínas Hemolisinas , Imunidade Celular , Listeria monocytogenes , Listeriose , Adjuvantes Imunológicos/farmacologia , Animais , Proteínas Hemolisinas/imunologia , Proteínas Hemolisinas/farmacologia , Imunidade Celular/efeitos dos fármacos , Listeria monocytogenes/imunologia , Listeriose/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Vacinas de Produtos Inativados/imunologia
18.
Front Immunol ; 11: 1579, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32849537

RESUMO

Staphylococcus aureus can cause life-threatening diseases, and hospital- as well as community-associated antibiotic-resistant strains are an emerging global public health problem. Therefore, prophylactic vaccines or immune-based therapies are considered as alternative treatment opportunities. To develop such novel treatment approaches, a better understanding of the bacterial virulence and immune evasion mechanisms and their potential effects on immune-based therapies is essential. One important staphylococcal virulence factor is alpha-toxin, which is able to disrupt the epithelial barrier in order to establish infection. In addition, alpha-toxin has been reported to modulate other cell types including immune cells. Since CD4+ T cell-mediated immunity is required for protection against S. aureus infection, we were interested in the ability of alpha-toxin to directly modulate CD4+ T cells. To address this, murine naïve CD4+ T cells were differentiated in vitro into effector T cell subsets in the presence of alpha-toxin. Interestingly, alpha-toxin induced death of Th1-polarized cells, while cells polarized under Th17 conditions showed a high resistance toward increasing concentrations of this toxin. These effects could neither be explained by differential expression of the cellular alpha-toxin receptor ADAM10 nor by differential activation of caspases, but might result from an increased susceptibility of Th1 cells toward Ca2+-mediated activation-induced cell death. In accordance with the in vitro findings, an alpha-toxin-dependent decrease of Th1 and concomitant increase of Th17 cells was observed in vivo during S. aureus bacteremia. Interestingly, corresponding subsets of innate lymphoid cells and γδ T cells were similarly affected, suggesting a more general effect of alpha-toxin on the modulation of type 1 and type 3 immune responses. In conclusion, we have identified a novel alpha-toxin-dependent immunomodulatory strategy of S. aureus, which can directly act on CD4+ T cells and might be exploited for the development of novel immune-based therapeutic approaches to treat infections with antibiotic-resistant S. aureus strains.


Assuntos
Toxinas Bacterianas/imunologia , Proteínas Hemolisinas/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Celular , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia , Caspases/metabolismo , Morte Celular , Citocinas/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th17/imunologia , Células Th17/metabolismo
19.
Microbiol Immunol ; 64(10): 666-678, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32786043

RESUMO

Antigen-specific CD8+ T-lymphocytes (cytotoxic T-lymphocytes: CTL), as well as CD4+ T-lymphocytes (helper T-lymphocytes: Th), simultaneously play an important role in the elimination of intracellular bacteria such as Mycobacterium tuberculosis and Listeria monocytogenes. Administration of T-cell epitope short peptide needs large numbers of peptides for effective vaccination due to its easily degradable nature in vivo. In this respect, biocompatible and biodegradable microparticles combined with CTL/Th-hybrid epitope long peptide (long peptide) have been used to diminish the degradation of loaded peptide. The aim of this study is to develop a novel T cell-oriented vaccine against intracellular bacteria that is composed of long peptide and poly (lactic-co-glycolic acid) (PLGA) microparticles. Mouse bone marrow-derived dendritic cells (BMDCs) were loaded with L. monocytogenes listeriolysin O (LLO)-derived or ovalbumin (OVA)-derived long peptide/PLGA or other comparative antigens. The antigen-loaded BMDCs were injected subcutaneously into the flank of mice twice, and then, the spleens were collected and lymphocyte proliferation and interferon-γ production were evaluated. The median diameter of the PLGA spheres was 1.38 µm. Both LLO- and OVA-long peptide/PLGA showed significantly more robust CTL and Th proliferations with higher interferon-γ production than the long peptide alone or CTL and Th short peptides/PLGA vaccination. Furthermore, the LLO-long peptide/PLGA vaccination showed a significantly lower bacterial burden in spleens compared with the long peptide alone or the CTL and Th short peptides/PLGA vaccination after the challenge of lethal amounts of L. monocytogenes. These results suggest that the novel vaccine taking advantages of CTL/Th-hybrid epitope long peptide and PLGA microparticle is effective for protection against intracellular bacteria.


Assuntos
Vacinas Bacterianas/imunologia , Células Dendríticas/transplante , Epitopos de Linfócito T/imunologia , Listeria monocytogenes/imunologia , Listeriose/prevenção & controle , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/imunologia , Animais , Toxinas Bacterianas/imunologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Células Cultivadas , Células Dendríticas/imunologia , Feminino , Proteínas de Choque Térmico/imunologia , Proteínas Hemolisinas/imunologia , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Linfócitos T Citotóxicos/imunologia , Vacinação
20.
Toxins (Basel) ; 12(7)2020 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-32664481

RESUMO

The pore-forming cytotoxin α-hemolysin, or Hla, is a critical Staphylococcus aureus virulence factor that promotes infection by causing tissue damage, excessive inflammation, and lysis of both innate and adaptive immune cells, among other cellular targets. In this study, we asked whether a virus-like particle (VLP)-based vaccine targeting Hla could attenuate S. aureus Hla-mediated pathogenesis. VLPs are versatile vaccine platforms that can be used to display target antigens in a multivalent array, typically resulting in the induction of high titer, long-lasting antibody responses. In the present study, we describe the first VLP-based vaccines that target Hla. Vaccination with either of two VLPs displaying a 21 amino-acid linear neutralizing domain (LND) of Hla protected both male and female mice from subcutaneous Hla challenge, evident by reduction in lesion size and neutrophil influx to the site of intoxication. Antibodies elicited by VLP-LND vaccination bound both the LND peptide and the native toxin, effectively neutralizing Hla and preventing toxin-mediated lysis of target cells. We anticipate these novel and promising vaccines being part of a multi-component S. aureus vaccine to reduce severity of S. aureus infection.


Assuntos
Toxinas Bacterianas/farmacologia , Vacinas Bacterianas/farmacologia , Proteínas Hemolisinas/farmacologia , Pele/efeitos dos fármacos , Infecções Cutâneas Estafilocócicas/prevenção & controle , Staphylococcus aureus/efeitos dos fármacos , Vacinas de Partículas Semelhantes a Vírus/farmacologia , Animais , Anticorpos Antibacterianos/sangue , Toxinas Bacterianas/imunologia , Vacinas Bacterianas/imunologia , Modelos Animais de Doenças , Epitopos , Feminino , Proteínas Hemolisinas/imunologia , Humanos , Imunogenicidade da Vacina , Células Jurkat , Masculino , Camundongos Endogâmicos BALB C , Testes de Neutralização , Pele/imunologia , Pele/microbiologia , Pele/patologia , Infecções Cutâneas Estafilocócicas/imunologia , Infecções Cutâneas Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/patologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade , Vacinação , Vacinas de Partículas Semelhantes a Vírus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA