Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Immunol ; 22(1): 20, 2021 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-33743606

RESUMEN

BACKGROUND: Bacillus ancthracis causes cutaneous, pulmonary, or gastrointestinal forms of anthrax. B. anthracis is a pathogenic bacterium that is potentially to be used in bioterrorism because it can be produced in the form of spores. Currently, protective antigen (PA)-based vaccines are being used for the prevention of anthrax, but it is necessary to develop more safe and effective vaccines due to their prolonged immunization schedules and adverse reactions. METHODS: We selected the lipoprotein GBAA0190, a potent inducer of host immune response, present in anthrax spores as a novel potential vaccine candidate. Then, we evaluated its immune-stimulating activity in the bone marrow-derived macrophages (BMDMs) using enzyme-linked immunosorbent assay (ELISA) and Western blot analysis. Protective efficacy of GBAA0190 was evaluated in the guinea pig (GP) model. RESULTS: The recombinant GBAA0190 (r0190) protein induced the expression of various inflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), and macrophage inflammatory protein-1α (MIP-1α) in the BMDMs. These immune responses were mediated through toll-like receptor 1/2 via activation of mitogen-activated protein (MAP) kinase and Nuclear factor-κB (NF-κB) pathways. We demonstrated that not only immunization of r0190 alone, but also combined immunization with r0190 and recombinant PA showed significant protective efficacy against B. anthracis spore challenges in the GP model. CONCLUSIONS: Our results suggest that r0190 may be a potential target for anthrax vaccine.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Carbunco/prevención & control , Bacillus anthracis/inmunología , Lipoproteínas/inmunología , Animales , Vacunas contra el Carbunco/administración & dosificación , Vacunas contra el Carbunco/genética , Citocinas/metabolismo , Cobayas , Inmunización , Lipoproteínas/administración & dosificación , Lipoproteínas/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Transducción de Señal , Esporas Bacterianas/inmunología , Receptores Toll-Like/metabolismo
2.
Microb Pathog ; 158: 105104, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34298126

RESUMEN

Anthrax, by Bacillus anthracis, remains a dreadful fatal hazard worldwide. The currently used anthrax vaccines are plagued by numerous issues that limit their widespread use. As an immunization approach targeting both extracellular antigens and toxins of B. anthracis may achieve better sterile immunity, the present investigation designed a bicistronic secretory anti-anthrax DNA vaccine targeting immune response against toxin and cells. The efficacy of the vaccine was compared with monocistronic DNA vaccines and the currently used anthrax vaccine. For this, mice were immunized with the developed vaccine containing pag (encoding protective antigen to block toxin) and eag genes (encoding EA1 to target cells) of B. anthracis through DNA-prime/Protein-boost (D/P) and DNA prime/DNA-boost (D/D) approaches. There was a >2 and > 5 fold increase in specific antibody level by D/D and D/P approaches respectively, on 42nd days post-immunization (dpi). Serum cytokine profiling showed that both Th1 and Th2 immune responses were elicited, with more Th2 responses in D/P strategy. More importantly, challenge with 100 times LD50 of B. anthracis at 42nd dpi exhibited maximum cumulative survival (83.33 %) by bicistronic D/P approach. Remarkably, immunization with EA1 delayed mortality onset in infection. The study forms the first report on complement-dependent bactericidal activity of antiEA1 antibodies. In short, co-immunization of PA and EA1 through the developed bicistronic DNA vaccine would be an effective immunization approach in anthrax vaccination. Further, D/P strategy could enhance vaccine-induced immunity against B. anthracis. Altogether, the study generates certain critical insights having direct applications in next-generation vaccine development against anthrax.


Asunto(s)
Vacunas contra el Carbunco , Bacillus anthracis , Vacunas de ADN , Animales , Vacunas contra el Carbunco/genética , Anticuerpos Antibacterianos , Antígenos Bacterianos/genética , Bacillus anthracis/genética , ADN , Inmunidad , Ratones , Ratones Endogámicos BALB C , Vacunación , Vacunas de ADN/genética
3.
Immunopharmacol Immunotoxicol ; 43(4): 495-502, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34259590

RESUMEN

CONTEXT: Bacillus anthracis secretes a tripartite toxin comprising protective antigen (PA), edema factor (EF), and lethal factor (LF). The human anthrax vaccine is mainly composed of the anthrax protective antigen (PA). Considerable efforts are being directed towards improving the efficacy of vaccines because the use of commercial anthrax vaccines (human/veterinary) is associated with several limitations. OBJECTIVE: In this study, a triple chimeric antigen referred to as ELP (gene accession no: MT590758) comprising highly immunogenic domains of PA, LF, and EF was designed, constructed, and assessed for the immunization capacity against anthrax in a guinea pig model. MATERIALS AND METHODS: Immunization was carried out considering antigen titration and immunization protocol. The immunoprotective efficacy of the ELP was evaluated in guinea pigs and compared with the potency of veterinary anthrax vaccine using a challenge test with B. anthracis 17JB strain spores. RESULTS: The results demonstrated that the ELP antigen induced strong humoral responses. The T-cell response of the ELP was found to be similar to PA, and showed that the ELP could protect 100%, 100%, 100%, 80% and 60% of the animals from 50, 70, 90, 100 and 120 times the minimum lethal dose (MLD, equal 5 × 105 spore/ml), respectively, which killed control animals within 48 h. DISCUSSION AND CONCLUSIONS: It is concluded that the ELP antigen has the necessary requirement for proper immunization against anthrax and it can be used to develop an effective recombinant vaccine candidate against anthrax.


Asunto(s)
Vacunas contra el Carbunco/administración & dosificación , Antígenos Bacterianos/administración & dosificación , Bacillus anthracis/efectos de los fármacos , Esporas Bacterianas/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Vacunas contra el Carbunco/genética , Vacunas contra el Carbunco/inmunología , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Bacillus anthracis/genética , Bacillus anthracis/inmunología , Femenino , Cobayas , Humanos , Esporas Bacterianas/inmunología , Resultado del Tratamiento
4.
Infect Immun ; 88(8)2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32393506

RESUMEN

Bacillus anthracis is the causative agent of anthrax disease, presents with high mortality, and has been at the center of bioweapon efforts. The only currently U.S. FDA-approved vaccine to prevent anthrax in humans is anthrax vaccine adsorbed (AVA), which is protective in several animal models and induces neutralizing antibodies against protective antigen (PA), the cell-binding component of anthrax toxin. However, AVA requires a five-course regimen to induce immunity, along with an annual booster, and is composed of undefined culture supernatants from a PA-secreting strain. In addition, it appears to be ineffective against strains that lack anthrax toxin. Here, we investigated a vaccine formulation consisting of recombinant proteins from a surface-localized heme transport system containing near-iron transporter (NEAT) domains and its efficacy as a vaccine for anthrax disease. The cocktail of five NEAT domains was protective against a lethal challenge of inhaled bacillus spores at 3 and 28 weeks after vaccination. The reduction of the formulation to three NEATs (IsdX1, IsdX2, and Bslk) was as effective as a five-NEAT domain cocktail. The adjuvant alum, approved for use in humans, was as protective as Freund's Adjuvant, and protective vaccination correlated with increased anti-NEAT antibody reactivity and reduced bacterial levels in organs. Finally, the passive transfer of anti-NEAT antisera reduced mortality and disease severity, suggesting the protective component is comprised of antibodies. Collectively, these results provide evidence that a vaccine based upon recombinant NEAT proteins should be considered in the development of a next-generation anthrax vaccine.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Carbunco/prevención & control , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Neutralizantes/biosíntesis , Antígenos Bacterianos/inmunología , Bacillus anthracis/efectos de los fármacos , Administración por Inhalación , Compuestos de Alumbre/administración & dosificación , Animales , Carbunco/inmunología , Carbunco/microbiología , Carbunco/mortalidad , Vacunas contra el Carbunco/administración & dosificación , Vacunas contra el Carbunco/genética , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/genética , Bacillus anthracis/inmunología , Bacillus anthracis/patogenicidad , Proteínas Bacterianas/administración & dosificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Portadoras/administración & dosificación , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Complemento C5/deficiencia , Femenino , Adyuvante de Freund/administración & dosificación , Humanos , Inmunogenicidad Vacunal , Ratones Noqueados , Análisis de Supervivencia , Vacunación/métodos
5.
Biologicals ; 61: 38-43, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31416791

RESUMEN

Tremendous efforts are being made to develop an anthrax vaccine with long term protection. The main component of traditional anthrax vaccine is protective antigen (PA) with the trace amount of other proteins and bacterial components. In this study, we developed a recombinant PA-LF chimera antigen of Bacillus anthracis by fusing the PA domain 2-4 with lethal factor (LF) domain 1 and evaluated its protective potential against B. anthracis in mouse model. The anti-PA-LF chimera serum reacted with both PA and LF antigen, individually. The chimera elicited a strong antibody titer in mice with predominance of IgG1 isotype followed by IgG2b, IgG2a and IgG3. Cytokines were assessed in splenocytes of immunized mice and a significant up-regulation in the expression of IL-4, IL-10, IFN-γ and TNF-α was observed. The PA-LF chimera immunized mice exhibited 80% survival after challenge with virulent spores of B. anthracis. Pathological studies showed normal architecture in vital organs (spleen, lung, liver and kidney) of recovered immunized mice on 20 DPI after spore challenge. These findings suggested that PA-LF chimera of B. anthracis elicited good humoral as well as cell mediated immune response in mice, and thus, can be a potent vaccine candidate against anthrax.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Carbunco/prevención & control , Antígenos Bacterianos/inmunología , Bacillus anthracis/inmunología , Toxinas Bacterianas/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Carbunco/inmunología , Carbunco/patología , Vacunas contra el Carbunco/genética , Antígenos Bacterianos/genética , Bacillus anthracis/genética , Toxinas Bacterianas/genética , Manejo de la Enfermedad , Evaluación de Medicamentos , Femenino , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión/genética
6.
Immunopharmacol Immunotoxicol ; 41(1): 25-31, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30621469

RESUMEN

Background: Anthrax is a zoonotic disease caused by Bacillus anthracis and it can be deadly in 6 days. Considerable efforts have been conducted toward developing more effective veterinary and human anthrax vaccines because these common vaccines have several limitations. B. anthracis secretes a tripartite toxin, comprising protective antigen (PA), edema factor (EF), and lethal factor (LF). Several studies have shown important role of PA in protection of anthrax. LF and EF induce production of toxin neutralizing antibodies too. PA in fusion form with LF/EF has synergistic effects as a potential subunit vaccine. Methods: In this study, for the first time, a triple chimeric protein called ELP was modeled by fusing three different domains of anthrax toxic antigens, the N-terminal domains of EF and LF, and the C-terminal domain of PA as a high immunogenic antigen using Modeller 9.19 software. Immunogenicity of the ELP was assessed in guinea pigs using enzyme-linked immunosorbent assay (ELISA) test and MTT assay. Results: Theoretical studies and molecular dynamics (MD) simulation results suggest that the ELP model had acceptable quality and stability. Sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) analysis of the purified ELP, its domains, and PA were matched with their molecular size and confirmed by western blotting analysis. In the immune guinea pigs, antibody was produced against all of the ELP domains. It was observed that ELP induced strong humoral response and could protect murine macrophage cell line (RAW 264.7 cells) against anthrax lethal toxin (LeTx). Conclusions: ELP chimeric antigen could be considered as a high immunogenic antigen.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Carbunco/prevención & control , Anticuerpos Neutralizantes/sangre , Antígenos Bacterianos/inmunología , Bacillus anthracis/inmunología , Toxinas Bacterianas/inmunología , Modelos Teóricos , Animales , Carbunco/inmunología , Vacunas contra el Carbunco/genética , Vacunas contra el Carbunco/toxicidad , Antígenos Bacterianos/genética , Antígenos Bacterianos/toxicidad , Bacillus anthracis/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/toxicidad , Cobayas , Ratones , Simulación de Dinámica Molecular , Pruebas de Neutralización , Células RAW 264.7 , Programas Informáticos , Vacunas Sintéticas
7.
Appl Microbiol Biotechnol ; 100(19): 8439-51, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27364624

RESUMEN

Bacillus anthracis chimeric molecule PALFn, comprising the immunodominant domains of protective antigen (PA) and lethal factor (LF), has been developed in the past and has been shown to confer enhanced protection against anthrax in mouse model when challenged with anthrax lethal toxin (LeTx). However, the immunological correlates for this chimeric antigen, both in terms of humoral as well as cell-mediated immune responses, have not been described in detail. To address this gap, we have determined the immunological responses both at humoral as well as cellular levels for the protection conferred by the novel chimeric antigen PALFn constructed in our laboratory in comparison to PA antigen. The biological functionality of the chimeric antigen was ascertained by the trypsin digestion assay. The trypsin cleavage activated the functionality of PALFn and rendered it to interact and bind with the LF molecule. Similarly, the LFn component in the chimera could independently interact and bind to the trypsin-activated wild-type PA. Further, it was observed that the PALFn-immunized mice sera could readily react to both PA and LF antigens while PA-immunized mice sera showed reaction to PA and PALFn alone and not to the individual LF antigen. The in vitro toxin neutralizing ability of PALFn antisera on macrophage cell line J774.1 was robust but with 1.3-fold lesser titer than PA-immunized antisera. PALFn-immunized mouse splenocytes showed a significant lymphocyte proliferation when stimulated with PALFn. There was a remarkable increase in the level of interleukin-2 (IL-2), interleukin-4 (IL-4), interleukin 10 (IL-10), interferon-γ (IFN- γ), and tumor necrosis factor α (TNFα) from PALFn- and PA-stimulated splenocytes. In addition, there was a significant increase in antigen-specific CD4+ and CD8+ T-cell counts from both PALFn- and PA-immunized mouse splenocytes. The results clearly demonstrate the ability of chimeric molecule PALFn in eliciting robust humoral and cell-mediated immune responses in mouse model that is parallel to the wild-type PA but has additional anti-LF antibody response. Considering the enhanced protection offered by the chimera PALFn, we can conclude that it can be a better alternative to the wild-type PA-based recombinant vaccine against anthrax.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Antígenos Bacterianos/inmunología , Bacillus anthracis/inmunología , Toxinas Bacterianas/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Carbunco/prevención & control , Vacunas contra el Carbunco/administración & dosificación , Vacunas contra el Carbunco/genética , Anticuerpos Antibacterianos/sangre , Anticuerpos Neutralizantes/sangre , Antígenos Bacterianos/genética , Antitoxinas/sangre , Toxinas Bacterianas/genética , Línea Celular , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Linfocitos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Ratones , Pruebas de Neutralización , Proteínas Recombinantes de Fusión/genética , Bazo/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
8.
Artículo en Ruso | MEDLINE | ID: mdl-27029122

RESUMEN

Live genetic engineering anthrax vaccines on the platform of avirulent and probiotic micro-organisms are a safe and adequate alternative to preparations based on attenuated Bacillus anthracis strains. Mucosal application results in a direct contact of the vaccine preparations with mucous membranes in those organs arid tissues of the macro-organisms, that are exposed to the pathogen in the first place, resulting in a development of local and systemic immune response. Live recombinant anthrax vaccines could be used both separately as well as in a prime-boost immunization scheme. The review focuses on immunogenic and protective properties of experimental live genetic engineering prearations, created based on members of geni of Salmonella, Lactobacillus and adenoviruses.


Asunto(s)
Vacunas contra el Carbunco/administración & dosificación , Carbunco/prevención & control , Bacillus anthracis/inmunología , Ingeniería Genética/métodos , Inmunidad Mucosa/efectos de los fármacos , Vacunación , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Carbunco/inmunología , Carbunco/microbiología , Vacunas contra el Carbunco/genética , Vacunas contra el Carbunco/inmunología , Bacillus anthracis/efectos de los fármacos , Bacillus anthracis/genética , Bacillus anthracis/patogenicidad , Vectores Genéticos/inmunología , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Lactobacillus/genética , Lactobacillus/inmunología , Ratones , Probióticos/metabolismo , Probióticos/farmacología , Salmonella/genética , Salmonella/inmunología , Vacunas Atenuadas , Vacunas Sintéticas
9.
Med Microbiol Immunol ; 204(4): 481-91, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25265876

RESUMEN

DNA vaccines are generally weak stimulators of the immune system. Fortunately, their efficacy can be improved using a viral replicon vector or by the addition of immunostimulatory CpG motifs, although the design of these engineered DNA vectors requires optimization. Our results clearly suggest that multiple copies of three types of CpG motifs or combinations of various types of CpG motifs cloned into a viral replicon vector backbone with strong immunostimulatory activities on human PBMC are efficient adjuvants for these DNA vaccines to modulate and enhance protective immunity against anthrax, although modifications with these different CpG forms in vivo elicited inconsistent immune response profiles. Modification with more copies of CpG motifs elicited more potent adjuvant effects leading to the generation of enhanced immunity, which indicated a CpG motif dose-dependent enhancement of antigen-specific immune responses. Notably, the enhanced and/or synchronous adjuvant effects were observed in modification with combinations of two different types of CpG motifs, which provides not only a contribution to the knowledge base on the adjuvant activities of CpG motifs combinations but also implications for the rational design of optimal DNA vaccines with combinations of CpG motifs as "built-in" adjuvants. We describe an efficient strategy to design and optimize DNA vaccines by the addition of combined immunostimulatory CpG motifs in a viral replicon DNA plasmid to produce strong immune responses, which indicates that the CpG-modified viral replicon DNA plasmid may be desirable for use as vector of DNA vaccines.


Asunto(s)
Adyuvantes Inmunológicos/metabolismo , Vacunas contra el Carbunco/inmunología , Carbunco/prevención & control , Oligodesoxirribonucleótidos/metabolismo , Vacunas de ADN/inmunología , Adyuvantes Inmunológicos/genética , Animales , Carbunco/inmunología , Vacunas contra el Carbunco/administración & dosificación , Vacunas contra el Carbunco/genética , Anticuerpos Antibacterianos/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Portadores de Fármacos , Femenino , Vectores Genéticos , Ratones Endogámicos BALB C , Oligodesoxirribonucleótidos/genética , Virus de los Bosques Semliki/genética , Análisis de Supervivencia , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
10.
Mol Gen Mikrobiol Virusol ; (2): 36-40, 2014.
Artículo en Ruso | MEDLINE | ID: mdl-25080817

RESUMEN

Comparative VNTR- and SNP-genotype analysis of four Bacillus anthracis strains (three vaccinal and one virulent) was carried out using modern molecular-genetic typing methods. It is established that these strains formed four SNP patterns completely corresponding to the VNTR-profiles. It was demonstrated that all strains tested except vaccinal B. anthracis STI-1 could not belong by SNP-profile to three global genetic lines of the anthrax agent extended in the world.


Asunto(s)
Vacunas contra el Carbunco/genética , Bacillus anthracis/genética , Genotipo , Repeticiones de Minisatélite , Tipificación de Secuencias Multilocus , Polimorfismo de Nucleótido Simple , Vacunas contra el Carbunco/clasificación , Bacillus anthracis/clasificación , Técnicas de Tipificación Bacteriana
11.
Infect Immun ; 81(1): 278-84, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23115046

RESUMEN

Long-term stability is a desired characteristic of vaccines, especially anthrax vaccines, which must be stockpiled for large-scale use in an emergency situation; however, spontaneous deamidation of purified vaccine antigens has the potential to adversely affect vaccine immunogenicity over time. In order to explore whether spontaneous deamidation of recombinant protective antigen (rPA)--the major component of new-generation anthrax vaccines--affects vaccine immunogenicity, we created a "genetically deamidated" form of rPA using site-directed mutagenesis to replace six deamidation-prone asparagine residues, at positions 408, 466, 537, 601, 713, and 719, with either aspartate, glutamine, or alanine residues. We found that the structure of the six-Asp mutant rPA was not significantly altered relative to that of the wild-type protein as assessed by circular dichroism (CD) spectroscopy and biological activity. In contrast, immunogenicity of aluminum-adjuvanted six-Asp mutant rPA, as measured by induction of toxin-neutralizing antibodies, was significantly lower than that of the corresponding wild-type rPA vaccine formulation. The six-Gln and six-Ala mutants also exhibited lower immunogenicity than the wild type. While the wild-type rPA vaccine formulation exhibited a high level of immunogenicity initially, its immunogenicity declined significantly upon storage at 25°C for 4 weeks. In contrast, the immunogenicity of the six-Asp mutant rPA vaccine formulation was low initially but did not change significantly upon storage. Taken together, results from this study suggest that spontaneous deamidation of asparagine residues predicted to occur during storage of rPA vaccines would adversely affect vaccine immunogenicity and therefore the storage life of vaccines.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Bacillus anthracis/genética , Bacillus anthracis/inmunología , Animales , Carbunco/inmunología , Carbunco/prevención & control , Vacunas contra el Carbunco/genética , Vacunas contra el Carbunco/metabolismo , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Antibacterianos/inmunología , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Neutralizantes/inmunología , Formación de Anticuerpos/genética , Formación de Anticuerpos/inmunología , Antígenos Bacterianos/química , Antígenos Bacterianos/metabolismo , Asparagina/inmunología , Asparagina/metabolismo , Bacillus anthracis/metabolismo , Células Cultivadas , Femenino , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Mutagénesis Sitio-Dirigida/métodos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/metabolismo
12.
Emerg Microbes Infect ; 12(1): 2191741, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36920800

RESUMEN

Pulmonary anthrax is the most fatal clinical form of anthrax and currently available injectable vaccines do not provide adequate protection against it. Hence, next-generation vaccines that effectively induce immunity against pulmonary anthrax are urgently needed. In the present study, we prepared an attenuated and low protease activity Bacillus anthracis strain A16R-5.1 by deleting five of its extracellular protease activity-associated genes and its lef gene through the CRISPR-Cas9 genome editing system. This mutant strain was then used to formulate a lethal toxin (LeTx)-free culture supernatant extract (CSE) anthrax vaccine, of which half was protective antigen (PA). We generated liquid, powder, and powder reconstituted formulations that could be delivered by aerosolized intratracheal inoculation. All of them induced strong humoral, cellular, and mucosal immune responses. The vaccines also produced LeTx neutralizing antibodies and conferred full protection against the lethal aerosol challenges of B. anthracis Pasteur II spores in mice. Compared to the recombinant PA vaccine, the CSE anthrax vaccine with equal PA content provided superior immunoprotection against pulmonary anthrax. The preceding results suggest that the CSE anthrax vaccine developed herein is suitable and scalable for use in inhalational immunization against pulmonary anthrax.


Asunto(s)
Vacunas contra el Carbunco , Carbunco , Bacillus anthracis , Ratones , Animales , Carbunco/prevención & control , Vacunas contra el Carbunco/genética , Antígenos Bacterianos/genética , Polvos , Bacillus anthracis/genética , Vacunas Sintéticas , Péptido Hidrolasas , Anticuerpos Antibacterianos
13.
Microb Pathog ; 53(5-6): 250-8, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22975444

RESUMEN

The current standard for Bacillus anthracis vaccination is the Anthrax Vaccine Adsorbed (AVA, BioThrax). While effective, the licensed vaccine schedule requires five intramuscular injections in the priming series and yearly boosters to sustain protection. One potential approach to maintain or improve the protection afforded by an anthrax vaccine, but requiring fewer doses, is through the use of purified proteins to enhance an antibody response, which could be used on their own or in combination with the current vaccine. This study describes a novel, high-throughput system to amplify and clone every gene in the B. anthracis pXO1 and pXO2 virulence plasmids. We attempted to express each cloned gene in Escherichia coli, and obtained full-length expression of 57% of the proteins. Expressed proteins were then used to identify immunogens using serum from three different mammalian infection models: Dutch-belted rabbits, BALB/c mice, and rhesus macaque monkeys. Ten proteins were detected by antibodies in all of these models, eight of which have not been identified as immunoreactive in other studies to date. Serum was also collected from humans who had received the AVA vaccine, and similar screens showed that antigens that were detected in the infection models were not present in the serum of vaccinated humans, suggesting that antibodies elicited by the current AVA vaccine do not react with the immunoreactive proteins identified in this study. These results will contribute to the future selection of targets in antigenicity and protection studies as one or more of these proteins may prove to be worthy of inclusion in future vaccine preparations.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Antígenos Bacterianos/inmunología , Bacillus anthracis/inmunología , Proteínas Bacterianas/inmunología , Reacciones Cruzadas , Animales , Vacunas contra el Carbunco/genética , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/genética , Bacillus anthracis/genética , Proteínas Bacterianas/genética , Clonación Molecular/métodos , Escherichia coli/genética , Expresión Génica , Vectores Genéticos , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos BALB C , Plásmidos , Conejos
14.
Mol Microbiol ; 75(2): 324-32, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19906175

RESUMEN

Microbial pathogens use adhesive surface proteins to bind to and interact with host tissues, events that are universal for the pathogenesis of infectious diseases. A surface adhesin of Bacillus anthracis, the causative agent of anthrax, required to mediate these steps has not been discovered. Previous work identified BslA, an S-layer protein, to be necessary and sufficient for adhesion of the anthrax vaccine strain, Bacillus anthracis Sterne, to host cells. Here we asked whether encapsulated bacilli require BslA for anthrax pathogenesis in guinea pigs. Compared with the highly virulent parent strain B. anthracis Ames, bslA mutants displayed a dramatic increase in the lethal dose and in mean time-to-death. Whereas all tissues of animals infected with B. anthracis Ames contained high numbers of bacilli, only few vegetative forms could be recovered from internal organs of animals infected with the bslA mutant. Surface display of BslA occurred at the poles of encapsulated bacilli and enabled the binding of vegetative forms to host cells. Together these results suggest that BslA functions as the surface adhesin of the anthrax pathogen B. anthracis strain Ames.


Asunto(s)
Carbunco/inmunología , Bacillus anthracis/genética , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/inmunología , Adhesinas Bacterianas/toxicidad , Animales , Carbunco/patología , Carbunco/prevención & control , Carbunco/transmisión , Vacunas contra el Carbunco/genética , Vacunas contra el Carbunco/inmunología , Vacunas contra el Carbunco/uso terapéutico , Bacillus anthracis/patogenicidad , Adhesión Bacteriana/genética , Adhesión Bacteriana/fisiología , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidad , Cobayas , Células HeLa/microbiología , Humanos , Immunoblotting , Mutación , Factores de Virulencia/deficiencia , Factores de Virulencia/genética , Factores de Virulencia/inmunología
15.
J Virol ; 84(16): 8300-7, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20504926

RESUMEN

Viral vectors based on influenza virus, rabies virus (RV), and vaccinia virus (VV) were used to express large polypeptide segments derived from the Bacillus anthracis protective antigen (PA). For the infectious influenza virus vector and recombinant VV constructs, the receptor binding domain (RBD or domain 4) or the lethal and edema factor binding domain (LEF or domain 1') were engineered into functional chimeric hemagglutinin (HA) glycoproteins. In the case of the RV vector, the viral glycoprotein (G) was used as a carrier for RBD in an inactivated form of the vector. These constructs were examined by using multiple homologous and heterologous prime/boost immunization regimens in order to optimize the induction of alpha-PA antibody responses. Several immunization combinations were shown to induce high titers of antibody recognizing the anthrax RBD and LEF domains, as well as the full-length PA protein in mice. The heterologous prime/boost immunization regimens that involved an initial intranasal administration of a live influenza virus vector, followed by an intramuscular boost with either the killed RV vector or the VV vector, were particularly effective, inducing antigen-specific antibodies at levels severalfold higher than homologous or alternative heterologous protocols. Furthermore, sera from several groups of the immunized mice demonstrated neutralization activity in an in vitro anthrax toxin neutralization assay. In some cases, such toxin-neutralizing activity was notably high, indicating that the mechanisms by which immunity is primed by live influenza virus vectors may have beneficial properties.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Anticuerpos Antibacterianos/inmunología , Anticuerpos Neutralizantes/inmunología , Antígenos Bacterianos/inmunología , Antitoxinas/inmunología , Toxinas Bacterianas/inmunología , Vectores Genéticos , Orthomyxoviridae/genética , Animales , Vacunas contra el Carbunco/genética , Femenino , Inmunización Secundaria/métodos , Ratones , Ratones Endogámicos BALB C , Virus de la Rabia/genética , Vacunación/métodos , Virus Vaccinia/genética
16.
Artículo en Ruso | MEDLINE | ID: mdl-21446166

RESUMEN

AIM: To study the ability of recombinant protective antigen (PA) to stimulate adaptive immune response in laboratory animals. MATERIALS AND METHODS: Vaccine, recombinant, and reference strains of Bacillus anthracis were used in the study. Laboratory animals were immunized subcutaneously with two doses of antigenic preparation or one dose of B. anthracis strain. After inoculation with reference strain of B. anthracis, measurement of LD50 as well as indexes of immunity was performed by specified methods. RESULTS: It was revealed that asporogenic recombinant strain has stable biological characteristics during passages in vitro and is effective producer of PA. Using 2-stage chromatography, highly purified protein was obtained. Experiments on different biomodels--BALB/c mice, guinea pigs, and rabbits--demonstrated high protective activity of PA obtained from asporogenic producer. Increase of immunity index was noted when EA1 protein from S-layer was added to preparation for immunization. CONCLUSION: Immunity indexes determined in experiments on laboratory animals point to high protective efficacy of recombinant PA. Further studies of its interaction with macroorganism's innate and adaptive immunity systems are promising.


Asunto(s)
Vacunas contra el Carbunco/farmacología , Carbunco/inmunología , Carbunco/prevención & control , Antígenos Bacterianos/farmacología , Bacillus anthracis/inmunología , Toxinas Bacterianas/farmacología , Glicoproteínas de Membrana/farmacología , Inmunidad Adaptativa/efectos de los fármacos , Animales , Carbunco/genética , Vacunas contra el Carbunco/genética , Vacunas contra el Carbunco/inmunología , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Bacillus anthracis/genética , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Cobayas , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Conejos
17.
Hum Vaccin Immunother ; 17(2): 560-565, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32614657

RESUMEN

Anthrax is a zoonotic disease caused by the gram-positive spore-forming bacteria Bacillus anthracis. There is a need for safe, highly effective, long-term storage vaccine formulations for mass vaccination. However, the development of new subunit vaccines based on recombinant protective antigen (rPA) faces the problem of vaccine antigen instability. Here, the potential of simultaneous application of two different approaches to stabilize rPA was demonstrated. Firstly, we employed spherical particles (SPs) obtained from the tobacco mosaic virus (TMV). Previously, we had reported that SPs can serve as an adjuvant and platform for antigen presentation. In the current work, SPs were shown to increase the stability of the full-size rPA without loss of its antigenic properties. The second direction was site-specific mutagenesis of asparagine residues to avoid deamidation that causes partial protein degradation. The modified recombinant protein comprising the PA immunogenic domains 3 and 4 (rPA3 + 4) was stable during storage at 4 and 25°C. rPA3 + 4 interacts with antibodies to rPA83 both individually and as a part of a complex with SPs. The results obtained can underpin the development of a recombinant vaccine with a full-size modified rPA (with similar amino acid substitutions that stabilize the protein) and SPs.


Asunto(s)
Vacunas contra el Carbunco , Carbunco , Bacillus anthracis , Toxinas Bacterianas , Carbunco/prevención & control , Vacunas contra el Carbunco/genética , Anticuerpos Antibacterianos , Antígenos Bacterianos/genética , Bacillus anthracis/genética , Toxinas Bacterianas/genética , Humanos , Proteínas Recombinantes/genética
18.
Antimicrob Agents Chemother ; 54(11): 4750-7, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20713663

RESUMEN

Effective measures for the prophylaxis and treatment of anthrax are still required for counteracting the threat posed by inhalation anthrax. In this study, we first demonstrated that the chimeric protein LFn-PA, created by fusing the protective antigen (PA)-binding domain of lethal factor (LFn) to PA, retained the functions of the respective molecules. On the basis of this observation, we attempted to develop an antitoxin that targets the binding of lethal factor (LF) and/or edema factor (EF) to PA and the transportation of LF/EF. Therefore, we replaced PA in LFn-PA with a dominant-negative inhibitory PA (DPA), i.e., PA(F427D). In in vitro models of anthrax intoxication, the LFn-DPA chimera showed 3-fold and 2-fold higher potencies than DPA in protecting sensitive cells against anthrax lethal toxin (LeTx) and edema toxin (EdTx), respectively. In animal models, LFn-DPA exhibited strong potency in rescuing mice from lethal challenge with LeTx. We also evaluated the immunogenicity and immunoprotective efficacy of LFn-DPA as an anthrax vaccine candidate. In comparison with recombinant PA, LFn-DPA induced significantly higher levels of the anti-PA immune response. Moreover, LFn-DPA elicited an anti-LF antibody response that could cross-react with EF. Mice immunized with LFn-DPA tolerated a LeTx challenge that was 5 times its 50% lethal dose. Thus, LFn-DPA represents a highly effective trivalent vaccine candidate for both preexposure and postexposure vaccination. Overall, we have developed a novel and dually functional reagent for the prophylaxis and treatment of anthrax.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Carbunco/inmunología , Antígenos Bacterianos/inmunología , Antitoxinas/inmunología , Toxinas Bacterianas/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Carbunco/prevención & control , Vacunas contra el Carbunco/genética , Vacunas contra el Carbunco/metabolismo , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Antitoxinas/genética , Antitoxinas/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Línea Celular , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Femenino , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
19.
Eur J Immunol ; 39(1): 159-77, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19130551

RESUMEN

Based on the hypothesis that immune outcome can be influenced by the form of antigen administered and its ability to access various antigen-processing pathways, we targeted the 63 kDa fragment of protective antigen (PA) of Bacillus anthracis to various subcellular locations by DNA chimeras bearing a set of signal sequences. These targeting signals, namely, lysosome-associated membrane protein 1 (LAMP1), tissue plasminogen activator (TPA) and ubiquitin, encoded various forms of PA viz. lysosomal, secreted and cytosolic, respectively. Examination of IgG subclass distribution arising as a result of DNA vaccination indicated a higher IgG1:IgG2a ratio whenever the groups were immunized with chimeras bearing TPA, LAMP1 signals alone or when combined together. Importantly, high end-point titers of IgG antibodies were maintained until 24 wk. It was paralleled by high avidity toxin neutralizing antibodies (TNA) and effective cellular adaptive immunity in the systemic compartment. Anti-PA and TNA titers of approximately 10(5) and approximately 10(3), respectively, provided protection to approximately 90% of vaccinated animals in the group pTPA-PA63-LAMP1. A significant correlation was found between survival percentage and post-challenge anti-PA titers and TNA titers. Overall, immune kinetics pointed that differential processing through various compartments gave rise to qualitative differences in the immune response generated by various chimeras.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Carbunco/prevención & control , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Vacunas de ADN/inmunología , Animales , Vacunas contra el Carbunco/administración & dosificación , Vacunas contra el Carbunco/genética , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/genética , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/genética , Biolística , Citocinas/inmunología , Femenino , Proteínas de Membrana de los Lisosomas/genética , Proteínas de Membrana de los Lisosomas/inmunología , Ratones , Ratones Endogámicos BALB C , Linfocitos T Citotóxicos/inmunología , Activador de Tejido Plasminógeno/genética , Activador de Tejido Plasminógeno/inmunología , Ubiquitina/genética , Ubiquitina/inmunología , Vacunación , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética
20.
Mol Ther ; 17(2): 373-9, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19002162

RESUMEN

Bacillus anthracis represents a formidable bioterrorism and biowarfare threat for which new vaccines are needed with improved safety and efficacy over current options. Toward this end, we created recombinant adeno-associated virus type 1 (rAAV1) vectors containing synthetic genes derived from the protective antigen (PA) or lethal factor (LF) of anthrax lethal toxin (LeTx) and tested them for immunogenicity and induction of toxin-neutralizing antibodies in rabbits. Codon-optimized segments encoding activated PA (PA63), or LF, were synthesized and cloned into optimized rAAV1 vectors containing a human cytomegalovirus (hCMV) promoter and synthetic optimized leader. Serum from rabbits immunized intramuscularly with rAAV1/PA (monovalent), rAAV1/LF (monovalent), rAAV1/PA + rAAV1/LF (bivalent), or rAAV1/enhanced green fluorescent protein (control) exhibited substantial PA- and LF-specific antibody responses at 4 weeks by both western blot (> 1:10,000 dilution) and enzyme-linked immunosorbent assay (ELISA) (mean end-point titer: 32,000-260,000), and contained anthrax LeTx-neutralizing activity in vitro, with peak titers approximating those of a rabbit hyperimmune antisera raised against soluble PA and LF. Compared to the monovalent groups (rAAV1/PA or rAAV1/LF), the bivalent group (rAAV1/PA + rAAV1/LF) exhibited marginally higher ELISA and neutralization activity with dual specificity for both PA and LF. The finding of robust neutralizing antibody responses after a single injection of these rAAV1-based vectors supports their further development as candidate anthrax vaccines.


Asunto(s)
Vacunas contra el Carbunco/genética , Dependovirus/genética , Vectores Genéticos/genética , Animales , Vacunas contra el Carbunco/inmunología , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Western Blotting , Citomegalovirus/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Regiones Promotoras Genéticas/genética , Conejos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA