Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
BMC Immunol ; 22(1): 20, 2021 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-33743606

RESUMO

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.


Assuntos
Vacinas contra Antraz/imunologia , Antraz/prevenção & controle , Bacillus anthracis/imunologia , Lipoproteínas/imunologia , Animais , Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/genética , Citocinas/metabolismo , Cobaias , Imunização , Lipoproteínas/administração & dosagem , Lipoproteínas/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Transdução de Sinais , Esporos Bacterianos/imunologia , Receptores Toll-Like/metabolismo
2.
J Pediatric Infect Dis Soc ; 7(4): 270-274, 2018 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-30113691

RESUMO

The Advisory Committee on Immunization Practices (ACIP), a group of medical and public health experts, meets 3 times per year to develop recommendations for vaccine use in the United States. The group has 15 voting members, and each member's term is 4 years. ACIP members and Centers for Disease Control and Prevention staff discuss the epidemiology of vaccine-preventable diseases and vaccine research, effectiveness, safety data, and clinical trial results. Representatives from the American Academy of Pediatrics (J. D. C. and D. W. K.) and the Pediatric Infectious Diseases Society (S. T. O.) are present as liaisons to the ACIP. The ACIP met on June 20 and 21, 2018, to discuss influenza vaccine effectiveness and safety, anthrax vaccination in the setting of a mass exposure, human papillomavirus vaccine, mumps vaccine, Japanese encephalitis vaccine, and pneumococcal vaccination among adults.


Assuntos
Esquemas de Imunização , Vacinação/normas , Vacinas/normas , Vacinas/uso terapêutico , Adolescente , Adulto , Comitês Consultivos , Idoso , Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/efeitos adversos , Vacinas contra Antraz/provisão & distribuição , Criança , Pré-Escolar , Humanos , Lactente , Vacinas contra Influenza/efeitos adversos , Vacinas contra Influenza/uso terapêutico , Vacinas contra Encefalite Japonesa/efeitos adversos , Vacinas contra Encefalite Japonesa/uso terapêutico , Pessoa de Meia-Idade , Vacina contra Caxumba/uso terapêutico , Vacinas contra Papillomavirus/uso terapêutico , Vacinas Pneumocócicas/efeitos adversos , Vacinas Pneumocócicas/uso terapêutico , Vacinação/efeitos adversos , Vacinas/efeitos adversos , Adulto Jovem
3.
J Pediatric Infect Dis Soc ; 7(3): 181-187, 2018 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-29961833

RESUMO

The Advisory Committee on Immunization Practices (ACIP), a group of medical and public health experts, meets 3 times per year to develop recommendations for vaccine use in the United States. There are 15 voting members, and their terms are for 4 years. ACIP members and Centers for Disease Control and Prevention staff discuss the epidemiology of vaccine-preventable diseases and vaccine research, effectiveness, safety data, and clinical trial results. Representatives from the American Academy of Pediatrics (including D. W. K.) and the Pediatric Infectious Diseases Society are present as liaisons to the ACIP. In the February 2018 meeting, important votes on the use of influenza vaccine and hepatitis vaccines were held, and updates on human papillomavirus, meningococcal, and anthrax vaccines, among others, were provided.


Assuntos
Vacinas Virais/uso terapêutico , Adolescente , Adulto , Comitês Consultivos , Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/efeitos adversos , Vacinas contra Antraz/uso terapêutico , Criança , Pré-Escolar , Análise Custo-Benefício , Feminino , Vacinas contra Hepatite A/efeitos adversos , Vacinas contra Hepatite A/provisão & distribuição , Vacinas contra Hepatite A/uso terapêutico , Humanos , Lactente , Vacinas contra Influenza/efeitos adversos , Vacinas contra Influenza/uso terapêutico , Vacinas contra Encefalite Japonesa/efeitos adversos , Vacinas contra Encefalite Japonesa/uso terapêutico , Masculino , Infecções Meningocócicas/tratamento farmacológico , Infecções Meningocócicas/prevenção & controle , Vacinas Meningocócicas/efeitos adversos , Vacinas Meningocócicas/economia , Vacinas Meningocócicas/uso terapêutico , Vacinas contra Papillomavirus/efeitos adversos , Vacinas contra Papillomavirus/uso terapêutico , Usos Terapêuticos , Estados Unidos , Vacinas Virais/efeitos adversos , Adulto Jovem
4.
Vaccine ; 35(18): 2511-2519, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28343781

RESUMO

Anthrax is caused by Bacillus anthracis, a zoonotic bacterial pathogen affecting humans and livestock worldwide. The current human anthrax vaccine, anthrax vaccine adsorbed (AVA), is an injected vaccine with a cumbersome administration schedule and fails to promote mucosal immunity. Bacterial enterotoxins, which stimulate production of the cyclic nucleotide cAMP are effective experimental mucosal vaccine adjuvants, but their inherent toxicity has precluded their use in humans. We investigated whether cyclic dinucleotides that target Stimulator of Interferon Gamma Genes (STING) in mammalian cells could represent an alternative to bacterial enterotoxins as adjuvant for sublingual immunization and promotion of mucosal immunity and secretory IgA responses in addition to systemic immunity. We found that sublingual immunization of mice with Bacillus anthracis protective antigen (PA) and the STING ligand 3'3'-cGAMP promotes PA-specific serum IgG Ab responses of the same magnitude as those induced after immunization with PA and the experimental adjuvant cholera toxin (CT). Interestingly, this STING ligand also promoted serum anti-PA IgA and IgA-producing cells in the bone marrow. Furthermore, the saliva of mice immunized with the STING ligand exhibited similar levels of PA-specific IgA Abs as groups immunized with CT as adjuvant. The adjuvant activity of 3'3'-cGAMP was associated with mixed Th1, Th2, and Th17 responses. This STING ligand also induced rapid IFN-ß and IL-10 responses in sublingual tissues and cervical lymph nodes, and TGF-ß responses in the cervical lymph nodes, which could contribute to promoting IgA responses after sublingual immunization.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas contra Antraz/imunologia , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Proteínas de Membrana/metabolismo , Nucleotídeos Cíclicos/administração & dosagem , Administração Sublingual , Animais , Vacinas contra Antraz/administração & dosagem , Antígenos de Bactérias/administração & dosagem , Antitoxinas/sangue , Toxinas Bacterianas/administração & dosagem , Citocinas/metabolismo , Feminino , Imunidade nas Mucosas , Imunoglobulina A/análise , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Leucócitos Mononucleares/imunologia , Linfonodos/imunologia , Camundongos Endogâmicos C57BL , Saliva/imunologia
5.
Appl Microbiol Biotechnol ; 100(19): 8439-51, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27364624

RESUMO

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.


Assuntos
Vacinas contra Antraz/imunologia , Antígenos de Bactérias/imunologia , Bacillus anthracis/imunologia , Toxinas Bacterianas/imunologia , Proteínas Recombinantes de Fusão/imunologia , Animais , Antraz/prevenção & controle , Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/genética , Anticorpos Antibacterianos/sangue , Anticorpos Neutralizantes/sangue , Antígenos de Bactérias/genética , Antitoxinas/sangue , Toxinas Bacterianas/genética , Linhagem Celular , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Linfócitos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Camundongos , Testes de Neutralização , Proteínas Recombinantes de Fusão/genética , Baço/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
6.
Artigo em Russo | MEDLINE | ID: mdl-27029122

RESUMO

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.


Assuntos
Vacinas contra Antraz/administração & dosagem , Antraz/prevenção & controle , Bacillus anthracis/imunologia , Engenharia Genética/métodos , Imunidade nas Mucosas/efeitos dos fármacos , Vacinação , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Antraz/imunologia , Antraz/microbiologia , Vacinas contra Antraz/genética , Vacinas contra Antraz/imunologia , Bacillus anthracis/efeitos dos fármacos , Bacillus anthracis/genética , Bacillus anthracis/patogenicidade , Vetores Genéticos/imunologia , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Lactobacillus/genética , Lactobacillus/imunologia , Camundongos , Probióticos/metabolismo , Probióticos/farmacologia , Salmonella/genética , Salmonella/imunologia , Vacinas Atenuadas , Vacinas Sintéticas
7.
Clin Vaccine Immunol ; 23(4): 326-38, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26865594

RESUMO

Protective antigen (PA)-specific antibody and cell-mediated immune (CMI) responses to annual and alternate booster schedules of anthrax vaccine adsorbed (AVA; BioThrax) were characterized in humans over 43 months. Study participants received 1 of 6 vaccination schedules: a 3-dose intramuscular (IM) priming series (0, 1, and 6 months) with a single booster at 42 months (4-IM); 3-dose IM priming with boosters at 18 and 42 months (5-IM); 3-dose IM priming with boosters at 12, 18, 30, and 42 months (7-IM); the 1970 licensed priming series of 6 doses (0, 0.5, 1, 6, 12, and 18 months) and two annual boosters (30 and 42 months) administered either subcutaneously (SQ) (8-SQ) or IM (8-IM); or saline placebo control at all eight time points. Antibody response profiles included serum anti-PA IgG levels, subclass distributions, avidity, and lethal toxin neutralization activity (TNA). CMI profiles included frequencies of gamma interferon (IFN-γ)- and interleukin 4 (IL-4)-secreting cells and memory B cells (MBCs), lymphocyte stimulation indices (SI), and induction of IFN-γ, IL-2, IL-4, IL-6, IL-1ß, and tumor necrosis factor alpha (TNF-α) mRNA. All active schedules elicited high-avidity PA-specific IgG, TNA, MBCs, and T cell responses with a mixed Th1-Th2 profile and Th2 dominance. Anti-PA IgG and TNA were highly correlated (e.g., month 7,r(2)= 0.86,P< 0.0001, log10 transformed) and declined in the absence of boosters. Boosters administered IM generated the highest antibody responses. Increasing time intervals between boosters generated antibody responses that were faster than and superior to those obtained with the final month 42 vaccination. CMI responses to the 3-dose IM priming remained elevated up to 43 months. (This study has been registered at ClinicalTrials.gov under registration no. NCT00119067.).


Assuntos
Vacinas contra Antraz/imunologia , Anticorpos Antibacterianos/sangue , Esquemas de Imunização , Imunização Secundária/métodos , Leucócitos Mononucleares/imunologia , Vacinas contra Antraz/administração & dosagem , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Ensaios Clínicos como Assunto , Estudos de Coortes , Citocinas/metabolismo , Humanos , Imunoglobulina G/sangue , Injeções Intramusculares , Injeções Subcutâneas , Testes de Neutralização , Placebos/administração & dosagem
8.
J Immunol ; 196(1): 284-97, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26608924

RESUMO

Nanoparticulate delivery systems for vaccine adjuvants, designed to enhance targeting of secondary lymphoid organs and activation of APCs, have shown substantial promise for enhanced immunopotentiation. We investigated the adjuvant activity of synthetic oligonucleotides containing CpG-rich motifs linked to the sucrose polymer Ficoll, forming soluble 50-nm particles (DV230-Ficoll), each containing >100 molecules of the TLR9 ligand, DV230. DV230-Ficoll was evaluated as an adjuvant for a candidate vaccine for anthrax using recombinant protective Ag (rPA) from Bacillus anthracis. A single immunization with rPA plus DV230-Ficoll induced 10-fold higher titers of toxin-neutralizing Abs in cynomolgus monkeys at 2 wk compared with animals immunized with equivalent amounts of monomeric DV230. Monkeys immunized either once or twice with rPA plus DV230-Ficoll were completely protected from challenge with 200 LD50 aerosolized anthrax spores. In mice, DV230-Ficoll was more potent than DV230 for the induction of innate immune responses at the injection site and draining lymph nodes. DV230-Ficoll was preferentially colocalized with rPA in key APC populations and induced greater maturation marker expression (CD69 and CD86) on these cells and stronger germinal center B and T cell responses, relative to DV230. DV230-Ficoll was also preferentially retained at the injection site and draining lymph nodes and produced fewer systemic inflammatory responses. These findings support the development of DV230-Ficoll as an adjuvant platform, particularly for vaccines such as for anthrax, for which rapid induction of protective immunity and memory with a single injection is very important.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas contra Antraz/imunologia , Antraz/prevenção & controle , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Oligonucleotídeos/imunologia , Infecções Respiratórias/prevenção & controle , Animais , Antraz/imunologia , Antraz/microbiologia , Vacinas contra Antraz/administração & dosagem , Antígenos de Bactérias/genética , Antígenos CD/biossíntese , Antígenos de Diferenciação de Linfócitos T/biossíntese , Linfócitos B/imunologia , Antígeno B7-2/biossíntese , Bacillus anthracis/imunologia , Bacillus anthracis/patogenicidade , Toxinas Bacterianas/genética , Células Dendríticas/imunologia , Ficoll/imunologia , Sequência Rica em GC/genética , Lectinas Tipo C/biossíntese , Macaca fascicularis , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nanopartículas , Neutrófilos/imunologia , Oligonucleotídeos/genética , Proteínas Recombinantes/imunologia , Infecções Respiratórias/imunologia , Infecções Respiratórias/microbiologia , Linfócitos T/imunologia , Vacinação , Vacinas Sintéticas/imunologia
9.
Vaccine ; 33(48): 6745-51, 2015 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-26514421

RESUMO

The current anthrax vaccine requires improvements for rapidly invoking longer-lasting neutralizing antibody responses with fewer doses from a well-defined formulation. Designing antigens that target neutralizing antibody epitopes of anthrax protective antigen, a component of anthrax toxin, may offer a solution for achieving a vaccine that can induce strong and long lasting antibody responses with fewer boosters. Here we report implementation of a strategy for developing epitope focused virus nanoparticle vaccines against anthrax by using immunogenic virus particles to present peptides derived from anthrax toxin previously identified in (1) neutralizing antibody epitope mapping studies, (2) toxin crystal structure analyses to identify functional regions, and (3) toxin mutational analyses. We successfully expressed two of three peptide epitopes from anthrax toxin that, in previous reports, bound antibodies that were partially neutralizing against toxin activity, discovered cross-reactivity between vaccine constructs and toxin specific antibodies raised in goats against native toxin and showed that antibodies induced by our vaccine constructs also cross-react with native toxin. While protection against intoxication in cellular and animal studies were not as effective as in previous studies, partial toxin neutralization was observed in animals, demonstrating the feasibility of using plant-virus nanoparticles as a platform for epitope defined anthrax vaccines.


Assuntos
Vacinas contra Antraz/imunologia , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Portadores de Fármacos , Epitopos/imunologia , Vírus do Mosaico do Tabaco/genética , Animais , Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/genética , Anticorpos Antibacterianos/sangue , Anticorpos Neutralizantes/sangue , Antígenos de Bactérias/genética , Toxinas Bacterianas/genética , Reações Cruzadas , Epitopos/genética , Feminino , Vetores Genéticos , Cabras , Camundongos Endogâmicos C57BL , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
10.
Clin Vaccine Immunol ; 22(4): 430-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25673303

RESUMO

AdVAV is a replication-deficient adenovirus type 5-vectored vaccine expressing the 83-kDa protective antigen (PA83) from Bacillus anthracis that is being developed for the prevention of disease caused by inhalation of aerosolized B. anthracis spores. A noninferiority study comparing the efficacy of AdVAV to the currently licensed Anthrax Vaccine Absorbed (AVA; BioThrax) was performed in New Zealand White rabbits using postchallenge survival as the study endpoint (20% noninferiority margin for survival). Three groups of 32 rabbits were vaccinated with a single intranasal dose of AdVAV (7.5 × 10(7), 1.5 × 10(9), or 3.5 × 10(10) viral particles). Three additional groups of 32 animals received two doses of either intranasal AdVAV (3.5 × 10(10) viral particles) or intramuscular AVA (diluted 1:16 or 1:64) 28 days apart. The placebo group of 16 rabbits received a single intranasal dose of AdVAV formulation buffer. All animals were challenged via the inhalation route with a targeted dose of 200 times the 50% lethal dose (LD50) of aerosolized B. anthracis Ames spores 70 days after the initial vaccination and were followed for 3 weeks. PA83 immunogenicity was evaluated by validated toxin neutralizing antibody and serum anti-PA83 IgG enzyme-linked immunosorbent assays (ELISAs). All animals in the placebo cohort died from the challenge. Three of the four AdVAV dose cohorts tested, including two single-dose cohorts, achieved statistical noninferiority relative to the AVA comparator group, with survival rates between 97% and 100%. Vaccination with AdVAV also produced antibody titers with earlier onset and greater persistence than vaccination with AVA.


Assuntos
Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/imunologia , Antraz/prevenção & controle , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Portadores de Fármacos , Mastadenovirus/genética , Infecções Respiratórias/prevenção & controle , Administração Intranasal , Animais , Antraz/imunologia , Vacinas contra Antraz/genética , Anticorpos Antibacterianos/sangue , Anticorpos Neutralizantes/sangue , Antígenos de Bactérias/genética , Antitoxinas/sangue , Toxinas Bacterianas/genética , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Vetores Genéticos , Imunoglobulina G/sangue , Masculino , Testes de Neutralização , Coelhos , Infecções Respiratórias/imunologia , Análise de Sobrevida , Vacinação/métodos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
11.
Mol Immunol ; 59(1): 91-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24513572

RESUMO

Recombinant bivalent chimeric protein was generated comprising of domain 4 of protective antigen (PA4) and carboxy terminal region of extractable antigen 1 (EA1C) by overlap extension PCR. The immunogenicity and protective efficacy of recombinant chimeric protein (PE) and protein mixture (PAEA) along with the individual components, PA4 and EA1C were evaluated in this study. We found that PE and PAEA exhibited higher endpoint titer and elevated IgG1 response. Compared to PA4 and EA1C, the chimeric protein PE and protein mixture PAEA exhibited 1.52 and 1.39 times more proliferative effect on lymphocytes in vitro. The spore uptake by anti-PE and anti-PAEA antibodies was significantly more than the individual components. We further evaluated the effects of antisera on the toxins in vitro and in vivo. Anti-PE and anti-PAEA antibodies displayed nearly 80% protection against crude toxin activity on RAW 264.7 cell lines. We further demonstrated that the anti-PE and anti-PAEA antibodies displayed better protection in controlling the edema induced by crude toxin. Passive immunization with anti-PE and anti-PAEA provided protection against toxin challenge in mice. The present study reveals that the chimeric protein consisting of heterologous regions of PA and EA1 can render better protection than PA4 or EA1C alone against toxins and bacilli.


Assuntos
Vacinas contra Antraz/imunologia , Antraz/imunologia , Antígenos de Bactérias/imunologia , Bacillus anthracis/imunologia , Proteínas Recombinantes de Fusão/imunologia , Animais , Antraz/microbiologia , Antraz/prevenção & controle , Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/genética , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Bacillus anthracis/genética , Bacillus anthracis/fisiologia , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Linhagem Celular , Proliferação de Células , Sobrevivência Celular/imunologia , Edema/imunologia , Edema/prevenção & controle , Ensaio de Imunoadsorção Enzimática , Feminino , Soros Imunes/imunologia , Imunização/métodos , Imunoglobulina G/imunologia , Linfócitos/citologia , Linfócitos/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/genética , Baço/citologia , Baço/imunologia , Análise de Sobrevida
12.
J Biotechnol ; 176: 1-10, 2014 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-24548460

RESUMO

In concern with frequent recurrence of anthrax in endemic areas and inadvertent use of its spores as biological weapon, the development of an effective anthrax vaccine suitable for both human and veterinary needs is highly desirable. A simple oral delivery through expression in plant system could offer promising alternative to the current methods that rely on injectable vaccines extracted from bacterial sources. In the present study, we have expressed protective antigen (PA) gene in Indian mustard by Agrobacterium-mediated transformation and in tobacco by plastid transformation. Putative transgenic lines were verified for the presence of transgene and its expression by molecular analysis. PA expressed in transgenic lines was biologically active as evidenced by macrophage lysis assay. Intraperitoneal (i.p.) and oral immunization with plant PA in murine model indicated high serum PA specific IgG and IgA antibody titers. PA specific mucosal immune response was noted in orally immunized groups. Further, antibodies indicated lethal toxin neutralizing potential in-vitro and conferred protection against in-vivo toxin challenge. Oral immunization experiments demonstrated generation of immunoprotective response in mice. Thus, our study examines the feasibility of oral PA vaccine expressed in an edible plant system against anthrax.


Assuntos
Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/imunologia , Antraz/imunologia , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/imunologia , Toxinas Bacterianas/metabolismo , Mostardeira/genética , Nicotiana/genética , Administração Oral , Animais , Antraz/prevenção & controle , Antígenos de Bactérias/genética , Bacillus anthracis/imunologia , Toxinas Bacterianas/genética , Cloroplastos/genética , Cloroplastos/metabolismo , Humanos , Imunidade nas Mucosas/imunologia , Imunoglobulina A/sangue , Imunoglobulina A/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Folhas de Planta/genética , Folhas de Planta/metabolismo , Plantas Geneticamente Modificadas , Rhizobium/genética , Rhizobium/metabolismo
13.
Clin Vaccine Immunol ; 21(2): 156-64, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24307239

RESUMO

Developing an effective anthrax vaccine that can induce a rapid and sustained immune response is a priority for the prevention of bioterrorism-associated anthrax infection. Here, we developed a recombinant replication-deficient adenovirus serotype 5-based vaccine expressing the humanized protective antigen (Ad5-PAopt). A single intramuscular injection of Ad5-PAopt resulted in rapid and robust humoral and cellular immune responses in Fisher 344 rats. Animals intramuscularly inoculated with a single dose of 108 infectious units of Ad5-PAopt achieved 100% protection from challenge with 10 times the 50% lethal dose (LD50) of anthrax lethal toxin 7 days after vaccination. Although preexisting intranasally induced immunity to Ad5 slightly weakened the humoral and cellular immune responses to Ad5-PAopt via intramuscular inoculation, 100% protection was achieved 15 days after vaccination in Fisher 344 rats. The protective efficacy conferred by intramuscular vaccination in the presence of preexisting intranasally induced immunity was significantly better than that of intranasal delivery of Ad5-PAopt and intramuscular injection with recombinant PA and aluminum adjuvant without preexisting immunity. As natural Ad5 infection often occurs via the mucosal route, the work here largely illuminates that intramuscular inoculation with Ad5-PAopt can overcome the negative effects of immunity induced by prior adenovirus infection and represents an efficient approach for protecting against emerging anthrax.


Assuntos
Adenoviridae/genética , Vacinas contra Antraz/imunologia , Antraz/prevenção & controle , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Vetores Genéticos , Animais , Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/genética , Antígenos de Bactérias/genética , Toxinas Bacterianas/genética , Linhagem Celular , Modelos Animais de Doenças , Feminino , Humanos , Imunidade Celular , Imunidade Humoral , Injeções Intramusculares , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Análise de Sobrevida , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
14.
J Control Release ; 173: 148-57, 2014 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-24188959

RESUMO

Aluminum hydroxide is used as a vaccine adjuvant in various human vaccines. Unfortunately, despite its favorable safety profile, aluminum hydroxide can only weakly or moderately potentiate antigen-specific antibody responses. When dispersed in an aqueous solution, aluminum hydroxide forms particulates of 1-20µm. There is increasing evidence that nanoparticles around or less than 200nm as vaccine or antigen carriers have a more potent adjuvant activity than large microparticles. In the present study, we synthesized aluminum hydroxide nanoparticles of 112nm. Using ovalbumin and Bacillus anthracis protective antigen protein as model antigens, we showed that protein antigens adsorbed on the aluminum hydroxide nanoparticles induced a stronger antigen-specific antibody response than the same protein antigens adsorbed on the traditional aluminum hydroxide microparticles of around 9.3µm. The potent adjuvant activity of the aluminum hydroxide nanoparticles was likely related to their ability to more effectively facilitate the uptake of the antigens adsorbed on them by antigen-presenting cells. Finally, the local inflammation induced by aluminum hydroxide nanoparticles in the injection sites was milder than that induced by microparticles. Simply reducing the particle size of the traditional aluminum hydroxide adjuvant into nanometers represents a novel and effective approach to improve its adjuvanticity.


Assuntos
Adjuvantes Imunológicos/farmacologia , Hidróxido de Alumínio/imunologia , Vacinas contra Antraz/imunologia , Antígenos de Bactérias/imunologia , Bacillus anthracis/imunologia , Toxinas Bacterianas/imunologia , Nanopartículas/ultraestrutura , Ovalbumina/imunologia , Adjuvantes Imunológicos/administração & dosagem , Hidróxido de Alumínio/administração & dosagem , Animais , Antraz/imunologia , Antraz/prevenção & controle , Vacinas contra Antraz/administração & dosagem , Formação de Anticorpos , Antígenos de Bactérias/administração & dosagem , Toxinas Bacterianas/administração & dosagem , Linhagem Celular , Células Cultivadas , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nanopartículas/administração & dosagem , Nanopartículas/metabolismo , Ovalbumina/administração & dosagem , Tamanho da Partícula , Vacinas/administração & dosagem , Vacinas/imunologia
15.
Hum Vaccin Immunother ; 9(3): 544-52, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23324615

RESUMO

The potential use of Bacillus anthracis as a bioterrorism weapon threatens the security of populations globally, requiring the immediate availability of safe, efficient and easily delivered anthrax vaccine for mass vaccination. Extensive research efforts have been directed toward the development of recombinant subunit vaccines based on protective antigen (PA), the principal virulence factor of B. anthracis. Among the emerging technologies for the production of these vaccine antigens is our launch vector-based plant transient expression system. Using this system, we have successfully engineered, expressed, purified and characterized full-length PA (pp-PA83) in Nicotiana benthamiana plants using agroinfiltration. This plant-produced antigen elicited high toxin neutralizing antibody titers in mice and rabbits after two vaccine administrations with Alhydrogel. In addition, immunization with this vaccine candidate protected 100% of rabbits from a lethal aerosolized B. anthracis challenge. The vaccine effects were dose-dependent and required the presence of Alhydrogel adjuvant. In addition, the vaccine antigen formulated with Alhydrogel was stable and retained immunogenicity after two-week storage at 4°C, the conditions intended for clinical use. These results support the testing of this vaccine candidate in human volunteers and the utility of our plant expression system for the production of a recombinant anthrax vaccine.


Assuntos
Vacinas contra Antraz/imunologia , Antraz/prevenção & controle , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/imunologia , Adjuvantes Imunológicos/administração & dosagem , Aerossóis , Hidróxido de Alumínio/administração & dosagem , Animais , Antraz/imunologia , Vacinas contra Antraz/administração & dosagem , Anticorpos Antibacterianos/sangue , Anticorpos Neutralizantes/sangue , Antígenos de Bactérias/genética , Antígenos de Bactérias/isolamento & purificação , Toxinas Bacterianas/genética , Toxinas Bacterianas/isolamento & purificação , Modelos Animais de Doenças , Exposição por Inalação , Camundongos Endogâmicos BALB C , Plantas Geneticamente Modificadas/genética , Coelhos , Análise de Sobrevida , Nicotiana/genética , Resultado do Tratamento , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
16.
Biochim Biophys Acta ; 1830(3): 2804-12, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23201204

RESUMO

BACKGROUND: The poly-gamma-D-glutamic acid (PGA) capsule, a major virulence factor of Bacillus anthracis, protects bacilli from immune surveillance and allows its unimpeded growth in the host. Recently, the importance of the PGA in the pathogenesis of anthrax infection has been reported. The PGA capsule is associated with lethal toxin (LT) in the blood of experimentally infected animals and enhances the cytotoxicity of LT. METHODS: To investigate the role of anti-PGA Abs on progression of anthrax infection, two mouse anti-PGA mAbs with K(d) values of 0.8 microM and 2.6 microM respectively were produced and in silico three dimensional (3D) models of mAbs with their cognitive PGA antigen complex were analyzed. RESULTS: Anti-PGA mAbs specifically bound encapsulated B. anthracis H9401 and showed opsonophagocytosis activity against the bacteria with complement. The enhancement effect of PGA on LT-mediated cytotoxicity was confirmed ex vivo using mouse bone marrow-derived macrophages and was effectively inhibited by anti-PGA mAb. Passive immunization of mAb completely protected mice from PGA-enhanced LT toxicity and partially rescued mice from anthrax spore challenges. 3D structure models of these mAbs and PGA complex support specific interactions between CDR and cognitive PGA. These results indicate that mouse mAb against PGA capsule prevents the progress of anthrax disease not only by eliminating the vegetative form of encapsulated B. anthracis but also by inhibiting the enhanced cytotoxic activity of LT by PGA through specific binding with PGA capsule antigen. GENERAL SIGNIFICANCE: Our results suggest a potential role for PGA antibodies in preventing and treating anthrax infection.


Assuntos
Vacinas contra Antraz/administração & dosagem , Antraz/prevenção & controle , Anticorpos Antibacterianos/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Antígenos de Bactérias/imunologia , Cápsulas Bacterianas/imunologia , Imunização Passiva , Ácido Poliglutâmico/análogos & derivados , Animais , Antraz/imunologia , Antraz/microbiologia , Antraz/mortalidade , Anticorpos Antibacterianos/biossíntese , Anticorpos Monoclonais/biossíntese , Bacillus anthracis/efeitos dos fármacos , Bacillus anthracis/imunologia , Bacillus anthracis/patogenicidade , Toxinas Bacterianas/imunologia , Células Cultivadas , Feminino , Humanos , Cinética , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Simulação de Acoplamento Molecular , Ácido Poliglutâmico/antagonistas & inibidores , Ácido Poliglutâmico/imunologia , Esporos Bacterianos/efeitos dos fármacos , Esporos Bacterianos/imunologia , Esporos Bacterianos/patogenicidade , Análise de Sobrevida , Fatores de Virulência/antagonistas & inibidores , Fatores de Virulência/imunologia
17.
Clin Vaccine Immunol ; 20(1): 1-8, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23100479

RESUMO

Bacillus anthracis is the causative agent of anthrax, and its spores have been developed into lethal bioweapons. To mitigate an onslaught from airborne anthrax spores that are maliciously disseminated, it is of paramount importance to develop a rapid-response anthrax vaccine that can be mass administered by nonmedical personnel during a crisis. We report here that intranasal instillation of a nonreplicating adenovirus vector encoding B. anthracis protective antigen could confer rapid and sustained protection against inhalation anthrax in mice in a single-dose regimen in the presence of preexisting adenovirus immunity. The potency of the vaccine was greatly enhanced when codons of the antigen gene were optimized to match the tRNA pool found in human cells. In addition, an adenovirus vector encoding lethal factor can confer partial protection against inhalation anthrax and might be coadministered with a protective antigen-based vaccine.


Assuntos
Administração Intranasal , Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/imunologia , Antraz/prevenção & controle , Adenoviridae/genética , Animais , Antraz/imunologia , Bacillus anthracis/genética , Modelos Animais de Doenças , Portadores de Fármacos , Feminino , Vetores Genéticos , Camundongos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
18.
Vaccine ; 30(5): 846-52, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22172509

RESUMO

Bacillus anthracis, the causative agent of anthrax, is recognized as one of the most serious bioterrorism threats. The current human vaccines are based on the protective antigen component of the anthrax toxins. Concern about possible vaccine resistant strains and reliance on a single antigen has prompted the search for additional immunogens. Bacterial capsules, as surface-expressed virulence factors, are well-established components of several licensed vaccines. In a previous study we showed that an anthrax vaccine consisting of the B. anthracis poly-γ-D-glutamic acid capsule covalently conjugated to the outer membrane protein complex of Neisseria meningitidis serotype B protected mice against parenteral B. anthracis challenge. Here we tested this vaccine in rabbits and monkeys against an aerosol spore challenge. The vaccine induced anti-capsule antibody responses in both species, measured by ELISA and a macrophage opsono-adherence assay. While rabbits were not protected against a high aerosol challenge dose, significant protection was observed in monkeys receiving the capsule conjugate vaccine. The results confirm that the capsule is a protective immunogen against anthrax, being the first non-toxin antigen shown to be efficacious in monkeys and suggest that addition of capsule may broaden and enhance the protection afforded by protective antigen-based vaccines.


Assuntos
Vacinas contra Antraz/imunologia , Antraz/prevenção & controle , Cápsulas Bacterianas/imunologia , Proteínas da Membrana Bacteriana Externa/química , Animais , Antraz/imunologia , Vacinas contra Antraz/administração & dosagem , Anticorpos Antibacterianos/sangue , Cápsulas Bacterianas/química , Proteínas da Membrana Bacteriana Externa/isolamento & purificação , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Macaca mulatta , Macrófagos/imunologia , Masculino , Neisseria meningitidis/química , Proteínas Opsonizantes/sangue , Fagocitose , Coelhos , Análise de Sobrevida , Vacinas Conjugadas/administração & dosagem , Vacinas Conjugadas/imunologia
20.
Int Immunopharmacol ; 11(9): 1195-204, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21492746

RESUMO

Anthrax is an infectious disease caused by Bacillus anthracis. The currently licensed human anthrax vaccines contain protective antigen (PA) as a major protective component and alum as an adjuvant. In this study, we investigated whether CIA05, a TLR4 agonist, is able to promote the immune response to an anthrax vaccine adjuvanted with alum. BALB/c mice were immunized intraperitoneally three times at 2-week intervals with a recombinant B. anthracis PA alone or in combination with CIA05 in the absence or presence of alum, and immune responses were determined 2 or 3 weeks after the third immunization. The results showed that the combination of CIA05 and alum significantly increased both serum anti-PA IgG antibody and toxin-neutralizing antibody titers, and the adjuvant effects were greater when lower antigen doses were used for immunization. Both CIA05 and alum stimulated PA-specific splenocyte secretion of interleukin (IL)-4, IL-5, and IL-6. A combination of the two yielded synergistic effects on IL-4 secretion, but CIA05 tended to repress IL-5 and IL-6 secretions induced by alum. Co-administration of CIA05 and alum also increased GL7 expression in B220(+)CD24(+) splenic cells, indicating the ability to activate B cells. These data suggest that CIA05, combined with alum, could be used to achieve higher immune responses to PA, leading to the development of an effective anthrax vaccine.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Compostos de Alúmen/farmacologia , Bacillus anthracis/imunologia , Substâncias Protetoras/administração & dosagem , Receptor 4 Toll-Like/agonistas , Animais , Antraz/imunologia , Antraz/prevenção & controle , Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/imunologia , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Antígenos de Bactérias/imunologia , Antígenos de Diferenciação/imunologia , Linfócitos B/imunologia , Toxinas Bacterianas/imunologia , Imunização/métodos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Interleucinas/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Baço/efeitos dos fármacos , Baço/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA