Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 740
Filtrar
1.
Sci Rep ; 12(1): 251, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34997046

RESUMEN

Shigellosis is characterized as diarrheal disease that causes a high mortality rate especially in children, elderly and immunocompromised patients. More recently, the World Health Organization advised safe vaccine designing against shigellosis due to the emergence of Shigella dysenteriae resistant strains. Therefore, the aim of this study is to identify novel drug targets as well as the design of the potential vaccine candidates and chimeric vaccine models against Shigella dysenteriae. A computational based Reverse Vaccinology along with subtractive genomics analysis is one of the robust approaches used for the prioritization of drug targets and vaccine candidates through direct screening of genome sequence assemblies. Herein, a successfully designed peptide-based novel highly antigenic chimeric vaccine candidate against Shigella dysenteriae sd197 strain is proposed. The study resulted in six epitopes from outer membrane WP_000188255.1 (Fe (3+) dicitrate transport protein FecA) that ultimately leads to the construction of twelve vaccine models. Moreover, V9 construct was found to be highly immunogenic, non-toxic, non-allergenic, highly antigenic, and most stable in terms of molecular docking and simulation studies against six HLAs and TLRS/MD complex. So far, this protein and multiepitope have never been characterized as vaccine targets against Shigella dysenteriae. The current study proposed that V9 could be a significant vaccine candidate against shigellosis and to ascertain that further experiments may be applied by the scientific community focused on shigellosis.


Asunto(s)
Antibacterianos/farmacología , Vacunas Bacterianas/farmacología , Diseño de Fármacos , Disentería Bacilar/prevención & control , Shigella dysenteriae/efectos de los fármacos , Desarrollo de Vacunas/métodos , Vacunología/métodos , Animales , Antígenos Bacterianos/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/microbiología , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Diseño Asistido por Computadora , Disentería Bacilar/inmunología , Disentería Bacilar/metabolismo , Disentería Bacilar/microbiología , Epítopos , Interacciones Huésped-Patógeno , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Farmacología en Red , Shigella dysenteriae/inmunología , Shigella dysenteriae/patogenicidad , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/microbiología
2.
Int J Mol Sci ; 23(2)2022 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-35055181

RESUMEN

The clinical symptoms of shigellosis, a gastrointestinal infection caused by Shigella spp. range from watery diarrhea to fulminant dysentery. Endemic infections, particularly among children in developing countries, represent the majority of clinical cases. The situation is aggravated due to the high mortality rate of shigellosis, the rapid dissemination of multi-resistant Shigella strains and the induction of only serotype-specific immunity. Thus, infection prevention due to vaccination, encompassing as many of the circulating serotypes as possible, has become a topic of interest. However, vaccines have turned out to be ineffective so far. Outer membrane vesicles (OMVs) are promising novel targets for vaccination. OMVs are constitutively secreted by Gram-negative bacteria including Shigella during growth. They are composed of soluble luminal portions and an insoluble membrane and can contain toxins, bioactive periplasmic and cytoplasmic (lipo-) proteins, (phospho-) lipids, nucleic acids and/or lipopolysaccharides. Thus, OMVs play an important role in bacterial cell-cell communication, growth, survival and pathogenesis. Furthermore, they modulate the secretion and transport of biomolecules, the stress response, antibiotic resistance and immune responses of the host. Thus, OMVs serve as novel secretion machinery. Here, we discuss the current literature and highlight the properties of OMVs as potent vaccine candidates because of their immunomodulatory, antigenic and adjuvant properties.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/uso terapéutico , Disentería Bacilar/prevención & control , Shigella/crecimiento & desarrollo , Animales , Vacunas Bacterianas/farmacología , Modelos Animales de Enfermedad , Desarrollo de Medicamentos , Disentería Bacilar/inmunología , Humanos , Viabilidad Microbiana/efectos de los fármacos , Shigella/efectos de los fármacos , Shigella/metabolismo , Vacunación
3.
Front Immunol ; 12: 730116, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34745099

RESUMEN

Klebsiella pneumoniae found in the normal flora of the human oral and intestinal tract mainly causes hospital-acquired infections but can also cause community-acquired infections. To date, most clinical trials of vaccines against K. pneumoniae have ended in failure. Furthermore, no single conserved protein has been identified as an antigen candidate to accelerate vaccine development. In this study, we identified five outer membrane proteins of K. pneumoniae, namely, Kpn_Omp001, Kpn_Omp002, Kpn_Omp003, Kpn_Omp004, and Kpn_Omp005, by using reliable second-generation proteomics and bioinformatics. Mice vaccinated with these five KOMPs elicited significantly higher antigen-specific IgG, IgG1, and IgG2a. However, only Kpn_Omp001, Kpn_Omp002, and Kpn_Omp005 were able to induce a protective immune response with two K. pneumoniae infection models. These protective effects were accompanied by the involvement of different immune responses induced by KOMPs, which included KOMPs-specific IFN-γ-, IL4-, and IL17A-mediated immune responses. These findings indicate that Kpn_Omp001, Kpn_Omp002, and Kpn_Omp005 are three potential Th1, Th2, and Th17 candidate antigens, which could be developed into multivalent and serotype-independent vaccines against K. pneumoniae infection.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/farmacología , Vacunas Bacterianas/farmacología , Infecciones por Klebsiella/prevención & control , Klebsiella pneumoniae/inmunología , Desarrollo de Vacunas , Animales , Carga Bacteriana , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/genética , Vacunas Bacterianas/inmunología , Modelos Animales de Enfermedad , Células HL-60 , Humanos , Inmunogenicidad Vacunal , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/genética , Ratones Endogámicos BALB C , Fagocitos/inmunología , Fagocitos/microbiología , Fagocitosis , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología , Linfocitos T/inmunología , Linfocitos T/microbiología , Vacunación , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/farmacología
4.
Front Immunol ; 12: 736360, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34671354

RESUMEN

Bacterial infections cause huge losses in aquaculture and a wide range of health issues in humans. A vaccine is the most economical, efficient, and environment-friendly agent for protecting hosts against bacterial infections. This study aimed to identify broad, cross-protective antigens from the extracellular secretory proteome of the marine bacterium Vibrio alginolyticus. Of the 69 predicted extracellular secretory proteins in its genome, 16 were randomly selected for gene cloning to construct DNA vaccines, which were used to immunize zebrafish (Danio rerio). The innate immune response genes were also investigated. Among the 16 DNA vaccines, 3 (AT730_21605, AT730_22220, and AT730_22910) were protective against V. alginolyticus infection with 47-66.7% increased survival compared to the control, while other vaccines had lower or no protective effects. Furthermore, AT730_22220, AT730_22910, and AT730_21605 also exhibited cross-immune protective effects against Pseudomonas fluorescens and/or Aeromonas hydrophila infection. Mechanisms for cross-protective ability was explored based on conserved epitopes, innate immune responses, and antibody neutralizing ability. These results indicate that AT730_21605, AT730_22220, and AT730_22910 are potential polyvalent vaccine candidates against bacterial infections. Additionally, our results suggest that the extracellular secretory proteome is an antigen pool that can be used for the identification of cross-protective immunogens.


Asunto(s)
Antígenos Bacterianos/farmacología , Infecciones Bacterianas/prevención & control , Vacunas Bacterianas/farmacología , Desarrollo de Vacunas , Vacunas Combinadas/farmacología , Vibrio alginolyticus/inmunología , Animales , Anticuerpos Antibacterianos/metabolismo , Anticuerpos Neutralizantes/metabolismo , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Vacunas Bacterianas/genética , Vacunas Bacterianas/inmunología , Reacciones Cruzadas , Modelos Animales de Enfermedad , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Bacterias Gramnegativas/prevención & control , Inmunidad Innata/efectos de los fármacos , Inmunización , Inmunogenicidad Vacunal , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/prevención & control , Vacunas Combinadas/genética , Vacunas Combinadas/inmunología , Vibriosis/inmunología , Vibriosis/microbiología , Vibriosis/prevención & control , Vibrio alginolyticus/genética , Pez Cebra
5.
Front Immunol ; 12: 698737, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34249004

RESUMEN

Efficacious vaccines are needed to control genital chlamydial diseases in humans and the veterinary industry. We previously reported a C. abortus (Cab) vaccine comprising recombinant Vibrio cholerae ghosts (rVCG) expressing the conserved and immunogenic N-terminal region of the Cab polymorphic membrane protein D (rVCG-Pmp18.1) protein that protected mice against intravaginal challenge. In this study, we investigated the immunomodulatory effect of the hematopoietic progenitor activator cytokine, Fms-like tyrosine kinase 3-ligand (FL) when co-administered with the rVCG-Pmp18.1 vaccine as a strategy to enhance the protective efficacy and the potential mechanism of immunomodulation. Groups of female C57BL/6J mice were immunized and boosted twice intranasally (IN) with rVCG-PmpD18.1 with and without FL or purified rPmp18.1 or rVCG-gD2 (antigen control) or PBS (medium) per mouse. The results revealed that co-administration of the vaccine with FL enhanced antigen-specific cellular and humoral immune responses and protected against live Cab genital infection. Comparative analysis of immune cell phenotypes infiltrating mucosal and systemic immune inductive tissue sites following immunization revealed that co-administration of rVCG-Pmp18.1 with FL significantly enhanced the number of macrophages, dendritic and NK cells, γδ and NK T cells in the spleen (systemic) and iliac lymph nodes (ILN) draining the genital tract (mucosal) tissues compared to rVCG-Pmp18.1 alone. Furthermore, FL enhanced monocyte infiltration in the ILN, while CD19+ B cells and CD4+ T cells were enhanced in the spleen. These results indicate that the immunomodulatory effect of FL is associated with its ability to mobilize innate immune cells and subsequent activation of robust antigen-specific immune effectors in mucosal and systemic lymphoid tissues.


Asunto(s)
Adyuvantes de Vacunas/farmacocinética , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/farmacología , Infecciones por Chlamydia , Proteínas de la Membrana/inmunología , Animales , Chlamydia , Femenino , Ratones , Ratones Endogámicos C57BL , Vibrio cholerae
6.
Front Immunol ; 12: 704621, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34322135

RESUMEN

The lack of tools for the precise manipulation of the tick microbiome is currently a major limitation to achieve mechanistic insights into the tick microbiome. Anti-tick microbiota vaccines targeting keystone bacteria of the tick microbiota alter tick feeding, but their impact on the taxonomic and functional profiles of the tick microbiome has not been tested. In this study, we immunized a vertebrate host model (Mus musculus) with live bacteria vaccines targeting keystone (i.e., Escherichia-Shigella) or non-keystone (i.e., Leuconostoc) taxa of tick microbiota and tested the impact of bacterial-specific antibodies (Abs) on the structure and function of tick microbiota. We also investigated the effect of these anti-microbiota vaccines on mice gut microbiota composition. Our results showed that the tick microbiota of ticks fed on Escherichia coli-immunized mice had reduced Escherichia-Shigella abundance and lower species diversity compared to ticks fed on control mice immunized with a mock vaccine. Immunization against keystone bacteria restructured the hierarchy of nodes in co-occurrence networks and reduced the resistance of the bacterial network to taxa removal. High levels of E. coli-specific IgM and IgG were negatively correlated with the abundance of Escherichia-Shigella in tick microbiota. These effects were not observed when Leuconostoc was targeted with vaccination against Leuconostoc mesenteroides. Prediction of functional pathways in the tick microbiome using PICRUSt2 revealed that E. coli vaccination reduced the abundance of lysine degradation pathway in tick microbiome, a result validated by qPCR. In contrast, the gut microbiome of immunized mice showed no significant alterations in the diversity, composition and abundance of bacterial taxa. Our results demonstrated that anti-tick microbiota vaccines are a safe, specific and an easy-to-use tool for manipulation of vector microbiome. These results guide interventions for the control of tick infestations and pathogen infection/transmission.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Bacterias , Vacunas Bacterianas , Microbioma Gastrointestinal/inmunología , Ixodes , Animales , Bacterias/clasificación , Bacterias/inmunología , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/farmacología , Ixodes/inmunología , Ixodes/microbiología , Ratones
7.
Chem Biol Interact ; 345: 109537, 2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34062171

RESUMEN

The Breast Cancer Resistance Protein (BCRP/ABCG2) is an ATP-binding cassette efflux transporter that is expressed in the apical membrane of cells from relevant tissues involved in drug pharmacokinetics such as liver, intestine, kidney, testis, brain and mammary gland, among others. Tolfenamic acid is an anti-inflammatory drug used as an analgesic and antipyretic in humans and animals. Recently, tolfenamic acid has been repurposed as an antitumoral drug and for use in chronic human diseases such as Alzheimer. The aim of this work was to study whether tolfenamic acid is an in vitro Abcg2 substrate, and to investigate the potential role of Abcg2 in plasma exposure, secretion into milk and tissue accumulation of this drug. Using in vitro transepithelial assays with cells transduced with Abcg2, we showed that tolfenamic acid is an in vitro substrate of Abcg2. The in vivo effect of this transporter was tested using wild-type and Abcg2-/- mice, showing that after oral and intravenous administration of tolfenamic acid, its area under the plasma concentration-time curve in Abcg2-/- mice was between 1.7 and 1.8-fold higher compared to wild-type mice. Abcg2-/- mice also showed higher liver and testis accumulation of tolfenamic acid after intravenous administration. In this study, we demonstrate that tolfenamic acid is transported in vitro by Abcg2 and that its plasma levels as well as its tissue distribution are affected by Abcg2, with potential pharmacological and toxicological consequences.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Vacunas Bacterianas/sangre , Vacunas Bacterianas/farmacocinética , ortoaminobenzoatos/sangre , ortoaminobenzoatos/farmacocinética , Animales , Vacunas Bacterianas/farmacología , Transporte Biológico , Ratones , Distribución Tisular , ortoaminobenzoatos/farmacología
8.
Drugs ; 81(10): 1153-1169, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34097283

RESUMEN

Neisseria gonorrhoeae is the second most common bacterial sexually transmitted infection in the world after Chlamydia trachomatis. The pathogen has developed resistance to every antibiotic currently approved for treatment, and multidrug-resistant strains have been identified globally. The current treatment recommended by the World Health Organization is ceftriaxone and azithromycin dual therapy. However, resistance to azithromycin and ceftriaxone are increasing and treatment failures have been reported. As a result, there is a critical need to develop novel strategies for mitigating the spread of antimicrobial-resistant N. gonorrhoeae through improved diagnosis and treatment of resistant infections. Strategies that are currently being pursued include developing molecular assays to predict resistance, utilizing higher doses of ceftriaxone, repurposing older antibiotics, and developing newer agents. In addition, efforts to discover a vaccine for N. gonorrhoeae have been reignited in recent years with the cross-protectivity provided by the N. meningitidis vaccine, with several new strategies and targets. Despite the significant progress that has been made, there is still much work ahead to combat antimicrobial-resistant N. gonorrhoeae globally.


Asunto(s)
Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Gonorrea/tratamiento farmacológico , Azitromicina/uso terapéutico , Vacunas Bacterianas/farmacología , Ceftriaxona/uso terapéutico , Ensayos Clínicos como Asunto , Farmacorresistencia Bacteriana/genética , Quimioterapia Combinada , Gonorrea/epidemiología , Gonorrea/microbiología , Humanos , Pruebas de Sensibilidad Microbiana , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/metabolismo , Polimorfismo de Nucleótido Simple , Guías de Práctica Clínica como Asunto
9.
Front Immunol ; 12: 644396, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33953716

RESUMEN

The development of effective vaccines and delivery systems in aquaculture is a long-term challenge for controlling emerging and reemerging infections. Cost-efficient and advanced nanoparticle vaccines are of tremendous applicability in prevention of infectious diseases of fish. In this study, dihydrolipoamide dehydrogenase (DLDH) antigens of Vibrio alginolyticus were loaded into mesoporous silica nanoparticles (MSN) to compose the vaccine delivery system. Hydroxypropyl methylcellulose phthalate (HP55) was coated to provide protection of immunogen. The morphology, loading capacity, acid-base triggered release were characterized and the toxicity of nanoparticle vaccine was determined in vitro. Further, the vaccine immune effects were evaluated in large yellow croaker via oral administration. In vitro studies confirmed that the antigen could be stable in enzymes-rich artificial gastric fluid and released under artificial intestinal fluid environment. In vitro cytotoxicity assessment demonstrated the vaccines within 120 µg/ml have good biocompatibility for large yellow croaker kidney cells. Our data confirmed that the nanoparticle vaccine in vivo could elicit innate and adaptive immune response, and provide good protection against Vibrio alginolyticus challenge. The MSN delivery system prepared may be a potential candidate carrier for fish vaccine via oral administration feeding. Further, we provide theoretical basis for developing convenient, high-performance, and cost-efficient vaccine against infectious diseases in aquaculture.


Asunto(s)
Proteínas Bacterianas , Vacunas Bacterianas , Dihidrolipoamida Deshidrogenasa , Enfermedades de los Peces , Nanopartículas , Perciformes , Dióxido de Silicio , Vibriosis , Vibrio alginolyticus , Administración Oral , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/farmacología , Vacunas Bacterianas/química , Vacunas Bacterianas/farmacología , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacología , Dihidrolipoamida Deshidrogenasa/química , Dihidrolipoamida Deshidrogenasa/farmacología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/prevención & control , Nanopartículas/química , Nanopartículas/uso terapéutico , Perciformes/inmunología , Perciformes/microbiología , Porosidad , Dióxido de Silicio/química , Dióxido de Silicio/farmacología , Vibriosis/inmunología , Vibriosis/prevención & control , Vibriosis/veterinaria , Vibrio alginolyticus/enzimología , Vibrio alginolyticus/inmunología
10.
Vet Res ; 52(1): 67, 2021 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-33964969

RESUMEN

Mycoplasma hyopneumoniae (M. hyopneumoniae) is one of the primary agents involved in the porcine respiratory disease complex, economically one of the most important diseases in pigs worldwide. The pathogen adheres to the ciliated epithelium of the trachea, bronchi, and bronchioles, causes damage to the mucosal clearance system, modulates the immune system and renders the animal more susceptible to other respiratory infections. The pathogenesis is very complex and not yet fully understood. Cell-mediated and likely also mucosal humoral responses are considered important for protection, although infected animals are not able to rapidly clear the pathogen from the respiratory tract. Vaccination is frequently practiced worldwide to control M. hyopneumoniae infections and the associated performance losses, animal welfare issues, and treatment costs. Commercial vaccines are mostly bacterins that are administered intramuscularly. However, the commercial vaccines provide only partial protection, they do not prevent infection and have a limited effect on transmission. Therefore, there is a need for novel vaccines that confer a better protection. The present paper gives a short overview of the pathogenesis and immune responses following M. hyopneumoniae infection, outlines the major limitations of the commercial vaccines and reviews the different experimental M. hyopneumoniae vaccines that have been developed and tested in mice and pigs. Most experimental subunit, DNA and vector vaccines are based on the P97 adhesin or other factors that are important for pathogen survival and pathogenesis. Other studies focused on bacterins combined with novel adjuvants. Very few efforts have been directed towards the development of attenuated vaccines, although such vaccines may have great potential. As cell-mediated and likely also humoral mucosal responses are important for protection, new vaccines should aim to target these arms of the immune response. The selection of proper antigens, administration route and type of adjuvant and carrier molecule is essential for success. Also practical aspects, such as cost of the vaccine, ease of production, transport and administration, and possible combination with vaccines against other porcine pathogens, are important. Possible avenues for further research to develop better vaccines and to achieve a more sustainable control of M. hyopneumoniae infections are discussed.


Asunto(s)
Vacunas Bacterianas/farmacología , Mycoplasma hyopneumoniae/efectos de los fármacos , Neumonía Porcina por Mycoplasma/prevención & control , Vacunación/veterinaria , Animales , Neumonía Porcina por Mycoplasma/microbiología , Sus scrofa , Porcinos
11.
Sci Rep ; 11(1): 9769, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33963240

RESUMEN

Shiga-toxin producing Escherichia coli O157:H7 (O157)-based vaccines can provide a potential intervention strategy to limit foodborne zoonotic transmission of O157. While the peripheral antibody response to O157 vaccination has been characterized, O157-specific cellular immunity at the rectoanal junction (RAJ), a preferred site for O157 colonization, remains poorly described. Vaccine induced mucosal O157-specific antibodies likely provide some protection, cellular immune responses at the RAJ may also play a role in protection. Distinct lymphoid follicles were increased in the RAJ of vaccinated/challenged animals. Additionally, increased numbers of interferon (IFN)γ-producing cells and γδ + T cells were detected in the follicular region of the RAJ of vaccinated/challenged animals. Likewise, adjuvanted-vaccine formulation is critical in immunogenicity of the O157 parenteral vaccine. Local T cell produced IFNγ may impact epithelial cells, subsequently limiting O157 adherence, which was demonstrated using in vitro attachment assays with bovine epithelial cells. Thus, distinct immune changes induced at the mucosa of vaccinated and challenged animals provide insight of mechanisms associated with limiting O157 fecal shedding. Enhancing mucosal immunity may be critical in the further development of efficacious vaccines for controlling O157 in ruminants and thus limiting O157 transmission to humans.


Asunto(s)
Vacunas Bacterianas/farmacología , Infecciones por Escherichia coli , Escherichia coli O157/inmunología , Inmunidad Mucosa/efectos de los fármacos , Interferón gamma/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Vacunas Bacterianas/inmunología , Bovinos , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/prevención & control , Infecciones por Escherichia coli/veterinaria , Humanos , Masculino , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología
12.
Sci Rep ; 11(1): 10389, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34001988

RESUMEN

Vaccines based on live attenuated Chlamydia elementary bodies (EBs) can cause disease in vaccinated animals and the comparably safer inactivated whole EBs are only marginally protective. Recent studies show that a vaccine formulation comprising UV-inactivated EBs (EB) and appropriate mucosal delivery systems and/or adjuvants induced significant protective immunity. We tested the hypothesis that intranasal delivery of UV-inactivated C. psittaci EB formulated in Vibrio cholerae ghosts (VCG)-chitosan nanoparticles will induce protective immunity against intranasal challenge in SPF chickens. We first compared the impact of VCG and CpG adjuvants on protective immunity following IN mucosal and IM systemic delivery of EB formulated in chitosan hydrogel/microspheres. Immunologic analysis revealed that IN immunization in the presence of VCG induced higher levels of IFN-γ response than IM delivery or the CpG adjuvanted groups. Also, vaccine efficacy evaluation showed enhanced pharyngeal bacterial clearance and protection against lung lesions with the VCG adjuvanted vaccine formulation, thereby establishing the superior adjuvanticity of VCG over CpG. We next evaluated the impact of different concentrations of VCG on protective immunity following IN mucosal immunization. Interestingly, the adjuvanticity of VCG was concentration-dependent, since protective immunity induced following IN mucosal immunization showed dose-dependent immune responses and protection. These studies reveal that formulation of inactivated chlamydial antigens with adjuvants, such as VCG and chitosan increases their ability to induce protective immune responses against challenge.


Asunto(s)
Quitosano/farmacología , Chlamydophila psittaci/inmunología , Nanopartículas/química , Psitacosis/tratamiento farmacológico , Administración Intranasal , Animales , Antígenos Bacterianos/farmacología , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/farmacología , Pollos/microbiología , Quitosano/química , Chlamydophila psittaci/patogenicidad , Humanos , Inmunidad Mucosa/inmunología , Inyecciones Intramusculares , Interferón gamma/genética , Ratones , Vibrio cholerae/inmunología , Vibrio cholerae/patogenicidad
13.
Front Immunol ; 12: 629480, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868248

RESUMEN

Passive immunization with specific egg yolk antibodies (immunoglobulin Y, IgY) is emerging as a promising alternative to antibiotics to control bacterial infections. Recently, we developed a novel conjugate vaccine that could trigger a strong immune response in rabbits directed against enterobactin (Ent), a highly conserved siderophore molecule utilized by different Gram-negative pathogens. However, induction of Ent-specific antibodies appeared to be affected by the choice of animal host and vaccination regimen. It is still unknown if the Ent conjugate vaccine can trigger a specific immune response in layers for the purpose of production of anti-Ent egg yolk IgY. In this study, three chicken vaccination trials with different regimens were performed to determine conditions for efficient production of anti-Ent egg yolk IgY. Purified Ent was conjugated to three carrier proteins, keyhole limpet hemocyanin (KLH), bovine serum albumin (BSA) and CmeC (a subunit vaccine candidate), respectively. Intramuscular immunization of Barred Rock layers with KLH-Ent conjugate four times induced strong immune response against whole conjugate vaccine but the titer of Ent-specific IgY did not change in yolk with only a 4 fold increase detected in serum. In the second trial, three different Ent conjugate vaccines were evaluated in Rhode Island Red pullets with four subcutaneous injections. The KLH-Ent or CmeC-Ent conjugate consistently induced high level of Ent-specific IgY in both serum (up to 2,048 fold) and yolk (up to 1,024 fold) in each individual chicken. However, the Ent-specific immune response was only temporarily and moderately induced using a BSA-Ent vaccination. In the third trial, ten White Leghorn layers were subcutaneously immunized three times with KLH-Ent, leading to consistent and strong immune response against both whole conjugate and the Ent molecule in each chicken; the mean titer of Ent-specific IgY increased approximately 32 and 256 fold in serum and yolk, respectively. Consistent with its potent binding to various Ent derivatives, the Ent-specific egg yolk IgY also inhibited in vitro growth of a representative Escherichia coli strain. Together, this study demonstrated that the novel Ent conjugate vaccine could induce strong, specific, and robust immune response in chickens. The Ent-specific hyperimmune egg yolk IgY has potential for passive immune intervention against Gram-negative infections.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Vacunas Bacterianas/farmacología , Proteínas del Huevo/inmunología , Yema de Huevo/inmunología , Enterobactina/farmacología , Escherichia coli/efectos de los fármacos , Inmunogenicidad Vacunal , Inmunoglobulinas/sangre , Animales , Vacunas Bacterianas/inmunología , Pollos , Enterobactina/inmunología , Escherichia coli/crecimiento & desarrollo , Escherichia coli/inmunología , Estudios de Factibilidad , Inmunización , Vacunas Conjugadas/inmunología , Vacunas Conjugadas/farmacología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/farmacología
14.
Front Immunol ; 12: 653092, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815413

RESUMEN

Q fever is caused by the obligate intracellular bacterium, Coxiella burnetii, a designated potential agent of bioterrorism because of its route of transmission, resistance to disinfectants, and low infectious dose. The only vaccine licensed for human use is Q-VAX® (Seqirus, licensed in Australia), a formalin-inactivated whole-cell vaccine, which produces severe local and systemic reactogenic responses in previously sensitized individuals. Accordingly, the U.S. Food and Drug Administration and other regulatory bodies around the world, have been reluctant to approve Q-VAX for widespread use. To obviate these adverse reactions, we prepared recombinant protein subunit vaccine candidates containing purified CBU1910, CBU0307, CBU0545, CBU0612, CBU0891, and CBU1398 proteins and TLR triagonist adjuvants. TLR triagonist adjuvants combine different TLR agonists to enhance immune responses to vaccine antigens. We tested both the protective efficacy and reactogenicity of our vaccine candidates in Hartley guinea pigs using intratracheal infection with live C. burnetii. While all of our candidates showed varying degrees of protection during challenge, local reactogenic responses were significantly reduced for one of our vaccine candidates when compared with a formalin-inactivated whole-cell vaccine. Our findings show that subunit vaccines combined with novel TLR triagonist adjuvants can generate protective immunity to C. burnetii infection while reducing reactogenic responses.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacunas Bacterianas/farmacología , Coxiella burnetii/inmunología , Fiebre Q/prevención & control , Receptores Toll-Like/antagonistas & inhibidores , Adyuvantes Inmunológicos/uso terapéutico , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/farmacología , Antígenos Bacterianos/uso terapéutico , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/genética , Vacunas Bacterianas/uso terapéutico , Modelos Animales de Enfermedad , Cobayas , Humanos , Inmunogenicidad Vacunal , Fiebre Q/inmunología , Fiebre Q/microbiología , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Vacunas de Subunidad/genética , Vacunas de Subunidad/farmacología , Vacunas de Subunidad/uso terapéutico , Vacunas Sintéticas/genética , Vacunas Sintéticas/farmacología , Vacunas Sintéticas/uso terapéutico
15.
Methods Mol Biol ; 2283: 1-14, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33765303

RESUMEN

Helicobacter pylori (H. pylori) represents one of the most widespread bacterial infections globally. Infection causes chronic gastritis and increases the risk of peptic ulcer disease, gastric adenocarcinoma, and mucosa-associated lymphoid tissue lymphoma. The pioneering discovery of H. pylori by Marshall and Warren in the early 1980s has initiated fervent research into H. pylori as a pathogen ever since. This chapter aims to provide an overview of our understanding of H. pylori infection and its management, with a focus on current options for diagnosis, the challenges associated with H. pylori eradication, and the need for alternative therapeutic strategies based on furthering our understanding of host: H. pylori interactions.


Asunto(s)
Infecciones por Helicobacter/diagnóstico , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori/patogenicidad , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Vacunas Bacterianas/farmacología , Vacunas Bacterianas/uso terapéutico , Manejo de la Enfermedad , Farmacorresistencia Bacteriana/efectos de los fármacos , Infecciones por Helicobacter/complicaciones , Helicobacter pylori/efectos de los fármacos , Humanos
16.
J Fish Dis ; 44(5): 513-520, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33682163

RESUMEN

Type III secretion system (T3SS)-dependent translocation has been used to deliver heterologous antigens by vaccine carriers into host cells. In this research, we identified the translocation signal of Edwardsiella piscicida T3SS effector EseG and constructed an antibiotic resistance-free balanced-lethal system as attenuated vaccine carrier to present antigens by T3SS. Edwardsiella piscicida LSE40 asd gene deletion mutant was constructed and complemented with pYA3342 harbouring the asd (aspartate ß-semialdehyde dehydrogenase) gene from Salmonella. Fusion proteins composed of EseG N-terminal 1-108 amino acids and the TEM1-ß-lactamase reporter were inserted in plasmid pYA3342. The fusion protein could secrete into the cell culture, translocate into HeLa cells, and localize in the membrane fraction. Then, the double gene deletion mutant LSE40ΔasdΔpurA was constructed as an attenuated vaccine carrier, and Aeromonas hydrophila GapA (glyceraldehyde-3-phosphate dehydrogenase) was fused with the translocation signal, instead of the TEM1-ß-lactamase reporter. The bivalent vaccine could protect blue gourami (Trichogaster trichopterus) against E. piscicida and A. hydrophila, with the relative per cent survival of 80.77% and 63.83%, respectively. These results indicated that EseG N-terminal 1-108 amino acid peptide was the translocation signal of E. piscicida T3SS, which could be used to construct bivalent vaccines based on an attenuated E. piscicida carrier.


Asunto(s)
Aeromonas hydrophila/inmunología , Vacunas Bacterianas/farmacología , Edwardsiella/inmunología , Enfermedades de los Peces/prevención & control , Sistemas de Secreción Tipo III/farmacología , Vacunas Combinadas/farmacología , Animales , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/prevención & control , Infecciones por Enterobacteriaceae/veterinaria , Enfermedades de los Peces/microbiología , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Bacterias Gramnegativas/prevención & control , Infecciones por Bacterias Gramnegativas/veterinaria , Vacunas Atenuadas/farmacología
17.
Fish Shellfish Immunol ; 112: 81-91, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33675991

RESUMEN

Columnaris, a highly contagious bacterial disease caused by Flavobacterium columnare, is recognized as one of the most important infectious diseases in farmed tilapia, especially during the fry and fingerling stages of production. The disease is associated with characteristic lesions in the mucosa of affected fish, particularly their skin and gills. Vaccines delivered via the mucosa are therefore of great interest to scientists developing vaccines for this disease. In the present study, we characterized field isolates of F. columnare obtained from clinical columnaris outbreaks in red tilapia to select an isolate to use as a candidate for our vaccine study. This included characterizing its colony morphology, genotype and virulence status. The isolate was incorporated into a mucoadhesive polymer chitosan-complexed nanovaccine (CS-NE), the efficacy of which was determined by experimentally infecting red tilapia that had been vaccinated with the nanoparticles by immersion. The experimental infection was performed 30-days post-vaccination (dpv), which resulted in 89% of the unvaccinated control fish dying, while the relative percentage survival (RPS) of the CS-NE vaccinated group was 78%. Histology of the mucosal associated lymphoid tissue (MALT) showed a significantly higher presence of leucocytes and a greater antigen uptake by the mucosal epithelium in CS-NE vaccinated fish compared to control fish and whole cell vaccinated fish, respectively, and there was statistically significant up-regulation of IgT, IgM, TNF α, IL1-ß and MHC-1 genes in the gill of the CS-NE vaccinated group. Overall, the results of our study confirmed that the CS-NE particles achieved better adsorption onto the mucosal surfaces of the fish, elicited great vaccine efficacy and modulated the MALT immune response better than the conventional whole cell-killed vaccine, demonstrating the feasibility of the mucoadhesive nano-immersion vaccine as an effective delivery system for the induction of a mucosal immune response against columnaris disease in tilapia.


Asunto(s)
Vacunas Bacterianas/farmacología , Materiales Biomiméticos/farmacología , Cíclidos/inmunología , Enfermedades de los Peces/inmunología , Inmunidad Mucosa , Tejido Linfoide/inmunología , Nanopartículas/administración & dosificación , Animales , Vacunas Bacterianas/administración & dosificación , Materiales Biomiméticos/administración & dosificación , Enfermedades de los Peces/microbiología , Infecciones por Flavobacteriaceae/inmunología , Infecciones por Flavobacteriaceae/microbiología , Infecciones por Flavobacteriaceae/veterinaria , Flavobacterium/fisiología , Tejido Linfoide/efectos de los fármacos , Vacunación/veterinaria
18.
Int J Biol Macromol ; 176: 567-577, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33581203

RESUMEN

Listeria monocytogenes is a cause of infectious food-borne disease in humans, characterized by neurological manifestations, abortion, and neonatal septicemia. It is intracellular bacterium, which limits the development of protective inactivated vacines. Adjuvants capable of stimulating cellular immune response are important tools for developing novel vaccines against intracellular bacteria. The aim of this study was to evaluate the vaccine potential of L. monocytogenes inactivated by gamma irradiation (KLM-γ) encapsulated in alginate microcapsules associated or not with chitosan against listeriosis in the murine model. At the fourth day after challenge there was a reduction in bacterial recovery in mice vaccinated with KLM-γ encapsulated with alginate or alginate-chitosan, with lower bacterial loads in the spleen (10 fold) and liver (100 fold) when compared to non-vaccinated mice. In vitro stimulation of splenocytes from mice vaccinated with alginate-chitosan-encapsulated KLM-γ resulted in lymphocyte proliferation, increase of proportion of memory CD4+ and CD8+ T cell and production of IL-10 and IFN-γ. Interestingly, the group vaccinated with alginate-chitosan-encapsulated KLM-γ had increased survival to lethal infection with lower L. monocytogenes-induced hepatic inflammation and necrosis. Therefore, KLM-γ encapsulation with alginate-chitosan proved to have potential for development of novel and safe inactivated vaccine formulations against listeriosis.


Asunto(s)
Alginatos , Vacunas Bacterianas , Quitosano , Rayos gamma , Listeria monocytogenes , Listeriosis , Alginatos/química , Alginatos/farmacología , Animales , Vacunas Bacterianas/química , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/farmacología , Quitosano/química , Quitosano/farmacología , Modelos Animales de Enfermedad , Femenino , Listeria monocytogenes/química , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/prevención & control , Ratones , Ratones Endogámicos BALB C , Vacunas de Productos Inactivados/química , Vacunas de Productos Inactivados/inmunología , Vacunas de Productos Inactivados/farmacología
19.
PLoS One ; 16(1): e0246141, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33508004

RESUMEN

A novel five year Test and Vaccinate or Remove (TVR) wildlife research intervention project in badgers (Meles meles) commenced in 2014 in a 100km2 area of Northern Ireland. It aimed to increase the evidence base around badgers and bovine TB and help create well-informed and evidence-based strategies to address the issue of cattle-to-cattle spread and spread between cattle and badgers. It involved real-time trap-side testing of captured badgers and vaccinating those that tested negative for bTB (BadgerBCG-BCG Danish 1331) and removal of those that tested bTB positive using the Dual-Path Platform VetTB test (DPP) for cervids (Chembio Diagnostic Systems, Medford, NY USA). Four diagnostic tests were utilised within the study interferon gamma release assay (IGRA), culture (clinical samples and post mortem), DPP using both whole blood and DPP using serum. BCG Sofia (SL222) was used in the final two years because of supply issues with BadgerBCG. Objectives for this study were to evaluate the performance of the DPP in field conditions and whether any trend was apparent in infection prevalence over the study period. A Bayesian latent class model of diagnostic test evaluation in the absence of a gold standard was applied to the data. Temporal variation in the sensitivity of DPP and interferon gamma release assay (IGRA) due to the impact of control measures was investigated using logistic regression and individual variability was assessed. Bayesian latent class analysis estimated DPP with serum to have a sensitivity of 0.58 (95% CrI: 0.40-0.76) and specificity of 0.97 (95% CrI: 0.95-0.98). The DPP with whole blood showed a higher sensitivity (0.69 (95% CrI: 0.48-0.88)) but similar specificity (0.98 (95% Crl: 0.96-0.99)). The change from BCG Danish to BCG Sofia significantly impacted on DPP serum test characteristics. In addition, there was weak evidence of increasing sensitivity of IGRA over time and differences in DPP test sensitivity between adults and cubs. An exponential decline model was an appropriate representation of the infection prevalence over the 5 years, with a starting prevalence of 14% (95% CrI: 0.10-0.20), and an annual reduction of 39.1% (95% CrI: 26.5-50.9). The resulting estimate of infection prevalence in year 5 of the study was 1.9% (95% CrI: 0.8-3.8). These results provide field evidence of a statistically significant reduction in badger TB prevalence supporting a TVR approach to badger intervention. They give confidence in the reliability and reproducibility in the DPP Whole Blood as a real time trap-side diagnostic test for badgers, and describe the effect of vaccination and reduced infection prevalence on test characteristics.


Asunto(s)
Animales Salvajes/microbiología , Vacunas Bacterianas/farmacología , Reservorios de Enfermedades , Modelos Biológicos , Mustelidae/microbiología , Mycobacterium bovis , Tuberculosis Bovina , Vacunación , Animales , Teorema de Bayes , Bovinos , Tuberculosis Bovina/epidemiología , Tuberculosis Bovina/prevención & control , Tuberculosis Bovina/transmisión
20.
Front Immunol ; 12: 760291, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35046936

RESUMEN

Human vaccination against leptospirosis has been relatively unsuccessful in clinical applications despite an expressive amount of vaccine candidates has been tested over years of research. Pathogenic Leptospira encompass a great number of serovars, most of which do not cross-react, and there has been a lack of genetic tools for many years. These obstacles have hampered the understanding of the bacteria's biology and, consequently, the identification of an effective antigen. Thus far, many approaches have been used in an attempt to find a cost-effective and broad-spectrum protective antigen(s) against the disease. In this extensive review, we discuss several strategies that have been used to develop an effective vaccine against leptospirosis, starting with Leptospira-inactivated bacterin, proteins identified in the genome sequences of pathogenic Leptospira, including reverse vaccinology, plasmid DNA, live vaccines, chimeric multi-epitope, and toll- and nod-like receptors agonists. This overview should be able to guide scientists working in the field to select potential antigens and to choose the appropriate formulation to administer the candidates.


Asunto(s)
Vacunas Bacterianas/inmunología , Leptospira/inmunología , Leptospirosis/prevención & control , Desarrollo de Vacunas , Vacunas Bacterianas/farmacología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...