Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 329
Filtrar
1.
Infect Immun ; 89(10): e0034221, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34227839

RESUMEN

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


Asunto(s)
Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Vacunas Combinadas/inmunología , Animales , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Reacciones Cruzadas/inmunología , Eritrocitos/inmunología , Eritrocitos/microbiología , Exotoxinas/inmunología , Proteínas Hemolisinas/inmunología , Humanos , Inmunización/métodos , Leucocidinas/inmunología , Neutrófilos/inmunología , Neutrófilos/microbiología , Conejos , Infecciones Estafilocócicas/microbiología , Toxoides/inmunología
2.
MAbs ; 13(1): 1869406, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33427589

RESUMEN

Due to their shared genetic history, antibodies from the same clonotype often bind to the same epitope. This knowledge is used in immune repertoire mining, where known binders are used to search bulk sequencing repertoires to identify new binders. However, current computational methods cannot identify epitope convergence between antibodies from different clonotypes, limiting the sequence diversity of antigen-specific antibodies that can be identified. We describe how the antibody binding site, the paratope, can be used to cluster antibodies with common antigen reactivity from different clonotypes. Our method, paratyping, uses the predicted paratope to identify these novel cross clonotype matches. We experimentally validated our predictions on a pertussis toxoid dataset. Our results show that even the simplest abstraction of the antibody binding site, using only the length of the loops involved and predicted binding residues, is sufficient to group antigen-specific antibodies and provide additional information to conventional clonotype analysis. Abbreviations: BCR: B-cell receptor; CDR: complementarity-determining region; PTx: pertussis toxoid.


Asunto(s)
Anticuerpos/inmunología , Antígenos/inmunología , Sitios de Unión de Anticuerpos/inmunología , Biología Computacional/métodos , Programas Informáticos , Toxoides/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Células Clonales/inmunología , Regiones Determinantes de Complementariedad/inmunología , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Ratones , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo , Análisis de la Célula Individual/métodos
3.
Arch Razi Inst ; 75(3): 385-395, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33025779

RESUMEN

Toxoid vaccines can provide protective immunity against clostridial diseases. Since the duration of the toxoid vaccine immunogenicity is short, these vaccines need to contain an adjuvant. The nanoparticles of chitosan can stimulate humoral and cell-mediated immune responses. In the present study, the effect of chitosan nanoparticles was investigated on the immunogenicity of the pentavalent clostridial toxoid vaccine containing Clostridium perfringens types D, C, and B, Clostridium septicum, as well as Clostridium novyi. Rabbits were immunized by two injections with 3-week intervals and checked clinically and through autopsy 2 weeks after the last injection. Hematological changes were investigated during immunization, including the changes of white and red blood cell counts, hemoglobin, packed cell volume, platelet, neutrophil, lymphocyte, eosinophil, basophile, monocyte, and Neut/Lymph. Biochemical factors, namely creatinine, blood urea nitrogen, glucose, alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, total protein, and albumin, were also studied. The changes in immune responses during the immunization period were investigated through indirect enzyme-linked immunosorbent assay (ELISA). The results of ELISA showed that chitosan significantly enhanced immunogenicity when accompanied with in the pentavalent clostridial toxoid vaccine. During the immunogenicity period and following that, no changes were observed in clinical behavior and internal organs after autopsy. The hematological and biochemical factors were reported with no significant pathologic changes during immunization in the control and vaccinated groups (p <0.05). The obtained findings revealed that the toxoid vaccines could not induce significant physiological changes in the body. The vaccine containing chitosan could stimulate humoral immunity 2-3 times higher than the nonchitosan vaccine. The humoral immune response was significantly duplicated due to the chitosan effect. Chitosan not only had no local or general side effects but also could be a good help with the enhancement of the immune system; therefore, it can be recommended as an appropriate safe adjuvant in the development of toxoid vaccines.


Asunto(s)
Vacunas Bacterianas/inmunología , Quitosano/inmunología , Clostridium perfringens/inmunología , Clostridium septicum/inmunología , Clostridium/inmunología , Nanopartículas/administración & dosificación , Vacunas Combinadas/inmunología , Quitosano/administración & dosificación , Inmunogenicidad Vacunal/inmunología , Toxoides/inmunología
4.
J Exp Med ; 217(9)2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32602902

RESUMEN

Staphylococcus aureus is responsible for various diseases in humans, and recurrent infections are commonly observed. S. aureus produces an array of bicomponent pore-forming toxins that target and kill leukocytes, known collectively as the leukocidins. The contribution of these leukocidins to impair the development of anti-S. aureus adaptive immunity and facilitate reinfection is unclear. Using a murine model of recurrent bacteremia, we demonstrate that infection with a leukocidin mutant results in increased levels of anti-S. aureus antibodies compared with mice infected with the WT parental strain, indicating that leukocidins negatively impact the generation of anti-S. aureus antibodies in vivo. We hypothesized that neutralizing leukocidin-mediated immune subversion by vaccination may shift this host-pathogen interaction in favor of the host. Leukocidin-immunized mice produce potent leukocidin-neutralizing antibodies and robust Th1 and Th17 responses, which collectively protect against bloodstream infections. Altogether, these results demonstrate that blocking leukocidin-mediated immune evasion can promote host protection against S. aureus bloodstream infection.


Asunto(s)
Bacteriemia/inmunología , Bacteriemia/prevención & control , Evasión Inmune , Leucocidinas/metabolismo , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Formación de Anticuerpos/inmunología , Bacteriemia/sangre , Bacteriemia/microbiología , Linfocitos T CD4-Positivos/inmunología , Citocinas/sangre , Interacciones Huésped-Patógeno/inmunología , Inmunidad , Inmunización , Inmunoglobulina G/sangre , Inflamación/patología , Staphylococcus aureus Resistente a Meticilina/inmunología , Ratones , Modelos Biológicos , Especificidad de Órganos , Recurrencia , Bazo/patología , Infecciones Estafilocócicas/sangre , Toxoides/inmunología
5.
Fish Shellfish Immunol ; 105: 310-318, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32702476

RESUMEN

Oral vaccines are highly demanded by aquaculture sector that requires alternatives to injectable vaccines, involving fish handling, stress-related immunosuppression and mortalities. However, most previous attempts to obtain effective oral vaccines have failed due to a restricted tolerance mechanisms in intestine, whose mucosa is at the frontline of antigen encounter and has to balance the equilibrium between tolerance and immunity in a microbe-rich environment. Thus, the search for oral adjuvants that could augment immune responses triggered by antigens allowing them to circumvent intestinal tolerance is of great relevance. The present work focuses on the adjuvant potential of the Escherichia coli LT(R192G/L211A) toxoid (dmLT). To undertake an initial screening of the potential that dmLT has as an oral adjuvant in rainbow trout (Oncorhynchus mykiss), we have analyzed its transcriptional effects alone or in combination with Aeromonas salmonicida subsp. salmonicida or viral hemorrhagic septicemia virus (VHSV) on rainbow trout intestinal epithelial cell line RTgutGC and gut explants. Our results show that although dmLT provoked no significant effects by itself, it increased the transcription of pro-inflammatory cytokines and antimicrobial genes induced by the bacteria. In contrast, when combined with VHSV, dmLT only increased the transcription of Mx and the intracellular adhesion molecule 1 (ICAM1). Therefore, the protocol designed is an effective method to initially evaluate the effects of potential oral adjuvants, and points to dmLT as an effective adjuvant for oral antibacterial vaccines.


Asunto(s)
Adyuvantes Inmunológicos/metabolismo , Escherichia coli/inmunología , Oncorhynchus mykiss/inmunología , Toxoides/inmunología , Aeromonas/fisiología , Animales , Línea Celular , Mucosa Intestinal/inmunología , Novirhabdovirus/fisiología , Oncorhynchus mykiss/genética , Transcripción Genética/inmunología
6.
Vaccine ; 37(43): 6405-6414, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31515145

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) causes diarrhoea by secreting enterotoxins into the small intestine. Human ETEC strains may secrete any combination of three enterotoxins: the heat-labile toxin (LT) and the heat-stable toxins (ST), of which there are two variants, called human ST (STh) and porcine ST (STp). Strains expressing STh, either alone or in combination with LT and/or STp, are among the four most important diarrhoea-causing pathogens affecting children in low- and middle-income countries. ST is therefore an attractive target for ETEC vaccine development. To produce a safe ST-based vaccine, several challenges must be solved. ST must be rendered immunogenic and non-toxic, and antibodies elicited by an ST vaccine should neutralize ST but not cross-react with the endogenous ligands uroguanylin and guanylin. Virus-like particles (VLPs) tend to be highly immunogenic and are increasingly being used as carriers for presenting heterologous antigens in new vaccines. In this study, we have coupled native STh and the STh-A14T toxoid to the coat protein of Acinetobacter phage AP205 by using the SpyCatcher system and immunized mice with these VLPs without the use of adjuvants. We found that both STs were efficiently coupled to the VLP, that both the STh and STh-A14T VLPs were immunogenic in mice, and that the resulting serum antibodies could completely neutralize the toxic activities of native STh. The serum antibodies showed a high degree of immunological cross-reaction to STp, while there was little or no unwanted cross-reaction to uroguanylin and guanylin. Moreover, compared to native STh, the STh-A14T mutation did not seem to negatively impact the immunogenicity of the construct or the neutralizing ability of the resulting sera. Taken together, these findings demonstrate that VLPs are suitable carriers for making STs immunogenic, and that the STh-A14T-coupled AP205 VLP represents a promising ETEC vaccine candidate.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Escherichia coli Enterotoxigénica/inmunología , Vacunas contra Escherichia coli/inmunología , Toxoides/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Acinetobacter/virología , Animales , Anticuerpos Neutralizantes/sangre , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/inmunología , Bacteriófagos , Reacciones Cruzadas , Infecciones por Escherichia coli/prevención & control , Vacunas contra Escherichia coli/administración & dosificación , Femenino , Hormonas Gastrointestinales/inmunología , Inmunización , Ratones , Ratones Endogámicos BALB C , Péptidos Natriuréticos/inmunología , Toxoides/administración & dosificación , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación
7.
Toxins (Basel) ; 11(6)2019 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-31207937

RESUMEN

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


Asunto(s)
Proteínas Bacterianas/inmunología , Vacunas Bacterianas , Leucocidinas/inmunología , Infecciones Estafilocócicas/prevención & control , Toxoides/inmunología , Animales , Proteínas Bacterianas/genética , Supervivencia Celular , Escherichia coli/genética , Femenino , Células HL-60 , Humanos , Leucocidinas/genética , Ratones Endogámicos ICR , Monocitos , Células THP-1 , Toxoides/genética
8.
Nano Lett ; 19(7): 4760-4769, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31184899

RESUMEN

Infections caused by multidrug-resistant Gram-negative bacteria have emerged as a major threat to public health worldwide. The high mortality and prevalence, along with the slow pace of new antibiotic discovery, highlight the necessity for new disease management paradigms. Here, we report on the development of a multiantigenic nanotoxoid vaccine based on macrophage membrane-coated nanoparticles for eliciting potent immunity against pathogenic Pseudomonas aeruginosa. The design of this biomimetic nanovaccine leverages the specific role of macrophages in clearing pathogens and their natural affinity for various virulence factors secreted by the bacteria. It is demonstrated that the macrophage nanotoxoid is able to display a wide range of P. aeruginosa antigens, and the safety of the formulation is confirmed both in vitro and in vivo. When used to vaccinate mice via different administration routes, the nanotoxoid is capable of eliciting strong humoral immune responses that translate into enhanced protection against live bacterial infection in a pneumonia model. Overall, the work presented here provides new insights into the design of safe, multiantigenic antivirulence vaccines using biomimetic nanotechnology and the application of these nanovaccines toward the prevention of difficult-to-treat Gram-negative infections.


Asunto(s)
Vacunas Bacterianas , Farmacorresistencia Bacteriana , Infecciones por Pseudomonas , Pseudomonas aeruginosa/inmunología , Toxoides , Vacunación , Factores de Virulencia/inmunología , Animales , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/inmunología , Inmunidad Humoral/efectos de los fármacos , Ratones , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/patología , Infecciones por Pseudomonas/prevención & control , Pseudomonas aeruginosa/patogenicidad , Toxoides/inmunología , Toxoides/farmacología
9.
Anaerobe ; 59: 72-75, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31129336

RESUMEN

The aim of this study was to evaluate the titers of neutralizing antibodies in cattle inoculated with multivalent commercial clostridial vaccines containing C. botulinum type C (BoNTC), C. botulinum type D (BoNTD), and C. perfringens epsilon (ETX) toxoids for a period of one year. Cattle (Bos taurus), aged 4-6 months and not previously immunized, were vaccinated under four different protocols at days 0 and 30 and followed over one year. Individual serum titration was performed by a serum neutralization test in mice or in MDCK cells. The number of animals with detectable neutralizing antibodies ranged from 40.6% to 78.1%, but only 12.5% of animals showed neutralizing antibodies against all tested antigens. Neutralizing antibodies were found only until 60 days for ETX, 120 days for BoNTC, and 180 days for BoNTD. The absence of detectable neutralizing antibodies against the three antigens before 360 days, suggests that cattle remained unprotected for a long period before the recommended booster vaccination.


Asunto(s)
Toxinas Bacterianas/inmunología , Toxinas Botulínicas/inmunología , Inmunidad Humoral , Toxoides/inmunología , Animales , Antitoxinas/sangre , Bovinos , Perros , Células de Riñón Canino Madin Darby , Ratones , Pruebas de Neutralización , Factores de Tiempo , Toxoides/administración & dosificación
10.
FEMS Microbiol Lett ; 366(4)2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30772899

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) producing type Ib heat-stable toxin (STa) are a main cause of children's diarrhea and travelers' diarrhea, thus STa needs to be targeted in ETEC vaccine development. However, because this 19-amino acid STa is poorly immunogenic, attempts to genetically fuse or chemically couple it to carrier proteins have been made to enhance STa immunogenicity. In this study, we selected one genetic fusion and one chemical conjugate to comparatively evaluate STa immunogenicity. The genetic fusion is 3xSTaN12S-mnLTR192G/L211A carrying three toxoid (STaN12S) genetically fused to a double mutant LT monomer (mnLTR192G/L211A); the chemical conjugate is BSA-STaA14T, which has toxoid STaA14T chemically coupled to bovine serum albumin (BSA). We immunized mice with the STa toxoid fusion and chemical conjugates, and examined antibody responses. Furthermore, we immunized pigs and evaluated derived antibodies for efficacy to passively provide protection against ETEC diarrhea using a piglet model. Data showed that mice subcutaneously immunized with BSA-STaA14T or 3xSTaN12S-mnLTR192G/L211A developed a strong anti-STa antibody, and the induced antibodies exhibited equivalent toxin-neutralizing activities. Pigs immunized with 3xSTaN12S-mnLTR192G/L211A or BSA-STaA14T developed similar levels of anti-STa antibodies; piglets with passively acquired antibodies induced by the genetic fusion appeared better protected against STa + ETEC. Results from the current study indicate that the fusion and conjugate approaches are viable options for facilitating STa immunogenicity and developing ETEC vaccines.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Inmunogenicidad Vacunal , Toxoides/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Conjugación Genética/inmunología , Escherichia coli Enterotoxigénica/genética , Escherichia coli Enterotoxigénica/inmunología , Infecciones por Escherichia coli/prevención & control , Fusión Génica/inmunología , Ratones , Porcinos
11.
J Pediatric Infect Dis Soc ; 8(2): 105-114, 2019 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-29438562

RESUMEN

BACKGROUND: Reduced-antigen-content tetanus, diphtheria, and acellular pertussis (Tdap) vaccine is recommended in many countries for boosting immunity in adolescents and adults. Although immunity to these antigens wanes with time, currently available Tdap products are not labeled for repeat administration in the United States. METHODS: We performed an observer-blinded, randomized controlled trial in 1330 adults aged 18 to <65 years who received either the Tdap (n = 1002) or tetanus-diphtheria (Td) (n = 328) vaccine 8 to 12 years after a dose of Tdap vaccine administered previously. Solicited adverse events following immunization were documented for 7 days after vaccination, and serious adverse events and adverse events of medical significance were documented for 6 months after vaccination. Levels of antibodies against component vaccine antigens were measured before and 1 month after vaccination. RESULTS: A solicited adverse event was reported by 87.7% of Tdap and 88.0% of Td vaccine recipients. We found no significant differences in the rates of injection-site reactions, systemic reactions, or serious adverse events between the vaccine groups. A robust antibody response to each pertussis antigen in the Tdap-vaccinated group was found; postvaccination-to-prevaccination geometric mean antibody concentration ratios were 8:1 (pertussis toxoid), 5.9 (filamentous hemagglutinin), 6.4 (pertactin), and 5.2 (fimbriae 2 and 3). Postvaccination geometric mean concentrations of tetanus antibody (4.20 and 4.74 IU/mL, respectively) and diphtheria antibody (10.1 and 12.6 IU/mL, respectively) were similar in the Tdap and Td groups, and the rates of seroprotection against tetanus and diphtheria were >99% in both groups. CONCLUSIONS: A second dose of Tdap vaccine in adults approximately 10 years after a previous dose was well tolerated and immunogenic. These data might facilitate consideration of providing Tdap booster doses to adults.


Asunto(s)
Vacunas contra Difteria, Tétanos y Tos Ferina Acelular/administración & dosificación , Vacunas contra Difteria, Tétanos y Tos Ferina Acelular/efectos adversos , Vacunas contra Difteria, Tétanos y Tos Ferina Acelular/inmunología , Inmunización Secundaria/métodos , Adhesinas Bacterianas/inmunología , Adolescente , Adulto , Anticuerpos Antibacterianos/sangre , Formación de Anticuerpos , Antígenos Bacterianos , Proteínas de la Membrana Bacteriana Externa/inmunología , Canadá , Difteria/prevención & control , Femenino , Fimbrias Bacterianas/inmunología , Humanos , Inmunogenicidad Vacunal , Reacción en el Punto de Inyección/inmunología , Masculino , Persona de Mediana Edad , Tétanos/prevención & control , Factores de Tiempo , Toxoides/inmunología , Estados Unidos , Vacunación , Factores de Virulencia de Bordetella/inmunología , Adulto Joven
12.
Toxicon ; 152: 30-36, 2018 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-29981815

RESUMEN

Hemiscorpius lepturus (H. lepturus) is one of the most dangerous scorpions and the most medically important scorpion in Iran. The clinical signs of H. lepturus envenomation, including dermonecrosis, hematuria, renal failure and early death, are attributed to phospholipase D activity. This study was conducted to develop a novel recombinant phospholipase D1 (rPLD1) toxoid and investigate its immunogenicity and protective effects against the lethality of H. lepturus venom. The lethal protein recombinant phospholipase D1 was expressed from PLD H. lepturus venom gland. The rPLD1 toxin was converted into toxoid (the first toxoid of H. lepturus PLD) with a 0.25% concentration of formalin and stored for ten days at room temperature. In the toxicity test, the lethal activity of recombinant phospholipase D1 was fully inhibited. When it reached up to 3 times higher than the maximal effective concentration of the purified toxin (11.1 µg), rPLD1 toxoid was used. The sphingomyelinase activity was inhibited when up to 5.4 times of the LD100 of the purified toxin (20 µg), toxoid was used. It was then used to produce an antibody in BALB/c as an antigen and the mice were then challenged with rPLD1 toxin and the whole venom. The immunogenicity of rPLD1 toxoid was evaluated and the maximum titer of the raised antibodies was determined by ELISA assay. The optimum titer for anti-rPLD1 toxoid sera was obtained at the third intraperitoneal injection of rPLD1 toxoid, and a high titer was reached at the fourth injection in the mice. This toxoid increased the amount of antibodies and produced a protective antiserum against the whole venom of H. lepturus and rPLD1 toxin. The in-vivo test results showed that the mice were completely resistant against 200 times the LD100 of recombinant phospholipase D1 and the whole venom of H. lepturus. To conclude, rPLD1 can be used in toxoid form as an immunogen in the production of a new generation of neutralizing antibodies against the lethality and toxicity of H. lepturus whole venom.


Asunto(s)
Fosfolipasa D/inmunología , Venenos de Escorpión/enzimología , Toxoides/inmunología , Animales , Anticuerpos Neutralizantes , Escherichia coli/inmunología , Formaldehído , Irán , Ratones Endogámicos BALB C , Fosfolipasa D/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Venenos de Escorpión/inmunología , Escorpiones , Esfingomielina Fosfodiesterasa , Toxoides/aislamiento & purificación
13.
Infect Immun ; 86(10)2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30012638

RESUMEN

Bordetella pertussis is the primary causative agent of pertussis (whooping cough), which is a respiratory infection that leads to a violent cough and can be fatal in infants. There is a need to develop more effective vaccines because of the resurgence of cases of pertussis in the United States since the switch from the whole-cell pertussis vaccines (wP) to the acellular pertussis vaccines (aP; diphtheria-tetanus-acellular-pertussis vaccine/tetanus-diphtheria-pertussis vaccine). Adenylate cyclase toxin (ACT) is a major virulence factor of B. pertussis that is (i) required for establishment of infection, (ii) an effective immunogen, and (iii) a protective antigen. The C-terminal repeats-in-toxin domain (RTX) of ACT is sufficient to induce production of toxin-neutralizing antibodies. In this study, we characterized the effectiveness of vaccines containing the RTX antigen against experimental murine infection with B. pertussis RTX was not protective as a single-antigen vaccine against B. pertussis challenge, and adding RTX to 1/5 human dose of aP did not enhance protection. Since the doses of aP used in murine studies are not proportionate to mouse/human body masses, we titrated the aP from 1/20 to 1/160 of the human dose. Mice receiving 1/80 human aP dose had bacterial burden comparable to those of naive controls. Adding RTX antigen to the 1/80 aP base resulted in enhanced bacterial clearance. Inclusion of RTX induced production of antibodies recognizing RTX, enhanced production of anti-pertussis toxin, decreased secretion of proinflammatory cytokines, such as interleukin-6, and decreased recruitment of total macrophages in the lung. This study shows that adding RTX antigen to an appropriate dose of aP can enhance protection against B. pertussis challenge in mice.


Asunto(s)
Adenilil Ciclasas/inmunología , Bordetella pertussis/inmunología , Vacuna contra la Tos Ferina/inmunología , Toxoides/inmunología , Tos Ferina/inmunología , Adenilil Ciclasas/administración & dosificación , Adenilil Ciclasas/genética , Animales , Anticuerpos Antibacterianos/inmunología , Anticuerpos Neutralizantes/inmunología , Bordetella pertussis/genética , Evaluación Preclínica de Medicamentos , Humanos , Ratones , Vacuna contra la Tos Ferina/administración & dosificación , Vacuna contra la Tos Ferina/genética , Toxoides/administración & dosificación , Toxoides/genética , Tos Ferina/microbiología
14.
J Anim Physiol Anim Nutr (Berl) ; 102(5): 1167-1180, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29905984

RESUMEN

Vitamin E (vit E), an essential antioxidant for maintaining the stability of biological membranes and the function of the immune system, is considered to support adaptive immune responses and performance in cattle. The principal virulence factor of Shiga toxin (Stx)-producing Escherichia coli (STEC), the eponymous Stx, modulates cellular immune responses in cattle, the primary STEC reservoir. Active and passive immunization of calves with Shiga toxoids (rStxMUT ) was recently shown to reduce the STEC shedding. Here, we examined the influence of vit E on calves' serum α-tocopherol, performance, haematology, blood chemistry and its interaction with rStxMUT immunization. Data from calves having received passive (colostrum from immunized cows) and active (intramuscularly at 5th and 8th weeks of life) vaccination with rStxMUT (n = 24) were compared to unvaccinated controls (n = 24; fed with low anti-Stx colostrum, placebo injected). For each vaccination group, data were analysed according to the level of vit E supplementation offered by milk replacer (188 IU all-rac-α-tocopheryl acetate daily [VitEM ] vs. 354 IU [VitEH ]). An increase by 79% in daily vit E supplementation led to slightly higher serum α-tocopherol level and earlier concentrate intake at the beginning of the experiment without significant differences in live weight gain, haematology, blood chemistry parameters and peripheral CD4+ and CD8+ T-cell subpopulations. rStxMUT vaccination modulated the CD4+ /CD8+ ratio irrespective of vit E supplementation but decreased concentrate intake in VitEH in a time-dependent manner. Results of our study indicate that an increase in daily vit E supplementation vastly fails to exert effects on laboratory parameters and growth performance. However, observed interactive effects of vit E supply and vaccination on the regulation of feed intake deserves further attention.


Asunto(s)
Bovinos/sangre , Bovinos/crecimiento & desarrollo , Toxoides/inmunología , Vitamina E/farmacología , alfa-Tocoferol/sangre , Alimentación Animal , Animales , Vacunas Bacterianas/inmunología , Suplementos Dietéticos , Masculino , Vacunación/veterinaria
15.
Int J Nanomedicine ; 13: 3251-3261, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29910613

RESUMEN

BACKGROUND: Melittin, the main active peptide ingredient of bee venom, can cause severe cell membrane lysis due to its robust interaction with negatively charged phospholipids. So far, no effective anti-melittin vaccine has been developed to protect people from undesired melittin intoxication. METHODS: Herein, we prepared a polydiacetylene (PDA) nanoparticle with cell membrane-mimic surface to complex melittin, forming an anti-melittin vaccine (PDA-melittin). RESULTS: PDA nanoparticles could effectively combine with melittin and neutralize its toxicity. PDA-melittin nanocomplex is demonstrated to enhance melittin uptake by DCs and stimulate strong melittin-specific immunity. Mice immunized with PDA-melittin nanocomplex showed higher survival rate after exposion to melittin than untreated mice. CONCLUSION: The PDA-melittin nanocomplex can efficiently and safely generate a specific immunity against melittin to protect body from melittin intoxication, providing a new method with potential clinical application for the treatment of melittin intoxication.


Asunto(s)
Venenos de Abeja/química , Meliteno/inmunología , Nanopartículas/química , Vacunas/química , Vacunas/inmunología , Células 3T3 , Animales , Venenos de Abeja/toxicidad , Biomimética , Células Dendríticas , Femenino , Meliteno/toxicidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Polímero Poliacetilénico , Polímeros/química , Poliinos/química , Toxoides/inmunología , Vacunas/farmacología
16.
Vet Res ; 49(1): 28, 2018 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-29514704

RESUMEN

The principal virulence factor of Shiga toxin (Stx)-producing Escherichia coli (STEC), the eponymous Stx, modulates cellular immune responses in cattle, the primary STEC reservoir. We examined whether immunization with genetically inactivated recombinant Shiga toxoids (rStx1MUT/rStx2MUT) influences STEC shedding in a calf cohort. A group of 24 calves was passively (colostrum from immunized cows) and actively (intra-muscularly at 5th and 8th week) vaccinated. Twenty-four calves served as unvaccinated controls (fed with low anti-Stx colostrum, placebo injected). Each group was divided according to the vitamin E concentration they received by milk replacer (moderate and high supplemented). The effective transfer of Stx-neutralizing antibodies from dams to calves via colostrum was confirmed by Vero cell assay. Serum antibody titers in calves differed significantly between the vaccinated and the control group until the 16th week of life. Using the expression of activation marker CD25 on CD4+CD45RO+ cells and CD8αhiCD45RO+ cells as flow cytometry based read-out, cells from vaccinated animals responded more pronounced than those of control calves to lysates of STEC and E. coli strains isolated from the farm as well as to rStx2MUT in the 16th week. Summarized for the entire observation period, less fecal samples from vaccinated calves were stx1 and/or stx2 positive than samples from control animals when calves were fed a moderate amount of vitamin E. This study provides first evidence, that transfer to and induction in young calves of Stx-neutralizing antibodies by Shiga toxoid vaccination offers the opportunity to reduce the incidence of stx-positive fecal samples in a calf cohort.


Asunto(s)
Derrame de Bacterias/inmunología , Vacunas Bacterianas/inmunología , Enfermedades de los Bovinos/prevención & control , Inmunización Pasiva/veterinaria , Escherichia coli Shiga-Toxigénica/fisiología , Toxoides/inmunología , Vacunación/veterinaria , Alimentación Animal/análisis , Animales , Bovinos , Enfermedades de los Bovinos/inmunología , Estudios de Cohortes , Calostro/inmunología , Dieta/veterinaria , Suplementos Dietéticos/análisis , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/prevención & control , Infecciones por Escherichia coli/veterinaria , Inmunidad Materno-Adquirida/inmunología , Inyecciones Intramusculares/veterinaria , Masculino , Vacunas Sintéticas/administración & dosificación
17.
Probiotics Antimicrob Proteins ; 10(2): 251-257, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28401477

RESUMEN

The alpha-toxin is one of the virulence factors of Clostridium perfringens for gas gangrene in humans and animals or necrotic enteritis in poultry. The C-terminal domain of this toxin ( cpa 247-370 ) was synthesized and cloned into pT1NX vector to construct the pT1NX-alpha plasmid. This surface-expressing plasmid was electroporated into Lactobacillus casei ATCC 393, generating the recombinant L. casei strain expressing alpha-toxoid (LC-α strain). Expression of this modified alpha-toxoid was confirmed by SDS-PAGE, immunoblotting, and direct immunofluorescence microscopy. BALB/c mice, immunized orally by the recombinant LC-α strain, elicited mucosal and significantly humoral immune responses (p < 0.05) and developed a protection against 900 MLD/mL of the standard alpha-toxin. This study showed that this recombinant LC-α strain could be a promising vaccine candidate against gas gangrene and necrotic enteritis.


Asunto(s)
Vacunas Bacterianas/administración & dosificación , Clostridium perfringens/inmunología , Enteritis/prevención & control , Gangrena Gaseosa/prevención & control , Lacticaseibacillus casei/genética , Probióticos/administración & dosificación , Toxoides/administración & dosificación , Administración Oral , Animales , Anticuerpos Antibacterianos/inmunología , Vacunas Bacterianas/genética , Vacunas Bacterianas/inmunología , Clonación Molecular , Clostridium perfringens/genética , Enteritis/inmunología , Femenino , Gangrena Gaseosa/inmunología , Expresión Génica , Humanos , Inmunización , Lacticaseibacillus casei/inmunología , Ratones , Ratones Endogámicos BALB C , Toxoides/genética , Toxoides/inmunología
18.
Appl Environ Microbiol ; 84(2)2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29079628

RESUMEN

Heat-stable toxin (STa)-producing enterotoxigenic Escherichia coli (ETEC) strains are a top cause of moderate-to-severe diarrhea in children from developing countries and a common cause of travelers' diarrhea. Recent progress in using STa toxoids and toxoid fusions to induce neutralizing anti-STa antibodies has accelerated ETEC vaccine development. However, concern remains regarding whether the derived anti-STa antibodies cross-react with STa-like guanylin and uroguanylin, two guanylate cyclase C (GC-C) ligands regulating fluid and electrolyte transportation in human intestinal and renal epithelial cells. To further divert STa from guanylin and uroguanylin structurally and antigenically and to eliminate anti-STa antibody cross-reactivity with guanylin and uroguanylin, we mutated STa at the 9th (leucine), 12th (asparagine), and 14th (alanine) residues for the double and triple mutants STaL9A/N12S, STaL9A/A14H, STaN12S/A14T, and STaL9A/N12S/A14H We then fused each STa mutant (three copies) to a monomeric heat-labile toxin (LT) mutant (mnLTR192G/L211A) for the toxoid fusions 3×STaL9A/N12S-mnLTR192G/L211A, 3×STaL9A/A14H-mnLTR192G/L211A, 3×STaN12S/A14T-mnLTR192G/L211A, and 3×STaL9A/N12S/A14H-mnLTR192G/L211A; examined each fusion for anti-STa immunogenicity; and assessed the derived antibodies for in vitro neutralization activity against STa toxicity and for cross-reactivity with guanylin and uroguanylin. Mice subcutaneously immunized with each fusion protein developed anti-STa antibodies, and the antibodies derived from 3×STaN12S-mnLTR192G/L211A, 3×STaL9A/N12S-mnLTR192G/L211A, or 3×STaN12S/A14T-mnLTR192G/L211A prevented STa from the stimulation of intracellular cGMP in T-84 cells. Competitive enzyme-linked immunosorbent assays (ELISAs) showed that guanylin and uroguanylin hardly blocked the binding of anti-STa antibodies to the coated STa-ovalbumin conjugate. These results indicated that antibodies derived from 3×STaN12S-mnLTR192G/L211A, 3×STaL9A/N12S-mnLTR192G/L211A, or 3×STaN12S/A14T-mnLTR192G/L211A neutralized STa and had little cross-reactivity with guanylin and uroguanylin, suggesting that these toxoid fusions are suitable antigens for ETEC vaccines.IMPORTANCE Enterotoxigenic Escherichia coli (ETEC) strains are a leading cause of children's diarrhea and travelers' diarrhea. Currently, there is no licensed vaccine against ETEC diarrhea. One key challenge is to identify safe antigens to induce antibodies neutralizing the key STa without cross-reacting with guanylin and uroguanylin, two important ligands controlling homeostasis in human intestinal and renal epithelial cells. In this study, we generated nontoxic fusion antigens that induced antibodies that neutralize STa enterotoxicity in vitro and do not cross-react with guanylin or uroguanylin. These fusions have become the preferred antigens for the development of ETEC vaccines to potentially prevent the deaths of hundreds of thousands of young children and hundreds of millions of diarrheal cases each year.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Escherichia coli Enterotoxigénica/inmunología , Hormonas Gastrointestinales/inmunología , Péptidos Natriuréticos/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Niño , Reacciones Cruzadas , Escherichia coli Enterotoxigénica/genética , Enterotoxinas/genética , Enterotoxinas/inmunología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/prevención & control , Femenino , Calor , Humanos , Inmunización , Ratones , Mutación , Toxoides/inmunología
19.
Infect Immun ; 86(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29263112

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) strains are a leading cause of children's diarrhea and travelers' diarrhea. Vaccines inducing antibodies to broadly inhibit bacterial adherence and to neutralize toxin enterotoxicity are expected to be effective against ETEC-associated diarrhea. 6×His-tagged adhesin-toxoid fusion proteins were shown to induce neutralizing antibodies to several adhesins and LT and STa toxins (X. Ruan, D. A. Sack, W. Zhang, PLoS One 10:e0121623, 2015, https://doi.org/10.1371/journal.pone.0121623). However, antibodies derived from His-tagged CFA/I/II/IV-2xSTaA14Q-dmLT or CFA/I/II/IV-2xSTaN12S-dmLT protein were less effective in neutralizing STa enterotoxicity and were not evaluated in vivo for efficacy against ETEC diarrhea. Additionally, His-tagged proteins are considered less desirable for human vaccines. In this study, we produced a tagless adhesin-toxoid MEFA (multiepitope fusion antigen) protein, enhanced anti-STa immunogenicity by including a third copy of STa toxoid STaN12S, and examined antigen immunogenicity in a murine model. Moreover, we immunized pregnant pigs with the tagless adhesin-toxoid MEFA protein and evaluated passive antibody protection against STa+ or LT+ ETEC infection in a pig challenge model. Results showed that tagless adhesin-toxoid MEFA CFA/I/II/IV-3xSTaN12S-mnLTR192G/L211A induced broad antiadhesin and antitoxin antibody responses in the intraperitoneally immunized mice and the intramuscularly immunized pigs. Mouse and pig serum antibodies significantly inhibited adherence of seven colonization factor antigen (CFA) adhesins (CFA/I and CS1 to CS6) and effectively neutralized both toxins. More importantly, suckling piglets born to the immunized mothers acquired antibodies and were protected against STa+ ETEC and LT+ ETEC diarrhea. These results indicated that tagless CFA/I/II/IV-3xSTaN12S-mnLTR192G/L211A induced broadly protective antiadhesin and antitoxin antibodies and demonstrate that this adhesin-toxoid MEFA is a potential antigen for developing broadly protective ETEC vaccines.


Asunto(s)
Adhesinas Bacterianas/administración & dosificación , Diarrea/prevención & control , Escherichia coli Enterotoxigénica/inmunología , Infecciones por Escherichia coli/prevención & control , Proteínas de Escherichia coli/administración & dosificación , Vacunas contra Escherichia coli/administración & dosificación , Toxoides/administración & dosificación , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Anticuerpos Neutralizantes/inmunología , Antígenos de Superficie/administración & dosificación , Antígenos de Superficie/genética , Antígenos de Superficie/inmunología , Adhesión Bacteriana/efectos de los fármacos , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Diarrea/inmunología , Diarrea/microbiología , Escherichia coli Enterotoxigénica/genética , Escherichia coli Enterotoxigénica/fisiología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/inmunología , Vacunas contra Escherichia coli/genética , Vacunas contra Escherichia coli/inmunología , Femenino , Proteínas Fimbrias/administración & dosificación , Proteínas Fimbrias/genética , Proteínas Fimbrias/inmunología , Ratones , Ratones Endogámicos BALB C , Porcinos , Toxoides/genética , Toxoides/inmunología
20.
Bioconjug Chem ; 29(3): 604-612, 2018 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-29241006

RESUMEN

As nanoparticles exhibit unique properties attractive for vaccine development, they have been progressively implemented as antigen delivery platforms and immune potentiators. Recently, cell membrane-coated nanoparticles have provided a novel approach for intercepting and neutralizing bacterial toxins by leveraging their natural affinity to cellular membranes. Such toxin-nanoparticle assemblies, termed nanotoxoids, allow rapid loading of different types of toxins and have been investigated for their ability to effectively confer protection against bacterial infection. This topical review will cover the current progress in antibacterial vaccine nanoformulations and highlight the nanotoxoid platform as a novel class of nanoparticulate vaccine. We aim to provide insights into the potential of nanotoxoids as a platform that is facile to implement and can be broadly applied to help address the rising threat of super pathogens.


Asunto(s)
Bacterias/inmunología , Infecciones Bacterianas/prevención & control , Toxinas Bacterianas/administración & dosificación , Vacunas Bacterianas/administración & dosificación , Membrana Celular/química , Nanopartículas/química , Toxoides/administración & dosificación , Animales , Infecciones Bacterianas/inmunología , Toxinas Bacterianas/química , Toxinas Bacterianas/inmunología , Vacunas Bacterianas/química , Vacunas Bacterianas/inmunología , Humanos , Nanotecnología/métodos , Toxoides/química , Toxoides/inmunología , Vacunación/métodos , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/química , Vacunas de Subunidad/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA