Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
2.
Avian Dis ; 65(2): 295-302, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34412461

RESUMEN

Several serotypes of non-host-specific or paratyphoid Salmonella have been linked with contamination of poultry meat, and eggs, resulting in foodborne outbreaks in humans. Vaccination of poultry against paratyphoid Salmonella is a frequent strategy used to reduce the levels of infection and transmission, which ultimately can lead to lower rates of human infections. Live vaccines have been developed and used in poultry immediately after hatching as a result of their ability to colonize the gut, stimulate a mucosal immune response, induce a competitive inhibitory effect against homologous wild strains, and reduce colonization and excretion of Salmonella. Furthermore, vaccines can competitively exclude some heterologous strains of Salmonella from colonizing the gastrointestinal tract when young poultry are immunologically immature. In addition, various studies have suggested that booster vaccination with live vaccines a few weeks after initial vaccination is essential to increase the level of protection and achieve better cross-protective immunity. Vaccination of breeders, broilers, layers, and turkeys with modified live Salmonella vaccines is a common intervention that has become an important component in poultry companies' multistep prevention programs to meet increasingly demanding customer and regulatory food safety requirements. Both live and inactivated vaccines play a critical role in a comprehensive control program for chicken and turkey breeders and commercial layers. This review examines the response and protection conferred by live modified vaccines against non-host-specific Salmonella that can be considered for the design and implementation of vaccination strategies in poultry.


Artículo regular­Inmunidad y protección que brindan las vacunas vivas modificadas contra salmonelas paratíficas en la avicultura­Una perspectiva aplicada. Varios serotipos de Salmonella paratífica no específica de huésped se han relacionado con la contaminación de la carne de pollo y huevos lo que ha provocado brotes de origen alimentario en los seres humanos. La vacunación de aves comerciales contra Salmonella paratífica es una estrategia que se utiliza con frecuencia para reducir los niveles de infección y transmisión, que en última instancia puede conducir a tasas más bajas de infecciones en humanos. Se han desarrollado y utilizado vacunas vivas en aves comerciales inmediatamente después de la eclosión como resultado de su capacidad para colonizar el intestino, estimular una respuesta inmunitaria de la mucosa, inducir un efecto inhibidor competitivo contra cepas silvestres homólogas y reducir la colonización y excreción de Salmonella. Además, las vacunas pueden excluir competitivamente algunas cepas heterólogas de Salmonella de colonizar el tracto gastrointestinal cuando las aves jóvenes son inmunológicamente inmaduras. Además, varios estudios han sugerido que la vacunación de refuerzo con vacunas vivas unas semanas después de la vacunación inicial es esencial para aumentar el nivel de protección y lograr una mejor inmunidad de protección cruzada. La vacunación de reproductoras, pollos de engorde, aves de postura y pavos con vacunas vivas modificadas contra Salmonella es una intervención común que se ha convertido en un componente importante en los programas de prevención de múltiples pasos de las empresas avícolas para cumplir con los requisitos de los clientes y regulatorios de seguridad alimentaria. Tanto las vacunas vivas como las inactivadas desempeñan un papel fundamental en un programa de control integral para productores de pollos, pavos y aves ponedoras comerciales. Esta revisión examina la respuesta y la protección conferidas por las vacunas vivas modificadas contra Salmonella no específica del huésped que pueden ser consideradas para el diseño e implementación de estrategias de vacunación en la avicultura.


Asunto(s)
Fiebre Paratifoidea/veterinaria , Enfermedades de las Aves de Corral/prevención & control , Salmonella enterica/inmunología , Vacunas Tifoides-Paratifoides , Animales , Pollos , Humanos , Inmunización Secundaria/veterinaria , Fiebre Paratifoidea/inmunología , Fiebre Paratifoidea/prevención & control , Aves de Corral , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/microbiología , Pavos , Vacunas Tifoides-Paratifoides/inmunología , Vacunas Tifoides-Paratifoides/normas , Vacunas Atenuadas/genética , Vacunas de Productos Inactivados/genética
3.
Pharmacol Res ; 169: 105652, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33975015

RESUMEN

Typhoid vaccine development has been impeded by inability of currently available vaccines to induce cellular immunity along with neutralizing antibodies against all serovars of S. Typhi and S. Paratyphi. Unfortunately, antibiotic treatment has shown to be an ineffective therapy due to development of resistance against multiple antibiotics. In the present study, we have explored the immunogenicity and protective efficacy of in-silico designed multi-epitope DnaK peptides as candidate vaccine molecules against Salmonella. Immunization studies in mouse typhoid model revealed three of these peptides (DP1, DP5 and DP7) are highly efficacious, stimulating both humoral and cell mediated immunity along with long lasting antibody memory response. There was significant increase in antibody titers (IgG, IgG1, IgG2a, IgA and IgM), lymphocyte proliferative responses and cytokine levels. Immunized groups showed marked reduction in organ bacterial load, fecal shedding and pronounced protection (upto 80%) as compared to unimmunized controls after challenge with S. typhimurium. Our results demonstrate the huge potential of DnaK peptide vaccine candidates (DP1, DP5 and DP7) to accord protective immunity with significant increase in survivability against Salmonella infection in mice, thus commending these molecules as promising agents to tackle typhoid.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Salmonella typhi/inmunología , Fiebre Tifoidea/prevención & control , Vacunas Tifoides-Paratifoides/uso terapéutico , Animales , Anticuerpos Neutralizantes/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunidad Celular/inmunología , Interleucinas/sangre , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Fiebre Paratifoidea/inmunología , Fiebre Paratifoidea/prevención & control , Salmonella paratyphi A/inmunología , Vacunas Tifoides-Paratifoides/inmunología
4.
Infect Immun ; 89(4)2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33318138

RESUMEN

Typhoid and paratyphoid fevers have a high incidence worldwide and coexist in many geographical areas, especially in low-middle-income countries (LMIC) in South and Southeast Asia. There is extensive consensus on the urgent need for better and affordable vaccines against systemic Salmonella infections. Generalized modules for membrane antigens (GMMA), outer membrane exosomes shed by Salmonella bacteria genetically manipulated to increase blebbing, resemble the bacterial surface where protective antigens are displayed in their native environment. Here, we engineered S Paratyphi A using the pDC5-viaB plasmid to generate GMMA displaying the heterologous S Typhi Vi antigen together with the homologous O:2 O antigen. The presence of both Vi and O:2 was confirmed by flow cytometry on bacterial cells, and their amount was quantified on the resulting vesicles through a panel of analytical methods. When tested in mice, such GMMA induced a strong antibody response against both Vi and O:2, and these antibodies were functional in a serum bactericidal assay. Our approach yielded a bivalent vaccine candidate able to induce immune responses against different Salmonella serovars, which could benefit LMIC residents and travelers.


Asunto(s)
Fiebre Paratifoidea/inmunología , Fiebre Paratifoidea/microbiología , Polisacáridos Bacterianos/inmunología , Polisacáridos Bacterianos/metabolismo , Salmonella paratyphi A/fisiología , Vesículas Transportadoras/metabolismo , Vacunas Combinadas/inmunología , Animales , Antígenos Bacterianos/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunización , Inmunogenicidad Vacunal/inmunología , Ratones , Antígenos O/inmunología , Fiebre Paratifoidea/prevención & control , Vacunas Combinadas/administración & dosificación
5.
PLoS Negl Trop Dis ; 14(10): e0008783, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33079959

RESUMEN

Enteric fever is a systemic infection caused by Salmonella Typhi or Paratyphi A. In many endemic areas, these serovars co-circulate and can cause multiple infection-episodes in childhood. Prior exposure is thought to confer partial, but incomplete, protection against subsequent attacks of enteric fever. Empirical data to support this hypothesis are limited, and there are few studies describing the occurrence of heterologous-protection between these closely related serovars. We performed a challenge-re-challenge study using a controlled human infection model (CHIM) to investigate the extent of infection-derived immunity to Salmonella Typhi or Paratyphi A infection. We recruited healthy volunteers into two groups: naïve volunteers with no prior exposure to Salmonella Typhi/Paratyphi A and volunteers previously-exposed to Salmonella Typhi or Paratyphi A in earlier CHIM studies. Within each group, participants were randomised 1:1 to oral challenge with either Salmonella Typhi (104 CFU) or Paratyphi A (103 CFU). The primary objective was to compare the attack rate between naïve and previously challenged individuals, defined as the proportion of participants per group meeting the diagnostic criteria of temperature of ≥38°C persisting for ≥12 hours and/or S. Typhi/Paratyphi bacteraemia up to day 14 post challenge. The attack-rate in participants who underwent homologous re-challenge with Salmonella Typhi was reduced compared with challenged naïve controls, although this reduction was not statistically significant (12/27[44%] vs. 12/19[63%]; Relative risk 0.70; 95% CI 0.41-1.21; p = 0.24). Homologous re-challenge with Salmonella Paratyphi A also resulted in a lower attack-rate than was seen in challenged naïve controls (3/12[25%] vs. 10/18[56%]; RR0.45; 95% CI 0.16-1.30; p = 0.14). Evidence of protection was supported by a post hoc analysis in which previous exposure was associated with an approximately 36% and 57% reduced risk of typhoid or paratyphoid disease respectively on re-challenge. Individuals who did not develop enteric fever on primary exposure were significantly more likely to be protected on re-challenge, compared with individuals who developed disease on primary exposure. Heterologous re-challenge with Salmonella Typhi or Salmonella Paratyphi A was not associated with a reduced attack rate following challenge. Within the context of the model, prior exposure was not associated with reduced disease severity, altered microbiological profile or boosting of humoral immune responses. We conclude that prior Salmonella Typhi and Paratyphi A exposure may confer partial but incomplete protection against subsequent infection, but with a comparable clinical and microbiological phenotype. There is no demonstrable cross-protection between these serovars, consistent with the co-circulation of Salmonella Typhi and Paratyphi A. Collectively, these data are consistent with surveillance and modelling studies that indicate multiple infections can occur in high transmission settings, supporting the need for vaccines to reduce the burden of disease in childhood and achieve disease control. Trial registration NCT02192008; clinicaltrials.gov.


Asunto(s)
Fiebre Paratifoidea/inmunología , Salmonella paratyphi A/fisiología , Salmonella typhi/fisiología , Fiebre Tifoidea/inmunología , Adolescente , Adulto , Protección Cruzada , Femenino , Humanos , Inmunidad Humoral , Masculino , Persona de Mediana Edad , Fiebre Paratifoidea/microbiología , Salmonella paratyphi A/inmunología , Salmonella typhi/inmunología , Fiebre Tifoidea/microbiología , Adulto Joven
7.
Microb Pathog ; 137: 103795, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31610219

RESUMEN

Paratyphoid fever caused by Salmonella Paratyphi A is a serious public health problem in many countries. In order to and develop a live attenuated candidate vaccine of Salmonella Paratyphi A, a Salmonella pathogenicity island 2 (SPI2, approximate 40 kb) deletion mutant of Salmonella Paratyphi A was constructed by lambda Red recombination, then the biological characteristics and protective ability of the Salmonella Paratyphi A SPI2 mutant were evaluated. Our results showed that the growth and biochemical properties of the SPI2 mutant were consistent with that of its parent strain, and the mutant was stable with the loss of SPI2. The mice lethal test showed that the virulence of the SPI2 mutant was significantly decreased, it can colonize and persistent more than 14 days in the liver and spleen of mice. Vaccination with the SPI2 mutant in mice revealed no significant effect on body weight and clinical symptoms compared to control animals, and specific humoral and cellular immune responses were also significantly induced. Immunization of mice offered efficient protection against Salmonella Paratyphi A strain challenge at 14 days post vaccination based on mortality and clinical symptoms relative to control group. Overall, these findings suggested that SPI2 plays an important role in pathogenicity of Salmonella Paratyphi A, and the SPI2 mutant showed its potential to develop a live attenuated vaccine candidate.


Asunto(s)
Islas Genómicas , Fiebre Paratifoidea/prevención & control , Vacunas contra la Salmonella/administración & dosificación , Salmonella paratyphi A/genética , Vacunas Tifoides-Paratifoides/administración & dosificación , Animales , Anticuerpos Antibacterianos/inmunología , Femenino , Humanos , Inmunización , Hígado/inmunología , Ratones , Ratones Endogámicos BALB C , Fiebre Paratifoidea/inmunología , Fiebre Paratifoidea/microbiología , Vacunas contra la Salmonella/genética , Vacunas contra la Salmonella/inmunología , Salmonella paratyphi A/inmunología , Salmonella paratyphi A/patogenicidad , Eliminación de Secuencia , Bazo/inmunología , Vacunas Tifoides-Paratifoides/genética , Vacunas Tifoides-Paratifoides/inmunología , Virulencia
8.
Immunobiology ; 224(3): 371-382, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30952553

RESUMEN

Enteric fever, caused by Salmonella enterica serovars, Typhi (S. Typhi) and Paratyphi (S. Paratyphi) is a major public health challenge for the developing nations. Globally, the disease affects ˜15-30 million individuals every year, resulting in >200,000 deaths. Multidrug-resistant S. Typhi H58 strain has emerged as the dominant circulating strain in a large part of the world and an extensively drug-resistant (XDR) subclade of the strain was recently reported. Many believe that vaccination of the susceptible populations is urgently needed and the best option to control the infection. However, the commercial live attenuated (Ty21a) vaccine is not recommended for children below six years of age while the Vi-polysaccharide-based vaccine has poor long-term efficacy against typhoid fever. Moreover, no vaccines are available against S. Paratyphi infection. Thus, a new formulation capable of providing long term protection against both the pathogens and safe for all age groups is immediately required. We show that recombinant, S. Typhi outer membrane protein STIV (rSTIV) is immunogenic in mice and elicits high serum titers of different immunoglobulin subtypes. STIV antibodies opsonize S. Typhi and S. Paratyphi A to promote antibody-dependent cellular cytotoxicity and complement-mediated lysis. Immunization with rSTIV also induces robust cell-mediated immunity, including antigen-specific T cell proliferation and cytotoxic T lymphocyte response. Finally, mice immunized with rSTIV are significantly protected against S. Typhi and S. Paratyphi A challenge, with reduced visceral bacterial load. Our results underscore the potential of rSTIV as a novel vaccine candidate for enteric fever.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Linfocitos T CD8-positivos/inmunología , Fiebre Paratifoidea/inmunología , Vacunas contra la Salmonella/inmunología , Salmonella typhi/fisiología , Fiebre Tifoidea/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Citotoxicidad Celular Dependiente de Anticuerpos , Proteínas de la Membrana Bacteriana Externa/genética , Línea Celular Tumoral , Proliferación Celular , Citotoxicidad Inmunológica , Humanos , Inmunidad Humoral , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/genética
9.
Nat Commun ; 9(1): 253, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29343684

RESUMEN

Mucosal-associated invariant T (MAIT) cells are innate-like T cells that can detect bacteria-derived metabolites presented on MR1. Here we show, using a controlled infection of humans with live Salmonella enterica serovar Paratyphi A, that MAIT cells are activated during infection, an effect maintained even after antibiotic treatment. At the peak of infection MAIT cell T-cell receptor (TCR)ß clonotypes that are over-represented prior to infection transiently contract. Select MAIT cell TCRß clonotypes that expand after infection have stronger TCR-dependent activation than do contracted clonotypes. Our results demonstrate that host exposure to antigen may drive clonal expansion of MAIT cells with increased functional avidity, suggesting a role for specific vaccination strategies to increase the frequency and potency of MAIT cells to optimize effector function.


Asunto(s)
Proliferación Celular , Células T Invariantes Asociadas a Mucosa/inmunología , Fiebre Paratifoidea/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Salmonella paratyphi A/inmunología , Adolescente , Adulto , Línea Celular Tumoral , Células Clonales/inmunología , Células Clonales/metabolismo , Células Clonales/microbiología , Voluntarios Sanos , Interacciones Huésped-Patógeno/inmunología , Humanos , Células Jurkat , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/microbiología , Persona de Mediana Edad , Células T Invariantes Asociadas a Mucosa/metabolismo , Células T Invariantes Asociadas a Mucosa/microbiología , Fiebre Paratifoidea/metabolismo , Fiebre Paratifoidea/microbiología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Salmonella paratyphi A/fisiología , Adulto Joven
10.
Expert Rev Vaccines ; 16(10): 1029-1043, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28856924

RESUMEN

INTRODUCTION: Typhoid and paratyphoid fever remain a global health problem, which - in non-endemic countries - are mainly seen in travelers, particularly in VFRs (visiting friends and relatives), with occasional local outbreaks occurring. A rise in anti-microbial resistance emphasizes the role of preventive measures, especially vaccinations against typhoid and paratyphoid fever for travelers visiting endemic countries. Areas covered: This state-of-the-art review recapitulates the epidemiology and mechanisms of disease of typhoid and paratyphoid fever, depicts the perspective of non-endemic countries and travelers (VFRs), and collectively presents current European recommendations for typhoid fever vaccination. We provide a brief overview of available (and developmental) vaccines in Europe, present current data on cross-protection to S. Paratyphi, and aim to provide a background for typhoid vaccine decision-making in travelers. Expert commentary: European recommendations are not harmonized. Experts must assess vaccination of travelers based on current country-specific recommendations. Travel health practitioners should be aware of the issues surrounding vaccination of travelers and be motivated to increase awareness of typhoid and paratyphoid fever risks.


Asunto(s)
Protección Cruzada , Fiebre Paratifoidea/prevención & control , Fiebre Tifoidea/prevención & control , Vacunas Tifoides-Paratifoides/administración & dosificación , Vacunación , Cápsulas Bacterianas/química , Cápsulas Bacterianas/inmunología , Portador Sano , Toma de Decisiones Clínicas , Países en Desarrollo , Europa (Continente)/epidemiología , Humanos , Fiebre Paratifoidea/epidemiología , Fiebre Paratifoidea/inmunología , Fiebre Paratifoidea/microbiología , Polisacáridos Bacterianos/química , Polisacáridos Bacterianos/inmunología , Guías de Práctica Clínica como Asunto , Salmonella enterica/inmunología , Salmonella enterica/patogenicidad , Salmonella typhi/inmunología , Salmonella typhi/patogenicidad , Viaje , Fiebre Tifoidea/epidemiología , Fiebre Tifoidea/inmunología , Fiebre Tifoidea/microbiología , Vacunas Conjugadas
11.
Microbiol Immunol ; 59(8): 443-51, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26084199

RESUMEN

The global epidemic features of enteric fever have changed greatly in recent years. The incidence of enteric fever caused by Salmonella enterica serovar Paratyphi A has progressively increased. In some areas of Asia, infections with S. Paratyphi A have exceeded those with S. Typhi, resulting in S. Paratyphi A becoming the main causative agent of enteric fever. However, two currently licensed typhoid vaccines do not confer adequate cross-protection against S. Paratyphi A infection. Therefore, development of specific vaccines against enteric fever caused by S. Paratyphi A is urgently needed. In the present study, an attenuated strain was constructed by double deletion of the htrA and yncD genes in a wild-type strain of S. Paratyphi A and its safety and immunogenicity assessed. In a mouse model, the 50% lethal dose of the double deletion mutant and the wild-type strain were 3.0 × 10(8) CFU and 1.9 × 10(3) CFU, respectively, suggesting that the double deletion resulted in remarkably decreased bacterial virulence. Bacterial colonization of the double deletion mutant in the livers and spleens of infected mice was strikingly less than that of the wild-type strain. A single nasal administration of the attenuated vaccine candidate elicited high concentrations of anti-LPS and anti-flagellin IgG in a mouse model and protected immunized mice against lethal challenge with the wild-type strain. Thus, our findings suggest that the attenuated vaccine strain is a promising candidate worthy of further evaluation both as a human enteric fever vaccine and as a vaccine delivery vector for heterologous antigens.


Asunto(s)
Eliminación de Gen , Fiebre Paratifoidea/prevención & control , Salmonella paratyphi A/crecimiento & desarrollo , Salmonella paratyphi A/inmunología , Vacunas Tifoides-Paratifoides/inmunología , Factores de Virulencia/deficiencia , Administración Intranasal , Animales , Anticuerpos Antibacterianos/sangre , Carga Bacteriana , Modelos Animales de Enfermedad , Femenino , Flagelina/inmunología , Inmunoglobulina G/sangre , Dosificación Letal Mediana , Lipopolisacáridos/inmunología , Hígado/microbiología , Ratones Endogámicos BALB C , Fiebre Paratifoidea/inmunología , Fiebre Paratifoidea/microbiología , Salmonella paratyphi A/genética , Bazo/microbiología , Análisis de Supervivencia , Vacunas Tifoides-Paratifoides/administración & dosificación , Vacunas Tifoides-Paratifoides/genética , Vacunas Tifoides-Paratifoides/aislamiento & purificación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/aislamiento & purificación , Virulencia
12.
Mucosal Immunol ; 8(6): 1349-59, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25872480

RESUMEN

Previously we have extensively characterized Salmonella enterica serovar Typhi (S. Typhi)-specific cell-mediated immune (CMI) responses in volunteers orally immunized with the licensed Ty21a typhoid vaccine. In this study we measured Salmonella-specific multifunctional (MF) CD8+ T-cell responses to further investigate whether Ty21a elicits crossreactive CMI against S. Paratyphi A and S. Paratyphi B that also cause enteric fever. Ty21a-elicited crossreactive CMI responses against all three Salmonella serotypes were predominantly observed in CD8+ T effector/memory (T(EM)) and, to a lesser extent, in CD8+CD45RA+ T(EM) (T(EMRA)) subsets. These CD8+ T-cell responses were largely mediated by MF cells coproducing interferon-γ and macrophage inflammatory protein-1ß and expressing CD107a with or without tumor necrosis factor-α. Significant proportions of Salmonella-specific MF cells expressed the gut-homing molecule integrin α4ß7. In most subjects, similar MF responses were observed to S. Typhi and S. Paratyphi B, but not to S. Paratyphi A. These results suggest that Ty21a elicits MF CMI responses against Salmonella that could be critical in clearing the infection. Moreover, because S. Paratyphi A is a major public concern and Ty21a was shown in field studies not to afford cross-protection to S. Paratyphi A, these results will be important in developing a S. Typhi/S. Paratyphi A bivalent vaccine against enteric fevers.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Fiebre Paratifoidea/prevención & control , Polisacáridos Bacterianos/inmunología , Fiebre Tifoidea/prevención & control , Vacunas Tifoides-Paratifoides/inmunología , Adulto , Reacciones Cruzadas/inmunología , Femenino , Citometría de Flujo , Humanos , Masculino , Persona de Mediana Edad , Fiebre Paratifoidea/inmunología , Salmonella paratyphi A/inmunología , Salmonella paratyphi B/inmunología , Salmonella typhi/inmunología , Fiebre Tifoidea/inmunología , Adulto Joven
14.
Scand J Immunol ; 79(3): 222-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24383914

RESUMEN

There are no vaccines in clinical use against paratyphoid fever, caused by Salmonella Paratyphi A and B or, rarely, C. Oral Salmonella Typhi Ty21a typhoid vaccine elicits a significant cross-reactive immune response against S. Paratyphi A and B, and some reports suggest cross-protective efficacy against the disease. These findings are ascribed to the O-12 antigen shared between the strains. The Vi capsular polysaccharide vaccine has been shown to elicit antibodies reactive with O-9,12. Twenty-five volunteers immunized with the parenteral Vi vaccine (Typherix(®) ) were explored for plasmablasts cross-reactive with paratyphoid strains; the responses were compared to those in 25 age- and gender-matched volunteers immunized with Ty21a (Vivotif(®) ). Before vaccination, 48/50 vaccinees had no plasmablasts reactive with the antigens. Seven days after vaccination, 15/25 and 22/25 Vi- and Ty21a-vaccinated volunteers had circulating plasmablasts producing antibodies cross-reactive with S. Paratyphi A, 18/25 and 23/25 with S. Paratyphi B and 16/25 and 9/25 with Paratyphi C, respectively. Compared to the Ty21a group, the Vi group showed significantly lower responses to S. Paratyphi A and B and higher to S. Paratyphi C. To conclude, the Vi vaccine elicited a cross-reactive plasmablast response to S. Paratyphi C (Vi antigen in common) and less marked responses to S. Paratyphi A and B than the Ty21a preparation. S. Paratyphi A and B both being Vi-negative, the result can be explained by trace amounts of bacterial cell wall O-12 antigen in the Vi preparation, despite purification. The clinical significance of this finding remains to be determined.


Asunto(s)
Polisacáridos Bacterianos/inmunología , Salmonella paratyphi A/inmunología , Salmonella paratyphi B/inmunología , Salmonella paratyphi C/inmunología , Vacunas Tifoides-Paratifoides/inmunología , Adulto , Anticuerpos Antibacterianos/inmunología , Reacciones Cruzadas/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Antígenos O/inmunología , Fiebre Paratifoidea/inmunología , Fiebre Paratifoidea/prevención & control , Vacunación , Adulto Joven
15.
Clin Vaccine Immunol ; 20(5): 712-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23486419

RESUMEN

Salmonella enterica serotype Paratyphi A is a human-restricted pathogen and the cause of paratyphoid A fever. Using a high-throughput immunoscreening technique, in vivo-induced antigen technology (IVIAT), we identified 20 immunogenic bacterial proteins expressed in humans who were bacteremic with S. Paratyphi A but not those expressed in S. Paratyphi A grown under standard laboratory conditions. The majority of these proteins have known or potential roles in the pathogenesis of S. enterica. These include proteins implicated in cell adhesion, fimbrial structure, adaptation to atypical conditions, oxidoreductase activity, proteolysis, antimicrobial resistance, and ion transport. Of particular interest among these in vivo-expressed proteins were S. Paratyphi A (SPA)2397, SPA2612, and SPA1604. SPA2397 and SPA2612 are prophage related, and SPA1604 is in Salmonella pathogenicity island 11 (SPI-11). Using real-time quantitative PCR (RT-qPCR), we confirmed increased levels of mRNA expressed by genes identified by IVIAT in a comparison of mRNA levels in organisms in the blood of bacteremic patients to those in in vitro cultures. Comparing convalescent- to acute-phase samples, we also detected a significant increase in the reaction of convalescent-phase antibodies with two proteins identified by IVIAT: SPA2397 and SPA0489. SPA2397 is a phage-related lysozyme, Gp19, and SPA0489 encodes a protein containing NlpC/P60 and cysteine, histidine-dependent amidohydrolase/peptidase (CHAP) domains. In a previous study utilizing a different approach, we found that transcripts for 11 and 7 of the genes identified by IVIAT were detectable in organisms in the blood of humans in Bangladesh who were bacteremic with S. Paratyphi A and Salmonella enterica serovar Typhi, respectively. S. Paratyphi A antigens identified by IVIAT warrant further evaluation for their contributions to pathogenesis and might have diagnostic, therapeutic, or preventive relevance.


Asunto(s)
Antígenos Bacterianos/sangre , Bacteriemia/microbiología , Proteínas Bacterianas/sangre , Proteínas Bacterianas/inmunología , Fiebre Paratifoidea/microbiología , Salmonella paratyphi A/inmunología , Antígenos Bacterianos/biosíntesis , Antígenos Bacterianos/inmunología , Bacteriemia/inmunología , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/aislamiento & purificación , Humanos , Fiebre Paratifoidea/diagnóstico , Fiebre Paratifoidea/inmunología , Profagos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Salmonella paratyphi A/genética , Salmonella paratyphi A/virología
16.
Microbes Infect ; 15(4): 270-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23376166

RESUMEN

Growing evidences suggest that Saccharomyces boulardii (SB) is efficacious against bacterial infections and inflammatory bowel diseases. This study investigated the effects of treatment with SB provided in a murine model of typhoid fever. Mice were divided into two groups: (1) control animals challenged with Salmonella Typhimurium (ST), and (2) animals receiving SB, and then challenged with ST. At days 0, 1, 5, 10 and 15 post-challenge, animals were euthanized and tissues collected to analyze bacterial translocation, cytokines, signaling pathways and histological analysis. Survival rate and animal weight were also evaluated. Treatment with SB increased survival rate and inhibited translocation of bacteria after ST challenge. Histological data showed that SB also protected mice against liver damage induced by ST. SB decreased levels of inflammatory cytokines and activation of mitogen-activated protein kinases (p38, JNK and ERK1/2), phospho-IκB, p65-RelA, phospho-jun and c-fos in the colon, signal pathways involved in the activation of inflammation induced by ST. Further experiments revealed that probiotic effects were due, at least in part, to the binding of ST to the yeast. Such binding diminishes ST translocation, resulting in decreased activation of signaling pathways which lead to intestinal inflammation in a murine model of typhoid fever.


Asunto(s)
Traslocación Bacteriana/inmunología , Fiebre Paratifoidea/inmunología , Saccharomyces/inmunología , Salmonella typhimurium/inmunología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Histocitoquímica , Hígado/inmunología , Hígado/microbiología , Hígado/patología , Ratones , Análisis de Supervivencia
17.
PLoS One ; 7(11): e47039, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23144798

RESUMEN

Enteric fevers remain a common and serious disease, affecting mainly children and adolescents in developing countries. Salmonella enterica serovar Typhi was believed to cause most enteric fever episodes, but several recent reports have shown an increasing incidence of S. Paratyphi A, encouraging the development of a bivalent vaccine to protect against both serovars, especially considering that at present there is no vaccine against S. Paratyphi A. The O-specific polysaccharide (O:2) of S. Paratyphi A is a protective antigen and clinical data have previously demonstrated the potential of using O:2 conjugate vaccines. Here we describe a new conjugation chemistry to link O:2 and the carrier protein CRM(197), using the terminus 3-deoxy-D-manno-octulosonic acid (KDO), thus leaving the O:2 chain unmodified. The new conjugates were tested in mice and compared with other O:2-antigen conjugates, synthesized adopting previously described methods that use CRM(197) as carrier protein. The newly developed conjugation chemistry yielded immunogenic conjugates with strong serum bactericidal activity against S. Paratyphi A.


Asunto(s)
Proteínas Bacterianas/química , Antígenos O/química , Fiebre Paratifoidea/prevención & control , Salmonella paratyphi A/inmunología , Vacunas Tifoides-Paratifoides/química , Vacunas Conjugadas/química , Animales , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/uso terapéutico , Secuencia de Carbohidratos , Femenino , Humanos , Ratones , Datos de Secuencia Molecular , Antígenos O/inmunología , Antígenos O/uso terapéutico , Fiebre Paratifoidea/sangre , Fiebre Paratifoidea/inmunología , Fiebre Paratifoidea/veterinaria , Salmonella paratyphi A/química , Vacunas Tifoides-Paratifoides/inmunología , Vacunas Tifoides-Paratifoides/uso terapéutico , Vacunas Conjugadas/inmunología , Vacunas Conjugadas/uso terapéutico
18.
Vaccine ; 30(42): 6047-53, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22858557

RESUMEN

BACKGROUND: There are no vaccines against paratyphoid fever in clinical use. The disease has become more wide-spread and there is a growing problem of antibiotic resistance among the strains. Previous reports suggest that the oral live Salmonella Typhi Ty21a-vaccine confers protection against paratyphoid B fever. Data on efficacy against paratyphoid A fever are somewhat contentious. The present study investigated the immunological basis for such efficacy reports at a single-cell level: plasmablasts (identified as antibody-secreting cells, ASC) were studied for secretion of antibodies cross-reactive with Salmonella Paratyphi in the circulation of patients with enteric fever and of volunteers vaccinated with Ty21a. MATERIALS AND METHODS: Thirty volunteers immunized with Ty21a and five patients with enteric fever were investigated for Salmonella Typhi and Salmonella Paratyphi A/B/C-specific circulating plasmablasts. PBMC were sorted by their expression of homing receptors (HR) for the intestine (α4ß7), peripheral lymph node (l-selectin) and skin (CLA) and typhoid- and paratyphoid-specific plasmablasts were enumerated with ELISPOT. RESULTS: Before vaccination, no cross-reactive ASC were found in the volunteers. In addition to the Salmonella Typhi-specific response, a significant cross-reactive immune response was mounted against Salmonella Paratyphi A and B both in the patients and the vaccinees. The magnitude of the response increased in the order Salmonella Paratyphi A (median 30 ASC/10(6) PBMC)→Salmonella Paratyphi B (median 81)→Salmonella Typhi (median 301) in the vaccinees. Both in patients and in vaccinees, the homing receptor (HR) selection favored homing to the gut, indicating a humoral intestinal immune response. CONCLUSIONS: These immunological data provide evidence consistent with previous reports describing certain levels of cross-protective efficacy of Ty21a against paratyphoid fever. Controlled studies are needed to evaluate cross-protective efficacy. In the current situation where paratyphoid fever is emerging and no vaccines are available, any level of cross-protective capacity is valuable.


Asunto(s)
Células Productoras de Anticuerpos/inmunología , Inmunidad Mucosa , Fiebre Paratifoidea/prevención & control , Polisacáridos Bacterianos/uso terapéutico , Receptores Mensajeros de Linfocitos/inmunología , Salmonella paratyphi A/patogenicidad , Vacunas Tifoides-Paratifoides/uso terapéutico , Administración Oral , Adolescente , Adulto , Protección Cruzada/inmunología , Reacciones Cruzadas/inmunología , Femenino , Humanos , Integrinas/metabolismo , Intestinos/inmunología , Selectina L/metabolismo , Ganglios Linfáticos/inmunología , Masculino , Persona de Mediana Edad , Fiebre Paratifoidea/inmunología , Polisacáridos Bacterianos/inmunología , Salmonella paratyphi A/inmunología , Piel/inmunología , Fiebre Tifoidea/inmunología , Fiebre Tifoidea/prevención & control , Vacunas Tifoides-Paratifoides/inmunología , Adulto Joven
19.
Clin Vaccine Immunol ; 19(2): 146-56, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22190395

RESUMEN

Enteric fever is an invasive life-threatening systemic disease caused by the Salmonella enterica human-adapted serovars Typhi and Paratyphi. Increasing incidence of infections with Salmonella enterica serovar Paratyphi A and the spreading of its antibiotic-resistant derivates pose a significant health concern in some areas of the world. Herein, we describe a molecular and phenotypic characterization of an S. Paratyphi A strain accounted for a recent paratyphoid outbreak in Nepal that affected at least 37 travelers. Pulsed-field gel electrophoresis analysis of the outbreak isolates revealed one genetic clone (pulsotype), confirming a single infecting source. Genetic profiling of the outbreak strain demonstrated the contribution of specific bacteriophages as a prime source of genetic diversity among clinical isolates of S. Paratyphi A. Phenotypic characterization in comparison with the S. Paratyphi A ATCC 9150 reference sequenced strain showed differences in flagellar morphology and increased abilities of the outbreak strain with respect to its motility, invasion into nonphagocytic cells, intracellular multiplication, survival within macrophages, and higher induction of interleukin-8 (IL-8) secreted by host cells. Collectively, these differences suggest an enhanced virulence potential of this strain and demonstrate an interesting phenotypic variation among S. Paratyphi A isolates. In vivo profiling of 16 inflammatory cytokines in patients infected with the outbreak strain revealed a common profile of a remarkable gamma interferon (IFN-γ) induction together with elevated concentrations of tumor necrosis factor alpha (TNF-α), IL-6, IL-8, IL-10, and IL-15, but not IL-12, which was previously demonstrated as elevated in nontyphoidal Salmonella infections. This apparent profile implies a distinct immune response to paratyphoid infections.


Asunto(s)
Fiebre Paratifoidea/inmunología , Fiebre Paratifoidea/microbiología , Salmonella paratyphi A/genética , Salmonella paratyphi A/inmunología , Adulto , Anciano , Animales , Bacteriófagos/genética , Células CACO-2 , Citocinas/análisis , ADN Bacteriano/genética , Farmacorresistencia Bacteriana , Femenino , Variación Genética , Genotipo , Células HeLa , Humanos , Interleucina-8/metabolismo , Israel , Macrófagos/microbiología , Masculino , Ratones , Pruebas de Sensibilidad Microbiana , Persona de Mediana Edad , Nepal/epidemiología , Fiebre Paratifoidea/epidemiología , Fenotipo , Salmonella paratyphi A/efectos de los fármacos , Salmonella paratyphi A/aislamiento & purificación , Análisis de Secuencia de ADN , Adulto Joven
20.
PLoS Negl Trop Dis ; 5(11): e1373, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22069504

RESUMEN

BACKGROUND: Antibiotic-resistant Salmonella enterica serovar Paratyphi A, the agent of paratyphoid A fever, poses an emerging public health dilemma in endemic areas of Asia and among travelers, as there is no licensed vaccine. Integral to our efforts to develop a S. Paratyphi A vaccine, we addressed the role of flagella as a potential protective antigen by comparing cell-associated flagella with exported flagellin subunits expressed by attenuated strains. METHODOLOGY: S. Paratyphi A strain ATCC 9150 was first deleted for the chromosomal guaBA locus, creating CVD 1901. Further chromosomal deletions in fliD (CVD 1901D) or flgK (CVD 1901K) were then engineered, resulting in the export of unpolymerized FliC, without impairing its overall expression. The virulence of the resulting isogenic strains was examined using a novel mouse LD(50) model to accommodate the human-host restricted S. Paratyphi A. The immunogenicity of the attenuated strains was then tested using a mouse intranasal model, followed by intraperitoneal challenge with wildtype ATCC 9150. RESULTS: Mucosal (intranasal) immunization of mice with strain CVD 1901 expressing cell-associated flagella conferred superior protection (vaccine efficacy [VE], 90%) against a lethal intraperitoneal challenge, compared with the flagellin monomer-exporting mutants CVD 1901K (30% VE) or CVD 1901D (47% VE). The superior protection induced by CVD 1901 with its cell-attached flagella was associated with an increased IgG2a:IgG1 ratio of FliC-specific antibodies with enhanced opsonophagocytic capacity. CONCLUSIONS: Our results clearly suggest that enhanced anti-FliC antibody-mediated clearance of S. Paratyphi A by phagocytic cells, induced by vaccines expressing cell-associated rather than exported FliC, might be contributing to the vaccine-induced protection from S. Paratyphi A challenge in vivo. We speculate that an excess of IgG1 anti-FliC antibodies induced by the exported FliC may compete with the IgG2a subtype and block binding to specific phagocyte Fc receptors that are critical for clearing an S. Paratyphi A infection.


Asunto(s)
Flagelos/inmunología , Fiebre Paratifoidea/prevención & control , Vacunas contra la Salmonella/inmunología , Salmonella paratyphi A/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Modelos Animales de Enfermedad , Femenino , Flagelos/genética , Eliminación de Gen , Inmunoglobulina G/sangre , Dosificación Letal Mediana , Ratones , Ratones Endogámicos BALB C , Proteínas Opsoninas/sangre , Fiebre Paratifoidea/inmunología , Fagocitosis , Vacunas contra la Salmonella/administración & dosificación , Vacunas contra la Salmonella/genética , Salmonella paratyphi A/genética , Análisis de Supervivencia , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...