Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Eur J Immunol ; 46(4): 874-84, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26681580

RESUMEN

Respiratory syncytial virus (RSV) causes severe bronchiolitis in infants worldwide. The immunological factors responsible for RSV susceptibility in infants are poorly understood. Here, we used the BALB/c mouse model of neonatal RSV infection to study the mechanisms leading to severe disease upon reexposure to the virus when adults. Two major deficiencies in neonatal lung innate responses were found: a poor DCs mobilization, and a weak engagement of the IFNI pathway. The administration of Flt3 ligand (Flt3-L), a growth factor that stimulates the proliferation of hematopoietic cells, to neonates before RSV-infection, resulted in increased lung DC number, and reconditioned the IFNI pathway upon RSV neonatal infection. Besides, neonates treated with Flt3-L were protected against exacerbated airway disease upon adult reexposure to RSV. This was associated with a reorientation of RSV-specific responses toward Th1-mediated immunity. Thus, the poor lung DCs and IFNI responses to RSV in neonates may be partly responsible for the deleterious long-term consequences revealed upon adult reexposure to RSV, which could be prevented by Flt3-L treatment. These results open new perspectives for developing neonatal immuno-modulating strategies to reduce the burden of bronchiolitis.


Asunto(s)
Bronquiolitis Viral/inmunología , Inmunidad Innata/inmunología , Proteínas de la Membrana/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Animales , Animales Recién Nacidos , Bronquiolitis Viral/prevención & control , Bronquiolitis Viral/virología , Proliferación Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Interferón Tipo I/inmunología , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/virología , Transducción de Señal/inmunología , Células TH1/inmunología
2.
J Virol ; 88(1): 325-38, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24155388

RESUMEN

In this study, subnucleocapsid nanorings formed by the recombinant nucleoprotein (N) of the respiratory syncytial virus were evaluated as a platform to anchor heterologous antigens. The ectodomain of the influenza virus A matrix protein 2 (M2e) is highly conserved and elicits protective antibodies when it is linked to an immunogenic carrier, making it a promising target to develop universal influenza vaccines. In this context, one or three M2e copies were genetically linked to the C terminus of N to produce N-M2e and N-3M2e chimeric recombinant nanorings. Mice were immunized intranasally with N-M2e or N-3M2e or with M2e or 3M2e control peptides. N-3M2e-vaccinated mice showed the strongest mucosal and systemic antibody responses. These mice presented a reduced viral load and minor weight loss, and all survived upon challenge with influenza virus A/PR8/34 (H1N1) (PR8). We compared the intranasal route to the subcutaneous route of N-3M2e immunization. Only the intranasal route induced a strong local IgA response and led to the protection of mice upon challenge. Finally, we demonstrated that the induction of anti-M2e antibodies by N-3M2e is not impaired by preexisting anti-N immunity. Overall, these results show that the N nanoring is a potent carrier for mucosal delivery of vaccinal antigens.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Membrana Mucosa/inmunología , Nanoestructuras , Proteínas de la Matriz Viral/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Vacunas contra la Influenza/inmunología , Ratones , Microscopía Electrónica de Transmisión , Nucleocápside , Reacción en Cadena en Tiempo Real de la Polimerasa , Carga Viral
3.
Vaccines (Basel) ; 9(3)2021 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-33803302

RESUMEN

The induction of long-lasting clinical and virological protection is needed for a successful vaccination program against the bovine respiratory syncytial virus (BRSV). In this study, calves with BRSV-specific maternally derived antibodies were vaccinated once, either with (i) a BRSV pre-fusion protein (PreF) and MontanideTM ISA61 VG (ISA61, n = 6), (ii) BRSV lacking the SH gene (ΔSHrBRSV, n = 6), (iii) a commercial vaccine (CV, n = 6), or were injected with ISA61 alone (n = 6). All calves were challenged with BRSV 92 days later and were euthanized 13 days post-infection. Based on clinical, pathological, and proteomic data, all vaccines appeared safe. Compared to the controls, PreF induced the most significant clinical and virological protection post-challenge, followed by ΔSHrBRSV and CV, whereas the protection of PreF-vaccinated calves was correlated with BRSV-specific serum immunoglobulin (Ig)G antibody responses 84 days post-vaccination, and the IgG antibody titers of ΔSHrBRSV- and CV-vaccinated calves did not differ from the controls on this day. Nevertheless, strong anamnestic BRSV- and PreF-specific IgG responses occurred in calves vaccinated with either of the vaccines, following a BRSV challenge. In conclusion, PreF and ΔSHrBRSV are two efficient one-shot candidate vaccines. By inducing a protection for at least three months, they could potentially improve the control of BRSV in calves.

4.
Vaccines (Basel) ; 8(2)2020 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-32443437

RESUMEN

Achieving safe and protective vaccination against respiratory syncytial virus (RSV) in infants and in calves has proven a challenging task. The design of recombinant antigens with a conformation close to their native form in virus particles is a major breakthrough. We compared two subunit vaccines, the bovine RSV (BRSV) pre-fusion F (preF) alone or with nanorings formed by the RSV nucleoprotein (preF+N). PreF and N proteins are potent antigenic targets for neutralizing antibodies and T cell responses, respectively. To tackle the challenges of neonatal immunization, three groups of six one-month-old calves with maternally derived serum antibodies (MDA) to BRSV received a single intramuscular injection of PreF, preF+N with MontanideTM ISA61 VG (ISA61) as adjuvant or only ISA61 (control). One month later, all calves were challenged with BRSV and monitored for virus replication in the upper respiratory tract and for clinical signs of disease over one week, and then post-mortem examinations of their lungs were performed. Both preF and preF+N vaccines afforded safe, clinical, and virological protection against BRSV, with little difference between the two subunit vaccines. Analysis of immune parameters pointed to neutralizing antibodies and antibodies to preF as being significant correlates of protection. Thus, a single shot vaccination with preF appears sufficient to reduce the burden of BRSV disease in calves with MDA.

5.
J Control Release ; 243: 146-159, 2016 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-27720994

RESUMEN

To put a Respiratory Syncytial Virus (RSV) vaccine onto the market, new vaccination strategies combining scientific and technical innovations need to be explored. Such a vaccine would also need to be adapted to the vaccination of young children that are the principal victims of acute RSV infection. In the present project, we describe the development and the preclinical evaluation of an original epicutaneous RSV vaccine that combines two technologies: Viaskin® epicutaneous patches as a delivery platform and RSV N-nanorings (N) as a subunit antigen. Such a needle-free vaccine may have a better acceptability for the vaccination of sensible population such as infants since it does not require any skin preparation. Moreover, this self-applicative vaccine would overcome some issues associated to injectable vaccines such as the requirement of sterile medical devices, the need of skilled health-care professionals and the necessity of stringent store conditions. Here, we demonstrate that Viaskin® patches loaded with a formulation containing N-nanorings (Viaskin®-N) are highly immunogenic in mice and promotes a Th1/Th17 oriented immune response. More importantly, Viaskin®-N epicutaneous vaccine confers a high level of protection against viral replication upon RSV challenge in mice, without exacerbating clinical symptoms. In swine, which provides the best experimental model for the transcutaneous passage of drug/antigen in human skin, we have shown that GFP fluorescent N-nanorings, delivered epicutaneously with Viaskin® patches, are taken up by epidermal Langerhans cells. We have also demonstrated that Viaskin®-N induced a significant RSV N-specific T-cell response in pig. In conclusion, Viaskin®-N epicutaneous vaccine seems efficient to protect against RSV infection in animal model.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Linfocitos T/inmunología , Replicación Viral/inmunología , Administración Cutánea , Animales , Femenino , Células de Langerhans/metabolismo , Ratones , Vacunas contra Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/farmacocinética , Absorción Cutánea , Especificidad de la Especie , Porcinos , Parche Transdérmico
6.
PLoS One ; 9(6): e100392, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24945377

RESUMEN

The development of safe and effective vaccines against both bovine and human respiratory syncytial viruses (BRSV, HRSV) to be used in the presence of RSV-specific maternally-derived antibodies (MDA) remains a high priority in human and veterinary medicine. Herein, we present safety and efficacy results from a virulent BRSV challenge of calves with MDA, which were immunized with one of three vaccine candidates that allow serological differentiation of infected from vaccinated animals (DIVA): an SH gene-deleted recombinant BRSV (ΔSHrBRSV), and two subunit (SU) formulations based on HRSV-P, -M2-1, and -N recombinant proteins displaying BRSV-F and -G epitopes, adjuvanted by either oil emulsion (Montanide ISA71VG, SUMont) or immunostimulating complex matrices (AbISCO-300, SUAbis). Whereas all control animals developed severe respiratory disease and shed high levels of virus following BRSV challenge, ΔSHrBRSV-immunized calves demonstrated almost complete clinical and virological protection five weeks after a single intranasal vaccination. Although mucosal vaccination with ΔSHrBRSV failed to induce a detectable immunological response, there was a rapid and strong anamnestic mucosal BRSV-specific IgA, virus neutralizing antibody and local T cell response following challenge with virulent BRSV. Calves immunized twice intramuscularly, three weeks apart with SUMont were also well protected two weeks after boost. The protection was not as pronounced as that in ΔSHrBRSV-immunized animals, but superior to those immunized twice subcutaneously three weeks apart with SUAbis. Antibody responses induced by the subunit vaccines were non-neutralizing and not directed against BRSV F or G proteins. When formulated as SUMont but not as SUAbis, the HRSV N, P and M2-1 proteins induced strong systemic cross-protective cell-mediated immune responses detectable already after priming. ΔSHrBRSV and SUMont are two promising DIVA-compatible vaccines, apparently inducing protection by different immune responses that were influenced by vaccine-composition, immunization route and regimen.


Asunto(s)
Anticuerpos Antivirales/inmunología , Genes Virales , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Virus Sincitial Respiratorio Bovino/genética , Virus Sincitial Respiratorio Bovino/inmunología , Virus Sincitial Respiratorio Humano/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/sangre , Bovinos , Epítopos/química , Epítopos/inmunología , Eliminación de Gen , Humanos , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ganglios Linfáticos/patología , Linfocitos/inmunología , Datos de Secuencia Molecular , Infecciones por Virus Sincitial Respiratorio/sangre , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Bovino/patogenicidad , Virus Sincitial Respiratorio Humano/inmunología , Especificidad de la Especie , Vacunación , Vacunas de Subunidad/efectos adversos , Vacunas de Subunidad/inmunología , Carga Viral , Virulencia
7.
PLoS One ; 7(5): e37722, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22655066

RESUMEN

BACKGROUND: The human respiratory syncytial virus (hRSV) is the leading cause of severe bronchiolitis in infants worldwide. The most severe RSV diseases occur between 2 and 6 months-of-age, so pediatric vaccination will have to be started within the first weeks after birth, when the immune system is prone to Th2 responses that may turn deleterious upon exposure to the virus. So far, the high risk to prime for immunopathological responses in infants has hampered the development of vaccine. In the present study we investigated the safety and efficacy of ring-nanostructures formed by the recombinant nucleoprotein N of hRSV (N(SRS)) as a mucosal vaccine candidate against RSV in BALB/c neonates, which are highly sensitive to immunopathological Th2 imprinting. METHODOLOGY AND PRINCIPAL FINDINGS: A single intranasal administration of N(SRS) with detoxified E. coli enterotoxin LT(R192G) to 5-7 day old neonates provided a significant reduction of the viral load after an RSV challenge at five weeks of age. However, neonatal vaccination also generated an enhanced lung infiltration by neutrophils and eosinophils following the RSV challenge. Analysis of antibody subclasses and cytokines produced after an RSV challenge or a boost administration of the vaccine suggested that neonatal vaccination induced a Th2 biased local immune memory. This Th2 bias and the eosinophilic reaction could be prevented by adding CpG to the vaccine formulation, which, however did not prevent pulmonary inflammation and neutrophil infiltration upon viral challenge. CONCLUSIONS/SIGNIFICANCE: In conclusion, protective vaccination against RSV can be achieved in neonates but requires an appropriate combination of adjuvants to prevent harmful Th2 imprinting.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Nanoestructuras/uso terapéutico , Nucleoproteínas/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/uso terapéutico , Virus Sincitiales Respiratorios/inmunología , Proteínas Virales/uso terapéutico , Adyuvantes Inmunológicos/administración & dosificación , Administración Intranasal , Animales , Islas de CpG , Humanos , Recién Nacido , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Nanoestructuras/administración & dosificación , Nanoestructuras/química , Nucleoproteínas/administración & dosificación , Nucleoproteínas/química , Nucleoproteínas/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/química , Vacunas contra Virus Sincitial Respiratorio/inmunología , Células Th2/inmunología , Células Th2/patología , Proteínas Virales/administración & dosificación , Proteínas Virales/química , Proteínas Virales/inmunología
8.
Vaccine ; 28(21): 3722-34, 2010 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-20307593

RESUMEN

Human and bovine respiratory syncytial viruses (HRSV and BRSV) are two closely related, worldwide prevalent viruses that are the leading cause of severe airway disease in children and calves, respectively. Efficacy of commercial bovine vaccines needs improvement and no human vaccine is licensed yet. We reported that nasal vaccination with the HRSV nucleoprotein produced as recombinant ring-shaped nanoparticles (N(SRS)) protects mice against a viral challenge with HRSV. The aim of this work was to evaluate this new vaccine that uses a conserved viral antigen, in calves, natural hosts for BRSV. Calves, free of colostral or natural anti-BRSV antibodies, were vaccinated with N(SRS) either intramuscularly, or both intramuscularly and intranasally using Montanide ISA71 and IMS4132 as adjuvants and challenged with BRSV. All vaccinated calves developed anti-N antibodies in blood and nasal secretions and N-specific cellular immunity in local lymph nodes. Clinical monitoring post-challenge demonstrated moderate respiratory pathology with local lung tissue consolidations for the non-vaccinated calves that were significantly reduced in the vaccinated calves. Vaccinated calves had lower viral loads than the non-vaccinated control calves. Thus N(SRS) vaccination in calves provided cross-protective immunity against BRSV infection without adverse inflammatory reaction.


Asunto(s)
Enfermedades de los Bovinos/prevención & control , Nucleoproteínas/inmunología , Infecciones por Virus Sincitial Respiratorio/veterinaria , Vacunas contra Virus Sincitial Respiratorio/inmunología , Proteínas Virales/inmunología , Adyuvantes Inmunológicos/farmacología , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/sangre , Formación de Anticuerpos , Bovinos , Enfermedades de los Bovinos/inmunología , Protección Cruzada , Inmunidad Celular , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Masculino , Datos de Secuencia Molecular , Nanopartículas , Proteínas Recombinantes/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Bovino/inmunología , Vacunas de Subunidad/inmunología , Carga Viral
9.
PLoS One ; 3(3): e1766, 2008 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-18335041

RESUMEN

BACKGROUND: Bronchiolitis caused by the respiratory syncytial virus (RSV) in infants less than two years old is a growing public health concern worldwide, and there is currently no safe and effective vaccine. A major component of RSV nucleocapsid, the nucleoprotein (N), has been so far poorly explored as a potential vaccine antigen, even though it is a target of protective anti-viral T cell responses and is remarkably conserved between human RSV A and B serotypes. We recently reported a method to produce recombinant N assembling in homogenous rings composed of 10-11 N subunits enclosing a bacterial RNA. These nanoparticles were named sub-nucleocapsid ring structure (N SRS). METHODOLOGY AND PRINCIPAL FINDINGS: The vaccine potential of N SRS was evaluated in a well-characterized and widely acknowledged mouse model of RSV infection. BALB/c adult mice were immunized intranasally with N SRS adjuvanted with the detoxified E. coli enterotoxin LT(R192G). Upon RSV challenge, vaccinated mice were largely protected against virus replication in the lungs, with a mild inflammatory lymphocytic and neutrophilic reaction in their airways. Mucosal immunization with N SRS elicited strong local and systemic immunity characterized by high titers of IgG1, IgG2a and IgA anti-N antibodies, antigen-specific CD8(+) T cells and IFN-gamma-producing CD4(+) T cells. CONCLUSIONS/SIGNIFICANCE: This is the first report of using nanoparticles formed by the recombinant nucleocapsid protein as an efficient and safe intra-nasal vaccine against RSV.


Asunto(s)
Nanopartículas , Virus Sincitiales Respiratorios/inmunología , Vacunas Virales/administración & dosificación , Administración Intranasal , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Microscopía Electrónica , Mutagénesis Sitio-Dirigida , Vacunas Virales/inmunología
10.
Vaccine ; 25(34): 6373-80, 2007 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-17629366

RESUMEN

Development of a safe, cheap and efficient vaccine against rotavirus is important to reduce the morbidity and mortality associated with gastroenteritis in infants worldwide. High quantities of two inner core rotavirus-derived proteins (VP2 and a nonglycosylated mutant VP6 (VP6(NG)) from the RF81 bovine strain) were produced in the milk of transgenic rabbits. We show here that rectal administration of partially purified milk-derived VP2 and VP6(NG) proteins with the detoxified LT(R192G) adjuvant almost completely prevented fecal shedding induced by a highly infectious challenge in mice with the murine ECw strain. The vaccine generated rotavirus-specific fecal secretory IgA, systemic IgG and IgA and a rotavirus-specific Th1 response. We thus demonstrate in clinically feasible settings that mass production of viral protein in transgenic milk is a promising way to generate subunit vaccine against rotavirus.


Asunto(s)
Antígenos Virales/inmunología , Proteínas de la Cápside/inmunología , Vacunas contra Rotavirus/inmunología , Vacunas Sintéticas/inmunología , Animales , Animales Modificados Genéticamente , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Proteínas de la Cápside/genética , Femenino , Inmunización , Ratones , Ratones Endogámicos BALB C , Leche/metabolismo , Conejos , Recto , Vacunas contra Rotavirus/administración & dosificación , Células TH1/inmunología , Células Th2/inmunología , Vacunas Sintéticas/administración & dosificación
11.
J Gen Virol ; 87(Pt 8): 2145-2148, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16847109

RESUMEN

Bovine respiratory syncytial virus (BRSV) is able to counteract the alpha/beta interferon (IFN-alpha/beta)-mediated antiviral response for efficient replication in a host-specific manner. Mice models have been developed for experimental infection with human, but not bovine, respiratory syncytial virus strains. Here, it is shown that BRSV can replicate efficiently on primary cell cultures derived from type I IFN receptor-deficient, but not from wild-type IFN-competent, mice. However, BRSV infection was not enhanced in mice devoid of the type I IFN receptor. These results show that type I IFN is a major host-range determinant for infection at the cellular level, but that other factors control virus replication and pathology in vivo.


Asunto(s)
Proteínas de la Membrana/fisiología , Receptores de Interferón/fisiología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Bovino/fisiología , Replicación Viral , Animales , Células Cultivadas , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Microscopía Fluorescente , Receptor de Interferón alfa y beta , Receptores de Interferón/genética
12.
J Virol ; 80(4): 1752-61, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16439532

RESUMEN

To evaluate whether the rectal route of immunization may be used to provide appropriate protection against enteric pathogens such as rotaviruses (RV), we studied the antibody response and the protection induced by rectal immunization of mice with RV virus-like particles (VLP). For this purpose, 6-week-old BALBc mice were rectally immunized twice with RV 8-2/6/7-VLP derived from the bovine RV RF81 strain either alone or combined with various adjuvants including four toxins [cholera toxin (CT) and three attenuated Escherichia coli-derived heat-labile toxins (LTs), LT(R192G), LT(R72), and LT(K63)] and two Toll-like receptor-targeting adjuvants (CpG and resiquimod). Six weeks after the second immunization, mice were challenged with murine RV strain ECw. RV VLP administered alone were not immunogenic and did not protect mice against RV challenge. By contrast, RV VLP combined with any of the toxin adjuvants were immunogenic (mice developed significant titers of anti-RV immunoglobulin A [IgA] in both serum and feces and of anti-RV IgG in serum) and either efficiently induced complete protection of the mice (no detectable fecal virus shedding) or, for LT(K63), reduced the amount of fecal virus shedding after RV challenge. When combined with RV VLP, CpG and resiquimod failed to achieve protection, although CpG efficiently induced an antibody response to RV. These results support the consideration of the rectal route for the development of new immunization strategies against RV infection. Rectal delivery of a VLP-based vaccine might allow the use of adjuvants less toxic than, but as efficient as, CT.


Asunto(s)
Anticuerpos Antivirales/análisis , Inmunidad Mucosa , Infecciones por Rotavirus/prevención & control , Vacunas contra Rotavirus/inmunología , Rotavirus/inmunología , Vacunación/métodos , Adyuvantes Inmunológicos/administración & dosificación , Administración Rectal , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/análisis , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Heces/virología , Femenino , Inmunización Secundaria , Inmunoglobulina A/análisis , Inmunoglobulina G/análisis , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos BALB C , Vacunas contra Rotavirus/administración & dosificación , Esparcimiento de Virus
13.
Vaccine ; 23(10): 1258-64, 2005 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-15652668

RESUMEN

The presence of unmethylated CpG motifs in bacterial plasmids is thought to provide necessary immunoadjuvant signals to DNA vaccination. We took advantage of CpG-unresponsive toll-like receptor 9 (TLR9) knock-out mice to study whether this pathway was required to generate immune responses to DNA vaccination. We compared two vectors, one encoding the surface glycoprotein C of pseudorabies virus shown to protect target animals against challenge, and the other encoding the cytoplasmic enzyme beta-galactosidase. In the absence of TLR9, bone marrow-derived dendritic cells lost their ability to secrete IL-12 and type I IFN in response not only to CpG as expected but also to the plasmids used for vaccination. In contrast, DNA vaccination experiments showed that TLR9-deficient mice were able to mount Th1-biased antigen-specific antibody and IFN-gamma responses, albeit at lower levels than normal mice. Thus, TLR9 signaling is not needed for eliciting T- and B-cell responses to DNA encoded antigens. However, TLR9 signaling tended to enhance plasmid-adjuvant effects on antigen-specific immune responses.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Proteínas de Unión al ADN/fisiología , Plásmidos/inmunología , Receptores de Superficie Celular/fisiología , Vacunas de ADN/inmunología , Animales , Anticuerpos/análisis , Cartilla de ADN , Proteínas de Unión al ADN/genética , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunidad Celular/genética , Inmunidad Celular/inmunología , Inmunización , Indicadores y Reactivos , Interferón gamma/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Superficie Celular/genética , Células TH1/inmunología , Receptor Toll-Like 9
14.
Pediatr Res ; 54(5): 739-46, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12904595

RESUMEN

Previous work has shown that the indigenous gut microflora in mice plays a protective role against Escherichia coli heat-labile enterotoxin (LT)-mediated abrogation of oral tolerance to an unrelated co-ingested protein. To assess potential protection by human gut microflora, we studied the effect of human gut microflora in a murine model. Oral tolerance was studied in adult gnotobiotic mice (i.e. ex-germ-free mice) colonized with the entire human fecal microflora and orally administered once with LT and ovalbumin. Systemic suppression of IgG, IgG1, IgG2a, and IgE antibody responses was assessed by ELISA. Both specific IgG subclasses and IgE hyporesponsiveness was induced in LT + ovalbumin-fed gnotobiotic mice, indicating that the human gut microflora can protect against the LT-mediated abrogation of oral tolerance. However, as confirmed with mouse gut microflora, this protective effect only occurs when the gut microflora is associated from birth on. Colonization of germ-free mice with a single bacterial strain, E. coli, predominant in the human and mouse gut microflora in the neonatal period, showed that this strain alone did not induce protection. These results supported the hypothesis that the natural establishment of the gut microflora in neonates crucially influenced resistance to LT-mediated abrogation of oral tolerance by reinforcing suppression of both T helper type 1- and T helper type 2-controlled responses, and suggested that sequential bacterial colonization of the gut rather than a single bacterial species may be involved in this phenomenon.


Asunto(s)
Toxinas Bacterianas/inmunología , Enterotoxinas/inmunología , Proteínas de Escherichia coli , Escherichia coli/metabolismo , Heces/microbiología , Vida Libre de Gérmenes , Tolerancia Inmunológica , Intestinos/microbiología , Ovalbúmina/inmunología , Administración Oral , Animales , Anticuerpos/inmunología , Anticuerpos/metabolismo , Toxinas Bacterianas/administración & dosificación , Enterotoxinas/administración & dosificación , Escherichia coli/inmunología , Calor , Humanos , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos C3H , Ovalbúmina/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA