Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Sci Rep ; 8(1): 18007, 2018 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-30573748

RESUMEN

Cell mediated immunity plays a vital role in defense against influenza infection in humans. Less is known about the role of vaccine-induced cell mediated immunity and the cytokine responses elicited. We measured CD4+ and CD8+ T-cell reactivity in human subjects following vaccination with licensed trivalent influenza vaccine and a novel virus-like particle based vaccine. We detected influenza-specific CD4+ T-cell responses following vaccination with the licensed trivalent influenza vaccine and found that these correlated with antibody measurements. Administration of the novel virus-like particle based vaccine elicited influenza-specific CD4+ and CD8+ T-cell responses and the induction of the cytokines IFN-γ, IL-17A, IL17F, IL-5, IL-13, IL-9, IL-10 and IL-21. Pre-existing cytokine responses influenced the profile of the cytokine response elicited by vaccination. In a subset of individuals the VLP vaccine changed pre-vaccination production of type 2 cytokines such as IL-5 and IL-13 to a post-vaccination type 1 cytokine signature characterized by IFN-γ. A transcriptional signature to vaccination was found to correlate with antibody titer, IFN-γ production by T-cells and expression of a putative RNA helicase, DDX17, on the surface of immune cells.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/fisiología , Citocinas/metabolismo , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Activación de Linfocitos/inmunología , Adulto , Formación de Anticuerpos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Estudios de Cohortes , Femenino , Humanos , Inmunidad Celular/inmunología , Gripe Humana/inmunología , Gripe Humana/metabolismo , Masculino , Persona de Mediana Edad , Vacunación , Adulto Joven
2.
Malar J ; 16(1): 115, 2017 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-28288639

RESUMEN

BACKGROUND: Non-human primates, such as the rhesus macaques, are the preferred model for down-selecting human malaria vaccine formulations, but the rhesus model is expensive and does not allow for direct efficacy testing of human malaria vaccines. Transgenic rodent parasites expressing genes of human Plasmodium are now routinely used for efficacy studies of human malaria vaccines. Mice have however rarely predicted success in human malaria trials and there is scepticism whether mouse studies alone are sufficient to move a vaccine candidate into the clinic. METHODS: A comparison of immunogenicity, fine-specificity and functional activity of two Alum-adjuvanted Plasmodium falciparum circumsporozoite protein (CSP)-based vaccines was conducted in mouse and rhesus models. One vaccine was a soluble recombinant protein (CSP) and the other was the same CSP covalently conjugated to the Qß phage particle (Qß-CSP). RESULTS: Mice showed different kinetics of antibody responses and different sensitivity to the NANP-repeat and N-terminal epitopes as compared to rhesus. While mice failed to discern differences between the protective efficacy of CSP versus Qß-CSP vaccine following direct challenge with transgenic Plasmodium berghei parasites, rhesus serum from the Qß-CSP-vaccinated animals induced higher in vivo sporozoite neutralization activity. CONCLUSIONS: Despite some immunologic parallels between models, these data demonstrate that differences between the immune responses induced in the two models risk conflicting decisions regarding potential vaccine utility in humans. In combination with historical observations, the data presented here suggest that although murine models may be useful for some purposes, non-human primate models may be more likely to predict the human response to investigational vaccines.


Asunto(s)
Vacunas contra la Malaria/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Animales , Femenino , Inmunogenicidad Vacunal , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/inmunología
3.
Front Immunol ; 8: 226, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28321220

RESUMEN

Since the discovery of the first virus-like particle (VLP) derived from hepatitis B virus in 1980 (1), the field has expanded substantially. Besides successful use of VLPs as safe autologous virus-targeting vaccines, the powerful immunogenicity of VLPs has been also harnessed to generate immune response against heterologous and even self-antigens (2-4). Linking adjuvants to VLPs displaying heterologous antigen ensures simultaneous delivery of all vaccine components to the same antigen-presenting cells. As a consequence, antigen-presenting cells, such as dendritic cells, will process and present the antigen displayed on VLPs while receiving costimulatory signals by the VLP-incorporated adjuvant. Similarly, antigen-specific B cells recognizing the antigen linked to the VLP are simultaneously exposed to the adjuvant. Here, we demonstrate in mice that physical association of antigen, carrier (VLPs), and adjuvant is more critical for B than T cell responses. As a model system, we used the E7 protein from human papilloma virus, which spontaneously forms oligomers with molecular weight ranging from 158 kDa to 10 MDa at an average size of 50 nm. E7 oligomers were either chemically linked or simply mixed with VLPs loaded with DNA rich in non-methylated CG motifs (CpGs), a ligand for toll-like receptor 9. E7-specific IgG responses were strongly enhanced if the antigen was linked to the VLPs. In contrast, both CD4+ and CD8+ T cell responses as well as T cell-mediated protection against tumor growth were comparable for linked and mixed antigen formulations. Therefore, our data show that B cell but not T cell responses require antigen-linkage to the carrier and adjuvant for optimal vaccination outcome.

4.
PLoS One ; 12(2): e0171923, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28178353

RESUMEN

Misfolding of the cellular prion protein (PrPC) into the scrapie prion protein (PrPSc) results in progressive, fatal, transmissible neurodegenerative conditions termed prion diseases. Experimental and epidemiological evidence point toward a protracted, clinically silent phase in prion diseases, yet there is no diagnostic test capable of identifying asymptomatic individuals incubating prions. In an effort to identify early biomarkers of prion diseases, we have compared global transcriptional profiles in brains from pre-symptomatic prion-infected mice and controls. We identified Cst7, which encodes cystatin F, as the most strongly upregulated transcript in this model. Early and robust upregulation of Cst7 mRNA levels and of its cognate protein was validated in additional mouse models of prion disease. Surprisingly, we found no significant increase in cystatin F levels in both cerebrospinal fluid or brain parenchyma of patients with Creutzfeldt-Jakob disease compared to Alzheimer's disease or non-demented controls. Our results validate cystatin F as a useful biomarker of early pathogenesis in experimental models of prion disease, and point to unexpected species-specific differences in the transcriptional responses to prion infections.


Asunto(s)
Cistatinas/metabolismo , Enfermedades por Prión/metabolismo , Animales , Biomarcadores , Encéfalo/metabolismo , Encéfalo/patología , Cistatinas/líquido cefalorraquídeo , Cistatinas/genética , Ensayo de Inmunoadsorción Enzimática , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Ratones , Enfermedades por Prión/líquido cefalorraquídeo , Enfermedades por Prión/genética , Enfermedades por Prión/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo
5.
Mol Ther ; 24(5): 1003-12, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26686385

RESUMEN

Interleukin-1ß (IL-1ß) is a key cytokine involved in inflammatory illnesses including rare hereditary diseases and common chronic inflammatory conditions as gout, rheumatoid arthritis, and type 2 diabetes mellitus, suggesting reduction of IL-1ß activity as new treatment strategy. The objective of our study was to assess safety, antibody response, and preliminary efficacy of a novel vaccine against IL-1ß. The vaccine hIL1bQb consisting of full-length, recombinant IL-1ß coupled to virus-like particles was tested in a preclinical and clinical, randomized, placebo-controlled, double-blind study in patients with type 2 diabetes. The preclinical simian study showed prompt induction of IL-1ß-specific antibodies upon vaccination, while neutralizing antibodies appeared with delay. In the clinical study with 48 type 2 diabetic patients, neutralizing IL-1ß-specific antibody responses were detectable after six injections with doses of 900 µg. The development of neutralizing antibodies was associated with higher number of study drug injections, lower baseline body mass index, improvement of glycemia, and C-reactive protein (CRP). The vaccine hIL1bQb was safe and well-tolerated with no differences regarding adverse events between patients receiving hIL1bQb compared to placebo. This is the first description of a vaccine against IL-1ß and represents a new treatment option for IL-1ß-dependent diseases such as type 2 diabetes mellitus (ClinicalTrials.gov NCT00924105).


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Neutralizantes/administración & dosificación , Diabetes Mellitus Tipo 2/terapia , Interleucina-1beta/inmunología , Vacunas/administración & dosificación , Adulto , Anciano , Animales , Diabetes Mellitus Tipo 2/inmunología , Método Doble Ciego , Femenino , Humanos , Macaca mulatta , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Vacunas/inmunología
6.
PLoS One ; 10(11): e0142035, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26571021

RESUMEN

Circumsporozoite protein (CSP) of Plasmodium falciparum is a promising malaria vaccine target. RTS,S, the most advanced malaria vaccine candidate consists of the central NANP repeat and carboxy-terminal region of CSP displayed on a hepatitis B virus-like particle (VLP). To build upon the success of RTS,S, we produced a near full-length Plasmodium falciparum CSP that also includes the conserved amino-terminal region of CSP. We recently showed that this soluble CSP, combined with a synthetic Toll-like-receptor-4 (TLR4) agonist in stable oil-in-water emulsion (GLA/SE), induces a potent and protective immune response in mice against transgenic parasite challenge. Here we have investigated whether the immunogenicity of soluble CSP could be further augmented by presentation on a VLP. Bacteriophage Qß VLPs can be readily produced in E.coli, they have a diameter of 25 nm and contain packaged E. coli RNA which serves as a built in adjuvant through the activation of TLR7/8. CSP was chemically conjugated to Qß and the CSP-Qß vaccine immunogenicity and efficacy were compared to adjuvanted soluble CSP in the C57Bl/6 mouse model. When formulated with adjuvants lacking a TLR4 agonist (Alum, SE and Montanide) the Qß-CSP induced higher anti-NANP repeat titers, higher levels of cytophilic IgG2b/c antibodies and a trend towards higher protection against transgenic parasite challenge as compared to soluble CSP formulated in the same adjuvant. The VLP and soluble CSP immunogenicity difference was most pronounced at low antigen dose, and within the CSP molecule, the titers against the NANP repeats were preferentially enhanced by Qß presentation. While a TLR4 agonist enhanced the immunogenicity of soluble CSP to levels comparable to the VLP vaccine, the TLR4 agonist did not further improve the immunogenicity of the Qß-CSP vaccine. The data presented here pave the way for further improvement in the Qß conjugation chemistry and evaluation of both the Qß-CSP and soluble CSP vaccines in the non-human primate model.


Asunto(s)
Vacunas contra la Malaria/química , Plasmodium falciparum/inmunología , Proteínas Protozoarias/química , Vacunas Sintéticas/química , Vacunas de Partículas Similares a Virus/inmunología , Adyuvantes Inmunológicos/química , Allolevivirus/metabolismo , Compuestos de Alumbre/química , Animales , Anticuerpos Antiprotozoarios/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Epítopos/química , Escherichia coli/genética , Femenino , Sistema Inmunológico , Inmunoglobulina G/inmunología , Lipopolisacáridos/química , Malaria/prevención & control , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Parasitemia/parasitología , Proteínas Protozoarias/genética , ARN Bacteriano/genética , Proteínas Recombinantes/química , Esporozoítos/química , Receptor Toll-Like 4/metabolismo
7.
Vaccine ; 32(39): 5041-8, 2014 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-25045806

RESUMEN

METHODS: A novel, fully bacterially produced recombinant virus-like particle (VLP) based influenza vaccine (gH1-Qbeta) against A/California/07/2009(H1N1) was tested in a double-blind, randomized phase I clinical trial at two clinical sites in Singapore. The trial evaluated the immunogenicity and safety of gH1-Qbeta in the presence or absence of alhydrogel adjuvant. Healthy adult volunteers with no or low pre-existing immunity against A/California/07/2009 (H1N1) were randomized to receive two intramuscular injections 21 days apart, with 100µg vaccine, containing 42µg hemagglutinin antigen. Antibody responses were measured before and 21 days after each immunization by hemagglutination inhibition (HAI) assays. The primary endpoint was seroconversion on Day 42, defined as percentage of subjects which reach a HAI titer ≥40 or achieve an at least 4-fold rise in HAI titer (with pre-existing immunity). The co-secondary endpoints were safety and seroconversion on Day 21. RESULTS: A total of 84 Asian volunteers were enrolled in this study and randomized to receive the adjuvanted (n=43) or the non-adjuvanted (n=41) vaccine. Of those, 43 and 37 respectively (95%) completed the study. There were no deaths or serious adverse events reported during this trial. A total of 535 adverse events occurred during treatment with 49.5% local solicited symptoms, of mostly (76.4%) mild severity. The most common treatment-related systemic symptom was fatigue. The non-adjuvanted vaccine met all primary and secondary endpoints and showed seroconversion in 62.2% and 70.3% of participants respectively on Day 21 and Day 42. While the adjuvanted vaccine showed an increased seroconversion from 25.5% (Day 21) to 51.2% (Day 42), it did not meet the immunogenicity endpoint. CONCLUSION: In summary, non-adjuvanted gH1-Qbeta showed similar antibody mediated immunogenicity and a comparable safety profile in healthy humans to commercially available vaccines. These results warrant the consideration of this VLP vaccine platform for the vaccination against influenza infection (HSA CTC1300092).


Asunto(s)
Formación de Anticuerpos , Subtipo H1N1 del Virus de la Influenza A , Vacunas contra la Influenza/uso terapéutico , Gripe Humana/prevención & control , Adyuvantes Inmunológicos/administración & dosificación , Adulto , Hidróxido de Aluminio/administración & dosificación , Anticuerpos Antivirales/sangre , Método Doble Ciego , Determinación de Punto Final , Femenino , Pruebas de Inhibición de Hemaglutinación , Humanos , Masculino , Persona de Mediana Edad , Singapur , Vacunas de Partículas Similares a Virus/uso terapéutico , Adulto Joven
8.
J Immunol ; 192(12): 5499-508, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24821969

RESUMEN

Extensive studies have been undertaken to describe naive B cells differentiating into memory B cells at a cellular and molecular level. However, relatively little is known about the fate of memory B cells upon Ag re-encounter. We have previously established a system based on virus-like particles (VLPs), which allows tracking of VLP-specific B cells by flow cytometry as well as histology. Using allotype markers, it is possible to adoptively transfer memory B cells into a naive mouse and track responses of naive and memory B cells in the same mouse under physiological conditions. We have observed that VLP-specific memory B cells quickly differentiated into plasma cells that drove the early onset of a strong humoral IgG response. However, neither IgM(+) nor IgG(+) memory B cells proliferated extensively or entered germinal centers. Remarkably, plasma cells derived from memory B cells preferentially homed to the bone marrow earlier and secreted increased levels of Abs when compared with primary plasma cells derived from naive B cells. Hence, memory B cells have the unique phenotype to differentiate into highly effective secondary plasma cells.


Asunto(s)
Allolevivirus/inmunología , Anticuerpos Antivirales/inmunología , Diferenciación Celular/inmunología , Memoria Inmunológica , Células Plasmáticas/inmunología , Animales , Inmunización , Ratones
9.
PLoS One ; 8(10): e76571, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24204639

RESUMEN

Influenza pandemics can spread quickly and cost millions of lives; the 2009 H1N1 pandemic highlighted the shortfall in the current vaccine strategy and the need for an improved global response in terms of shortening the time required to manufacture the vaccine and increasing production capacity. Here we describe the pre-clinical assessment of a novel 2009 H1N1 pandemic influenza vaccine based on the E. coli-produced HA globular head domain covalently linked to virus-like particles derived from the bacteriophage Qß. When formulated with alum adjuvant and used to immunize mice, dose finding studies found that a 10 µg dose of this vaccine (3.7 µg globular HA content) induced antibody titers comparable to a 1.5 µg dose (0.7 µg globular HA content) of the licensed 2009 H1N1 pandemic vaccine Panvax, and significantly reduced viral titers in the lung following challenge with 2009 H1N1 pandemic influenza A/California/07/2009 virus. While Panvax failed to induce marked T cell responses, the novel vaccine stimulated substantial antigen-specific interferon-γ production in splenocytes from immunized mice, alongside enhanced IgG2a antibody production. In ferrets the vaccine elicited neutralizing antibodies, and following challenge with influenza A/California/07/2009 virus reduced morbidity and lowered viral titers in nasal lavages.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Células TH1/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Adyuvantes Inmunológicos , Compuestos de Alumbre , Animales , Especificidad de Anticuerpos , Bacteriófagos/inmunología , Escherichia coli/genética , Escherichia coli/inmunología , Femenino , Hurones/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Inmunoglobulina G/inmunología , Interferón gamma/biosíntesis , Ratones , Infecciones por Orthomyxoviridae/prevención & control , ARN Bacteriano/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células TH1/metabolismo
10.
PLoS One ; 8(11): e78947, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24260136

RESUMEN

Although current influenza vaccines are effective in general, there is an urgent need for the development of new technologies to improve vaccine production timelines, capacities and immunogenicity. Herein, we describe the development of an influenza vaccine technology which enables recombinant production of highly efficient influenza vaccines in bacterial expression systems. The globular head domain of influenza hemagglutinin, comprising most of the protein's neutralizing epitopes, was expressed in E. coli and covalently conjugated to bacteriophage-derived virus-like particles produced independently in E.coli. Conjugate influenza vaccines produced this way were used to immunize mice and found to elicit immune sera with high antibody titers specific for the native influenza hemagglutinin protein and high hemagglutination-inhibition titers. Moreover vaccination with these vaccines induced full protection against lethal challenges with homologous and highly drifted influenza strains.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Potencia de la Vacuna , Animales , Anticuerpos Antivirales/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Inmunización , Subtipo H1N1 del Virus de la Influenza A/genética , Vacunas contra la Influenza/biosíntesis , Vacunas contra la Influenza/genética , Ratones
11.
Am J Rhinol Allergy ; 27(3): 206-12, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23710957

RESUMEN

BACKGROUND: The incidence of allergic rhinitis (AR) has increased constantly over the last decades. The disease can significantly lower quality of life and subsequently might progress to allergic asthma. Allergen-specific immunotherapy is mostly used to cope with the cause of the disease. However, incidence of systemic reactions or limited compliance hampers the widespread use of this therapeutic approach. Therefore, new candidates are examined to improve immunotherapy of allergies. Recently, a new technology was developed with the aim to positively influence the immune system of allergic patients. Virus-like particles (VLPs) represent a potent vaccine platform that has been proven to be immunogenic and clinically effective. To enhance immune cell activation, addition of Toll-like receptor ligands and/or depot-forming adjuvants seems to be helpful. In this context, CpG motifs represent intensive investigated and potent stimulators of T cells. This article focuses on the function of VLPs and CpG motifs and their clinical experience for treatment of AR. METHODS: A literature review was performed. RESULTS: Several published studies showed a beneficial impact of the treatment on allergic symptoms. They tested VLPs filled with or without CpG motifs in combination with or without allergen. CONCLUSION: Results encourage further investigations of VLPs and CpG motifs as adjuncts to or even alternative candidates for immunotherapy of allergic disorders.


Asunto(s)
Islas de CpG/inmunología , Inmunoterapia , Rinitis Alérgica Perenne/inmunología , Rinitis Alérgica Perenne/terapia , Receptores Toll-Like/administración & dosificación , Vacunas de Partículas Similares a Virus/administración & dosificación , Ensamble de Virus/inmunología , Alérgenos/administración & dosificación , Humanos , Inmunoterapia/métodos , Rinitis Alérgica , Resultado del Tratamiento
12.
Proc Natl Acad Sci U S A ; 109(50): 20566-71, 2012 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-23169669

RESUMEN

The lung is an important entry site for pathogens; its exposure to antigens results in systemic as well as local IgA and IgG antibodies. Here we show that intranasal administration of virus-like particles (VLPs) results in splenic B-cell responses with strong local germinal-center formation. Surprisingly, VLPs were not transported from the lung to the spleen in a free form but by B cells. The interaction between VLPs and B cells was initiated in the lung and occurred independently of complement receptor 2 and Fcγ receptors, but was dependent upon B-cell receptors. Thus, B cells passing through the lungs bind VLPs via their B-cell receptors and deliver them to local B cells within the splenic B-cell follicle. This process is fundamentally different from delivery of blood or lymph borne particulate antigens, which are transported into B cell follicles by binding to complement receptors on B cells.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/virología , Vacunas de Partículas Similares a Virus/inmunología , Administración Intranasal , Traslado Adoptivo , Animales , Anticuerpos Antivirales/biosíntesis , Antígenos Virales/administración & dosificación , Antígenos Virales/sangre , Movimiento Celular/inmunología , Femenino , Inmunoglobulina G/biosíntesis , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de Complemento 3d/inmunología , Receptores de IgG/inmunología , Bazo/inmunología , Bazo/virología , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/sangre
13.
Eur J Immunol ; 42(4): 863-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22531913

RESUMEN

A vaccine protecting against all influenza strains is a long-sought goal, particularly for emerging pandemics. As previously shown, vaccines based on the highly conserved extracellular domain of M2 (M2e) may protect against all influenza A strains. Here, we demonstrate that M2e-specific monoclonal antibodies (mAbs) protect mice from a lethal influenza infection. To be protective, antibodies had to be able to bind to Fc receptors and fix complement. Furthermore, mAbs of IgG2c isotype were protective in mice, while antibodies of identical specificity, but of the IgG1 isotype, failed to prevent disease. These findings readily translated into vaccine design. A vaccine targeting M2 in the absence of a toll-like receptor (TLR) 7 ligand primarily induced IgG1, whilst the same vaccine linked to a TLR7 ligand yielded high levels of IgG2c antibodies. Although both vaccines protected mice from a lethal challenge, mice treated with the vaccine containing a TLR7 ligand showed significantly lower morbidity. In accordance with these findings, vaccination of TLR7(-/-) mice with a vaccine containing a TLR7 ligand did not result in protection from a lethal challenge. Hence, the innate immune system is required to direct isotype switching toward the more protective IgG2a/c antibodies.


Asunto(s)
Anticuerpos Antivirales/inmunología , Inmunoglobulina G/inmunología , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Glicoproteínas de Membrana/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Transducción de Señal/inmunología , Receptor Toll-Like 7/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino/inmunología , Anticuerpos Monoclonales de Origen Murino/farmacología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/genética , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Cambio de Clase de Inmunoglobulina/efectos de los fármacos , Cambio de Clase de Inmunoglobulina/genética , Inmunoglobulina G/sangre , Inmunoglobulina G/genética , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Vacunas contra la Influenza/farmacología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/sangre , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Pandemias , Transducción de Señal/genética , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo , Vacunación
14.
Immunity ; 34(3): 375-84, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21353603

RESUMEN

The contribution of Toll-like receptor (TLR) signaling to T cell-dependent (TD) antibody responses was assessed by using mice lacking the TLR signaling adaptor MyD88 in individual cell types. When a soluble TLR9 ligand was used as adjuvant for a protein antigen, MyD88 was required in dendritic cells but not in B cells to enhance the TD antibody response, regardless of the inherent immunogenicity of the antigen. In contrast, a TLR9 ligand contained within a virus-like particle substantially augmented the TD germinal center IgG antibody response, and this augmentation required B cell MyD88. The ability of B cells to discriminate between antigens based on the physical form of a TLR ligand probably reflects an adaptation to facilitate strong antiviral antibody responses.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal , Receptor Toll-Like 9/inmunología , Animales , Anticuerpos Antivirales/sangre , Células Dendríticas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Inmunológicos , Orthomyxoviridae/inmunología
15.
J Allergy Clin Immunol ; 126(2): 375-83, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20624641

RESUMEN

BACKGROUND: Allergen-specific desensitization (SIT) is the most effective therapy for allergies. Although allergen-specific antibodies have an important role in the process, mechanisms of IgG-mediated inhibition of allergic reactions are not well defined. OBJECTIVE: We investigated mechanisms by which SIT-induced allergen-specific IgGs inhibit allergic reactions. METHODS: We generated mAbs that recognize 3 nonoverlapping epitopes of the major cat allergen Fel d 1. Each of the mAbs was produced as an IgE and different IgG isotype. RESULTS: IgEs against 2 nonoverlapping epitopes on Fel d 1 are necessary and sufficient to sensitize mast cells for maximal FcepsilonRI signaling and degranulation on exposure to monomeric Fel d 1. IgE antibodies of a third specificity did not further increase mast cell degranulation, indicating that formation of large FcepsilonRI clusters are not required to induce maximal activation of mast cells. A single IgG that was specific for an epitope different from those recognized by the IgEs was a potent inhibitor of Fel d 1-mediated mast cell activation in vitro and in vivo. This inhibition required Fcgamma receptor-IIB. In human beings, IgGs of a single specificity were able to block degranulation of basophils from individuals with cat allergy. The inhibitory potential of these antibodies increased when larger allergen-IgG complexes were formed. CONCLUSIONS: These data reconcile conflicting theories in the literature and might explain the reason IgE levels do not necessarily decrease during therapy, despite clinical efficacy. These findings have important implications for vaccine design.


Asunto(s)
Alérgenos/inmunología , Anticuerpos Monoclonales/farmacología , Desensibilización Inmunológica , Glicoproteínas/inmunología , Hipersensibilidad/terapia , Inmunoglobulina G/farmacología , Alérgenos/farmacología , Animales , Anticuerpos Monoclonales/inmunología , Epítopos/inmunología , Glicoproteínas/antagonistas & inhibidores , Glicoproteínas/farmacología , Humanos , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Inmunoglobulina E/inmunología , Inmunoglobulina G/inmunología , Mastocitos/inmunología , Mastocitos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptores de IgE/genética , Receptores de IgE/inmunología , Receptores de IgG/genética , Receptores de IgG/inmunología , Vacunas/genética , Vacunas/inmunología
16.
Vaccine ; 28(33): 5503-12, 2010 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-20307591

RESUMEN

Pre-existing immunity against vaccine carrier proteins has been reported to inhibit the immune response against antigens conjugated to the same carrier by a process termed carrier induced epitopic suppression (CIES). Hence understanding the phenomenon of CIES is of major importance for the development of conjugate vaccines. Virus-like particles (VLPs) are a novel class of potent immunological carriers which have been successfully used to enhance the antibody response to virtually any conjugated antigen. In the present study we investigated the impact of a pre-existing VLP-specific immune response on the development of antibody responses against a conjugated model peptide after primary, secondary and tertiary immunization. Although VLP-specific immune responses led to reduced peptide-specific antibody titers, we showed that CIES against peptide-VLP conjugates could be overcome by high coupling densities, repeated injections and/or higher doses of conjugate vaccine. Furthermore we dissected VLP-specific immunity by adoptively transferring VLP-specific antibodies, B-cells or T(helper) cells separately into naïve mice and found that the observed CIES against peptide-VLP conjugates was mainly mediated by carrier-specific antibodies.


Asunto(s)
Allolevivirus/inmunología , Anticuerpos Antivirales/inmunología , Antígenos/inmunología , Proteínas de la Cápside/inmunología , Cápside/inmunología , Epítopos/inmunología , Tolerancia Inmunológica , Péptidos/inmunología , Allolevivirus/genética , Animales , Formación de Anticuerpos , Linfocitos B/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Linfocitos T Colaboradores-Inductores/inmunología , Virosomas
17.
PLoS One ; 5(3): e9809, 2010 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-20352110

RESUMEN

BACKGROUND: Recombinant proteins and in particular single domains or peptides are often poorly immunogenic unless conjugated to a carrier protein. Virus-like-particles are a very efficient means to confer high immunogenicity to antigens. We report here the development of virus-like-particles (VLPs) derived from the RNA bacteriophage AP205 for epitope-based vaccines. METHODOLOGY/PRINCIPAL FINDINGS: Peptides of angiotensin II, S.typhi outer membrane protein (D2), CXCR4 receptor, HIV1 Nef, gonadotropin releasing hormone (GnRH), Influenza A M2-protein were fused to either N- or C-terminus of AP205 coat protein. The A205-peptide fusions assembled into VLPs, and peptides displayed on the VLP were highly immunogenic in mice. GnRH fused to the C-terminus of AP205 induced a strong antibody response that inhibited GnRH function in vivo. Exposure of the M2-protein peptide at the N-terminus of AP205 resulted in a strong M2-specific antibody response upon immunization, protecting 100% of mice from a lethal influenza infection. CONCLUSIONS/SIGNIFICANCE: AP205 VLPs are therefore a very efficient and new vaccine system, suitable for complex and long epitopes, of up to at least 55 amino acid residues in length. AP205 VLPs confer a high immunogenicity to displayed epitopes, as shown by inhibition of endogenous GnRH and protective immunity against influenza infection.


Asunto(s)
Epítopos/química , Virión/química , Animales , Clonación Molecular , Hormona Liberadora de Gonadotropina/química , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Péptidos/química , Estructura Terciaria de Proteína , Fagos ARN/metabolismo , Proteínas Recombinantes/química , Vacunación/instrumentación , Vacunación/métodos
18.
Eur J Immunol ; 40(1): 103-12, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19877013

RESUMEN

Innate stimuli, such as TLR ligands, are known to greatly facilitate cross-priming. Currently it is unclear whether innate stimuli enhance cross-priming at the level of cross-presentation or at the level of T-cell priming. In this study, we addressed this question by measuring cross-presentation as well as cross-priming by virus-like particles (VLP) displaying peptide p33 derived of lymphocytic choriomeningitis virus. Innate stimuli were varied by either packaging different TLR ligands into virus-like particles or using mice deficient in two key molecules of TLR-signaling, namely the adaptor molecule MyD88 as well as IFN-alpha/beta receptor. While efficient cross-presentation occurred despite strongly reduced activation of DC in the absence of TLR ligand-mediated signals, T-cell priming was abolished. Thus, innate stimuli regulate cross-priming at the level of DC licensing for T-cell activation and not antigen presentation.


Asunto(s)
Reactividad Cruzada , Células Dendríticas/inmunología , Inmunidad Innata , Transducción de Señal , Animales , Presentación de Antígeno , Células Dendríticas/metabolismo , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , Proteínas Virales/inmunología , Virión/inmunología , Virión/ultraestructura
19.
J Immunol ; 184(1): 26-9, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19949081

RESUMEN

Dendritic cells (DCs) are the most important APC. It was recently reported that there is a dichotomy for Ag presentation by DC subsets; exogenous Ags reach the MHC class I pathway, but not the MHC class II pathway, in CD8(+) DCs, whereas CD8(-) DCs only process Ags for the MHC class II pathway. In this study, we used virus-like particles (VLPs) to show that CD8(+) and CD8(-) DCs efficiently capture and process VLPs for presentation in association with MHC class II in vivo. In contrast, CD8(+) DCs, but not CD8(-) DCs, cross presented VLP-derived peptides. This pattern was changed in an FcgammaR-dependent fashion in the presence of VLP-specific Abs, because under those conditions both DC subsets failed to efficiently cross present. Thus, the presentation of viral particles to CD4(+) T cells is not restricted to distinct DC subsets, whereas the presentation of viral particles to CD8(+) T cells is limited to CD8(+) DCs.


Asunto(s)
Presentación de Antígeno/inmunología , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Virión/inmunología , Animales , Antígenos Virales/inmunología , Antígenos CD8/inmunología , Reactividad Cruzada/inmunología , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL
20.
Virol J ; 6: 224, 2009 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-20025741

RESUMEN

Influenza virus infection is a prevalent disease in humans. Antibodies against hemagglutinin have been shown to prevent infection and hence hemagglutinin is the major constituent of current vaccines. Antibodies directed against the highly conserved extracellular domain of M2 have also been shown to mediate protection against Influenza A infection in various animal models. Active vaccination is generally considered the best approach to combat viral diseases. However, passive immunization is an attractive alternative, particularly in acutely exposed or immune compromized individuals, young children and the elderly. We recently described a novel method for the rapid isolation of natural human antibodies by mammalian cell display. Here we used this approach to isolate human monoclonal antibodies directed against the highly conserved extracellular domain of the Influenza A M2 protein. The identified antibodies bound M2 peptide with high affinities, recognized native cell-surface expressed M2 and protected mice from a lethal influenza virus challenge. Moreover, therapeutic treatment up to 2 days after infection was effective, suggesting that M2-specific monoclonals have a great potential as immunotherapeutic agents against Influenza infection.


Asunto(s)
Anticuerpos Monoclonales , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae , Proteínas de la Matriz Viral/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Especificidad de Anticuerpos , Línea Celular , Cricetinae , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunización Pasiva , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Resultado del Tratamiento , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA