Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 438
Filtrar
2.
J Am Chem Soc ; 142(28): 12133-12139, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32524819

RESUMEN

Synthetic immune-stimulatory drugs such as agonists of the Toll-like receptors (TLR) 7/8 are potent activators of antigen-presenting cells (APCs), however, they also induce severe side effects due to leakage from the site of injection into systemic circulation. Here, we report on the design and synthesis of an amphiphilic polymer-prodrug conjugate of an imidazoquinoline TLR7/8 agonist that in aqueous medium forms vesicular structures of 200 nm. The conjugate contains an endosomal enzyme-responsive linker enabling degradation of the vesicles and release of the TLR7/8 agonist in native form after endocytosis, which results in high in vitro TLR agonist activity. In a mouse model, locally administered vesicles provoke significantly more potent and long-lasting immune stimulation in terms of interferon expression at the injection site and in draining lymphoid tissue compared to a nonamphiphilic control and the native TLR agonist. Moreover, the vesicles induce robust activation of dendritic cells in the draining lymph node in vivo.


Asunto(s)
Imidazoles/farmacología , Glicoproteínas de Membrana/agonistas , Profármacos/farmacología , Quinolinas/farmacología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , beta-Galactosidasa/inmunología , Animales , Imidazoles/química , Imidazoles/metabolismo , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Glicoproteínas de Membrana/inmunología , Ratones , Estructura Molecular , Tamaño de la Partícula , Polietilenglicoles/química , Polietilenglicoles/metabolismo , Polietilenglicoles/farmacología , Profármacos/química , Profármacos/metabolismo , Quinolinas/química , Quinolinas/metabolismo , Propiedades de Superficie , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 8/inmunología , beta-Galactosidasa/química , beta-Galactosidasa/metabolismo
3.
Amino Acids ; 52(3): 411-424, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32008091

RESUMEN

We have previously shown that particle-mediated epidermal delivery (PMED) of plasmids encoding ß-galactosidase (ßGal) under control of the fascin-1 promoter (pFascin-ßGal) yielded selective production of the protein in skin dendritic cells (DCs), and suppressed Th2 responses in a mouse model of type I allergy by inducing Th1/Tc1 cells. However, intranasal challenge of mice immunized with pFascin-ßGal induced airway hyperreactivity (AHR) and neutrophilic inflammation in the lung. The tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase (IDO) has been implicated in immune suppression and tolerance induction. Here we investigated the consequences of co-application of an IDO-encoding vector on the modulatory effect of DNA vaccination by PMED using pFascin-ßGal in models of eosinophilic allergic and non-eosinophilic intrinsic airway inflammation. IDO-encoding plasmids and pFascin-ßGal or pCMV-ßGal were co-applied to abdominal skin of BALB/c mice without, before or after sensitization with ßGal protein. Immune responses in the lung were analysed after intranasal provocation and airway reactivity was determined by whole body plethysmography. Co-application of pCMV-IDO with pFascin-ßGal, but not pCMV-ßGal inhibited the Th1/Tc1 immune response after PMED. Moreover, AHR in those mice was attenuated following intranasal challenge. Therapeutic vaccination of ßGal-sensitized mice with pFascin-ßGal plus pCMV-IDO slightly suppressed airway inflammation and AHR after provocation with ßGal protein, while prophylactic vaccination was not effective. Altogether, our data suggest that only the combination of DC-restricted antigen and ubiquitous IDO expression attenuated asthma responses in mice, most probably by forming a tryptophan-depleted and kynurenine-enriched micromilieu known to affect neutrophils and T cells.


Asunto(s)
Asma/terapia , Biolística/métodos , Hiperreactividad Bronquial/terapia , Células Dendríticas/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Vacunación/métodos , Animales , Antígenos/genética , Antígenos/inmunología , Asma/inmunología , Hiperreactividad Bronquial/inmunología , Proteínas Portadoras/genética , Vectores Genéticos , Ratones , Ratones Endogámicos BALB C , Proteínas de Microfilamentos/genética , Células 3T3 NIH , Infiltración Neutrófila , Linfocitos T Citotóxicos/inmunología , Células TH1/inmunología , Transgenes , beta-Galactosidasa/genética , beta-Galactosidasa/inmunología
4.
Science ; 366(6467): 881-886, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31727837

RESUMEN

Myocarditis can develop into inflammatory cardiomyopathy through chronic stimulation of myosin heavy chain 6-specific T helper (TH)1 and TH17 cells. However, mechanisms governing the cardiotoxicity programming of heart-specific T cells have remained elusive. Using a mouse model of spontaneous autoimmune myocarditis, we show that progression of myocarditis to lethal heart disease depends on cardiac myosin-specific TH17 cells imprinted in the intestine by a commensal Bacteroides species peptide mimic. Both the successful prevention of lethal disease in mice by antibiotic therapy and the significantly elevated Bacteroides-specific CD4+ T cell and B cell responses observed in human myocarditis patients suggest that mimic peptides from commensal bacteria can promote inflammatory cardiomyopathy in genetically susceptible individuals. The ability to restrain cardiotoxic T cells through manipulation of the microbiome thereby transforms inflammatory cardiomyopathy into a targetable disease.


Asunto(s)
Enfermedades Autoinmunes/complicaciones , Bacteroides/inmunología , Cardiomiopatía Dilatada/inmunología , Cardiomiopatía Dilatada/microbiología , Microbioma Gastrointestinal/inmunología , Miocarditis/complicaciones , Péptidos/inmunología , beta-Galactosidasa/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Humanos , Intestinos/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Miocarditis/inmunología , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/inmunología , Células Th17/inmunología
5.
Artículo en Inglés | MEDLINE | ID: mdl-30838180

RESUMEN

The development of new effective vaccines strongly depends on adjuvants and formulations able to stimulate not only strong humoral responses against a certain pathogen but also effector as well as memory CD4+ and CD8+ T cells (Dubensky et al., 2013). However, the majority of vaccines licensed for human use or currently under clinical investigation fail to stimulate efficient cellular responses. For example, vaccines against hepatitis B virus (HBV), human papillomavirus (HPV), diphtheria, tetanus and influenza are usually administered by intramuscular (i.m.) injection and contain aluminum salts (alum) as adjuvant. Alum has been shown to stimulate Th2 immune cells resulting in increased production of antigen-specific antibodies but to be incapable of stimulating robust Th1 or cytotoxic responses. To overcome such limitations recent research has focused on the development of adjuvant combinations (e.g., MF59, AS03 or AS04) to not only further strengthen antigen-specific immune responses but to also allow their modulation. We have shown previously that bis-(3',5')-cyclic dimeric adenosine monophosphate (c-di-AMP) constitutes a promising adjuvant candidate stimulating both effective Th1/Th2 and cytotoxic immune responses when included in mucosal or parenteral vaccine formulations. In the present work we demonstrate that c-di-AMP can be also combined with other adjuvants like alum resulting in increases in not only humoral responses but more striking also in cellular immune responses. This leads to improved vaccine efficacy against intracellular pathogens.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Compuestos de Alumbre/administración & dosificación , Fosfatos de Dinucleósidos/administración & dosificación , Sinergismo Farmacológico , Inmunidad Celular , Inmunidad Humoral , Animales , Inyecciones Intramusculares , Ratones Endogámicos BALB C , Células TH1/inmunología , Células Th2/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , beta-Galactosidasa/administración & dosificación , beta-Galactosidasa/inmunología
6.
PLoS One ; 14(2): e0211117, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30726287

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is characterized by a fibrotic stroma with a poor lymphocyte infiltrate, in part driven by cancer-associated fibroblasts (CAFs). CAFs, which express fibroblast activation protein (FAP), contribute to immune escape via exclusion of anti-tumor CD8+ T cells from cancer cells, upregulation of immune checkpoint ligand expression, immunosuppressive cytokine production, and polarization of tumor infiltrating inflammatory cells. FAP is a post-proline peptidase selectively expressed during tissue remodeling and repair, such as with wound healing, and in the tumor microenvironment by cancer-associated fibroblasts. We targeted FAP function using a novel small molecule inhibitor, UAMC-1110, and mice with germline knockout of FAP and concomitant knock-in of E. coli beta-galactosidase. We depleted CAFs by adoptive transfer of anti-ßgal T cells into the FAP knockout animals. Established syngeneic pancreatic tumors in immune competent mice were targeted with these 3 strategies, followed by focal radiotherapy to the tumor. FAP loss was associated with improved antigen-specific tumor T cell infiltrate and enhanced collagen deposition. However, FAP targeting alone or with tumor-directed radiation did not improve survival even when combined with anti-PD1 therapy. Targeting of CAFs alone or in combination with radiation did not improve survival. We conclude that targeting FAP and CAFs in combination with radiation is capable of enhancing anti-tumor T cell infiltrate and function, but does not result in sufficient tumor clearance to extend survival.


Asunto(s)
Anticuerpos/metabolismo , Carcinoma Ductal Pancreático/terapia , Gelatinasas/antagonistas & inhibidores , Proteínas de la Membrana/antagonistas & inhibidores , Neoplasias Pancreáticas/terapia , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Linfocitos T/trasplante , Traslado Adoptivo , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Quimioradioterapia , Terapia Combinada , Endopeptidasas , Gelatinasas/genética , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Humanos , Proteínas de la Membrana/genética , Ratones , Neoplasias Pancreáticas/metabolismo , Serina Endopeptidasas/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Linfocitos T/inmunología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto , beta-Galactosidasa/inmunología
7.
Fish Shellfish Immunol ; 80: 458-466, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29859312

RESUMEN

The aim of this study was the assessment of preloaded feed pellets as a delivery system for plasmid DNA (pDNA), with the purpose of evaluating the potential administration of DNA vaccines orally in aquacultured fish. Pellets were made up by usual feed ingredients, which were mixed with chitosan nanoparticles entrapping a model plasmid (pCMVß) expressible in eukaryotic cells before being elaborated. The plasmid is characterized by the insertion of the reporter gene lacZ, encoding for the bacterial enzyme ß-galactosidase (ß-gal). The possible in vivo expression of the exogenous gene was measured in different fish tissues of gilthead sea bream (Sparus aurata) juveniles by two different procedures. On the one hand, the activity of the enzyme ß-gal was detected and quantified in muscle, liver and intestine; on the other, specific IgM against ß-gal antigen was titrated in blood samples. Intramuscular (i.m.) injection of equal amounts of plasmid was also carried out for the purpose of comparison with oral administration. The expression of the reporter gene was detected in fish tissues following both oral and i. m. administration of pDNA up to 60 days. However, organ distribution of the gene expression was more evident after oral (ß-gal activity measured in gut, liver and muscle) than after parenteral administration (restricted to adjacent muscle tissues). In agreement, specific IgM titration indicated that humoral immune response was more intense and sustained throughout the experimental period after oral than after i. m. delivery of equal amounts of pDNA. These results suggest that feed pellets containing chitosan nanoparticles might enable efficient oral delivery of pDNA, a fact that might imply valuable applications in terms of on-farm mass immunization purposes, especially with regard to DNA-based vaccines and small size fish, in which i. m. administration remains unfeasible.


Asunto(s)
Quitosano/administración & dosificación , ADN/administración & dosificación , Nanopartículas/administración & dosificación , Dorada/inmunología , Vacunas de ADN , Administración Oral , Alimentación Animal , Animales , Técnicas de Transferencia de Gen , Genes Reporteros , Inmunoglobulina M/sangre , Intestinos/enzimología , Hígado/enzimología , Músculos/enzimología , Plásmidos , Dorada/metabolismo , beta-Galactosidasa/inmunología , beta-Galactosidasa/metabolismo
8.
Cell Death Dis ; 8(12): 3217, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29242529

RESUMEN

Aberrant DNA methylation observed in cancer can provide survival benefits to cells by silencing genes essential for anti-tumor activity. DNA-demethylating agents such as Decitabine (DAC)/Azacitidine (AZA) activate otherwise silenced tumor suppressor genes, alter immune response and epigenetically reprogram tumor cells. In this study, we show that non-cytotoxic nanomolar DAC concentrations modify the bladder cancer transcriptome to activate NOTCH1 at the mRNA and protein level, increase double-stranded RNA sensors and CK5-dependent differentiation. Importantly, DAC treatment increases ICN1 expression (the active intracellular domain of NOTCH1) significantly inhibiting cell proliferation and causing changes in cell size inducing morphological alterations reminiscent of senescence. These changes were not associated with ß-galactosidase activity or increased p16 levels, but instead were associated with substantial IL-6 release. Increased IL-6 release was observed in both DAC-treated and ICN1 overexpressing cells as compared to control cells. Exogenous IL-6 expression was associated with a similar enlarged cell morphology that was rescued by the addition of a monoclonal antibody against IL-6. Treatment with DAC, overexpression with ICN1 or addition of exogenous IL-6 showed CK5 reduction, a surrogate marker of differentiation. Overall this study suggests that in MIBC cells, DNA hypomethylation increases NOTCH1 expression and IL-6 release to induce CK5-related differentiation.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Azacitidina/farmacología , Regulación Neoplásica de la Expresión Génica , Interleucina-6/genética , Receptor Notch1/genética , Neoplasias de la Vejiga Urinaria/genética , Anticuerpos Monoclonales/farmacología , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/inmunología , Decitabina , Epigénesis Genética , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/inmunología , Queratina-5/genética , Queratina-5/metabolismo , Músculo Liso/inmunología , Músculo Liso/patología , Invasividad Neoplásica , Receptor Notch1/metabolismo , Transducción de Señal , Resultado del Tratamiento , Neoplasias de la Vejiga Urinaria/diagnóstico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/patología , beta-Galactosidasa/genética , beta-Galactosidasa/inmunología
9.
Gene Ther ; 24(2): 113-119, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28004656

RESUMEN

A significant problem affecting gene therapy approaches aiming at achieving long-term transgene expression is the immune response against the protein product of the therapeutic gene, which can reduce or eliminate the therapeutic effect. The problem is further exacerbated when therapy involves targeting an immunogenic tissue and/or one with a pre-existing inflammatory phenotype, such as dystrophic muscles. In this proof-of-principle study, we co-expressed a model antigen, bacterial ß-galactosidase, with an immunosuppressive factor, indoleamine 2,3-dioxygenase 1 (IDO1), in muscles of the mdx mouse model of Duchenne muscular dystrophy. This treatment prevented loss of expression of the transgene concomitant with significantly elevated expression of T-regulatory (Treg) markers in the IDO1-expressing muscles. Moreover, co-expression of IDO1 resulted in reduced serum levels of anti-ß-gal antibodies. These data indicate that co-expression of genes encoding immunomodulatory enzymes controlling kynurenine pathways provide a viable strategy for preventing loss of transgenes targeted into dystrophic muscles with pre-existing inflammation.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Músculo Esquelético/inmunología , Distrofia Muscular Animal/inmunología , Linfocitos T Reguladores/inmunología , Transgenes/fisiología , beta-Galactosidasa/metabolismo , Animales , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Indolamina-Pirrol 2,3,-Dioxigenasa/administración & dosificación , Quinurenina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo , beta-Galactosidasa/inmunología
10.
Oncotarget ; 8(4): 6857-6872, 2017 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-28036287

RESUMEN

N-glycosylation is generally accepted to enhance the immunogenicity of antigens because of two main reasons. First, the attachment of glycans enables recognition by endocytic receptors like the mannose receptor (MR) and hence increased uptake by dendritic cells (DCs). Second, foreign glycans are postulated to be immunostimulatory and their recognition could induce DC activation. However, a direct comparison between the immunogenicity of N-glycosylated vs. de-glycosylated proteins in vivo and a direct effect of N-glycosylated antigens on the intrinsic capacity of DCs to activate T cells have not been assessed so far.To analyze whether enforced N-glycosylation is a suited strategy to enhance the immunogenicity of non-glycosylated antigens for vaccination studies, we targeted non-glycoproteins towards the MR by introduction of artificial N-glycosylation using the methylotrophic yeast Komagataella phaffii (previously termed Pichia pastoris). We could demonstrate that the introduction of a single N-X-S/T motif was sufficient for efficient MR-binding and internalization. However, addition of N-glycosylated proteins neither influenced DC maturation nor their general capacity to activate T cells, pointing out that enforced N-glycosylation does not increase the immunogenicity of the antigen per se. Additionally, increased antigen-specific cytotoxic T cell responses in vivo after injection of N-glycosylated compared to de-glycosylated proteins were observed but this effect strongly depended on the epitope tested. A beneficial effect of N-glycosylation on antibody production could not be detected, which might be due to MR-cross-linking on DCs and to concomitant differences in IL-6 production by CD4+ T cells.These observations point out that the effect of N-glycosylation on antigen immunogenicity can vary between different antigens and therefore might have important implications for the development of vaccines using K. phaffii.


Asunto(s)
Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Activación de Linfocitos , Lectinas de Unión a Manosa/metabolismo , Procesamiento Proteico-Postraduccional , Receptores de Superficie Celular/metabolismo , Linfocitos T/metabolismo , beta-Galactosidasa/metabolismo , Animales , Comunicación Celular , Proliferación Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Epítopos , Glicosilación , Células HEK293 , Humanos , Inmunogenicidad Vacunal , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Ligandos , Receptor de Manosa , Lectinas de Unión a Manosa/deficiencia , Lectinas de Unión a Manosa/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/inmunología , Ovalbúmina/metabolismo , Pichia/genética , Pichia/metabolismo , Dominios y Motivos de Interacción de Proteínas , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Linfocitos T/inmunología , Factores de Tiempo , Transfección , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/metabolismo , beta-Galactosidasa/genética , beta-Galactosidasa/inmunología
11.
PLoS One ; 10(12): e0144738, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26658766

RESUMEN

We report a rapid, higher throughput method for measuring neutralizing antibody to herpes simplex virus (HSV) in human sera. Clinical isolates and sera from the Herpevac Trial for Women were used in a colorimetric assay in which infection of tissue culture (lack of neutralization) was indicated by substrate metabolism by beta-galactosidase induced in the ELVIS cell line. The neutralization assay was optimized by addition of guinea pig complement, which particularly enhanced neutralizing antibody titers to HSV-2. Higher neutralizing antibody titers were also achieved using virus particles isolated from the supernatant of infected cells rather than lysate of infected cells as the source of virus. The effect of assay incubation time and incubation time with substrate were also optimized. We found that incubating with substrate until a standard optical density of 1.0 was reached permitted a better comparison among virus isolates, and achieved reliable measurement of neutralizing antibody activity. Interestingly, in contrast to results in the absence of complement, addition of complement allowed sera from HSV-2 gD-vaccinated subjects to neutralize HSV-1 and HSV-2 clinical and laboratory isolates with equal potency.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Herpes Genital/inmunología , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 2/inmunología , Ensayos Analíticos de Alto Rendimiento , Animales , Línea Celular , Chlorocebus aethiops , Proteínas del Sistema Complemento/química , Cricetulus , Femenino , Genes Reporteros , Cobayas , Herpes Genital/diagnóstico , Herpes Genital/virología , Herpes Simple/diagnóstico , Herpes Simple/virología , Herpesvirus Humano 1/crecimiento & desarrollo , Herpesvirus Humano 2/crecimiento & desarrollo , Humanos , Sueros Inmunes/química , Operón Lac , Pruebas de Neutralización , Regiones Promotoras Genéticas , Células Vero , Virión/inmunología , beta-Galactosidasa/genética , beta-Galactosidasa/inmunología
12.
Hum Vaccin Immunother ; 11(12): 2913-22, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26697975

RESUMEN

Delivering vaccine antigens to mucosal surfaces is potentially very attractive, especially as protection from mucosal infections may be mediated by local immune responses. However, to date mucosal immunization has had limited successes, with issues of both safety and poor immunogenicity. One approach to improve immunogenicity is to develop adjuvants that are effective and safe at mucosal surfaces. Differences in immune responses between mice and men have overstated the value of some experimental adjuvants which have subsequently performed poorly in the clinic. Due to their closer similarity, non-human primates can provide a more accurate picture of adjuvant performance. In this study we immunised rhesus macaques (Macaca mulatta) using a unique matrix experimental design that maximised the number of adjuvants screened while reducing the animal usage. Macaques were immunised by the intranasal, sublingual and intrarectal routes with the model protein antigens keyhole limpet haemocyanin (KLH), ß-galactosidase (ß-Gal) and ovalbumin (OVA) in combination with the experimental adjuvants Poly(I:C), Pam3CSK4, chitosan, Thymic Stromal Lymphopoietin (TSLP), MPLA and R848 (Resiquimod). Of the routes used, only intranasal immunization with KLH and R848 induced a detectable antibody response. When compared to intramuscular immunization, intranasal administration gave slightly lower levels of antigen specific antibody in the plasma, but enhanced local responses. Following intranasal delivery of R848, we observed a mildly inflammatory response, but no difference to the control. From this we conclude that R848 is able to boost antibody responses to mucosally delivered antigen, without causing excess local inflammation.


Asunto(s)
Anticuerpos/sangre , Anticuerpos/inmunología , Formación de Anticuerpos/inmunología , Inmunidad Mucosa/inmunología , Inmunización/métodos , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Adyuvantes Inmunológicos , Administración Intranasal , Animales , Quitosano/inmunología , Citocinas/inmunología , Femenino , Hemocianinas/inmunología , Imidazoles/inmunología , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Inflamación/inmunología , Inyecciones Intramusculares , Lipopéptidos/inmunología , Macaca mulatta , Membrana Mucosa/inmunología , Ovalbúmina/inmunología , Poli I-C/inmunología , beta-Galactosidasa/inmunología , Linfopoyetina del Estroma Tímico
13.
PLoS One ; 10(6): e0128722, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26030383

RESUMEN

The skin accommodates multiple dendritic cell (DC) subsets with remarkable functional diversity. Immune reactions are initiated and modulated by the triggering of DC by pathogen-associated or endogenous danger signals. In contrast to these processes, the influence of intrinsic features of protein antigens on the strength and type of immune responses is much less understood. Therefore, we investigated the involvement of distinct DC subsets in immune reactions against two structurally different model antigens, E. coli beta-galactosidase (betaGal) and chicken ovalbumin (OVA) under otherwise identical conditions. After epicutaneous administration of the respective DNA vaccines with a gene gun, wild type mice induced robust immune responses against both antigens. However, ablation of langerin+ DC almost abolished IgG1 and cytotoxic T lymphocytes against betaGal but enhanced T cell and antibody responses against OVA. We identified epidermal Langerhans cells (LC) as the subset responsible for the suppression of anti-OVA reactions and found regulatory T cells critically involved in this process. In contrast, reactions against betaGal were not affected by the selective elimination of LC, indicating that this antigen required a different langerin+ DC subset. The opposing findings obtained with OVA and betaGal vaccines were not due to immune-modulating activities of either the plasmid DNA or the antigen gene products, nor did the differential cellular localization, size or dose of the two proteins account for the opposite effects. Thus, skin-borne protein antigens may be differentially handled by distinct DC subsets, and, in this way, intrinsic features of the antigen can participate in immune modulation.


Asunto(s)
Células Dendríticas/inmunología , Piel/inmunología , Vacunas de ADN/inmunología , Animales , Antígenos de Superficie/inmunología , Biolística/métodos , Pollos , Escherichia coli/inmunología , Inmunoglobulina G/inmunología , Células de Langerhans/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Linfocitos T Citotóxicos/inmunología , beta-Galactosidasa/inmunología
14.
Scand J Immunol ; 82(4): 320-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26095954

RESUMEN

DNA-mediated immunization of a tumour antigen is a possible immunotherapy for cancer, and interleukin (IL)-27 has diverse functions in adaptive immunity. In this study, we examined whether IL-27 DNA administration enhanced antitumour effects in mice vaccinated with DNA encoding a putative tumour antigen, ß-galactosidase (ß-gal). An intramuscular injection of cardiotoxin before DNA administration facilitated the exogenous gene expression. In mice received ß-gal and IL-27 DNA, growth of ß-gal-positive P815 tumours was retarded and survival of the mice was prolonged. Development of ß-gal-positive Colon 26 tumours was suppressed by vaccination of ß-gal DNA and further inhibited by additional IL-27 DNA administration or IL-12 family cytokines. Nevertheless, a population of ß-gal-specific CD8(+) T cells did not increase, and production of anti-ß-gal antibody was not enhanced by IL-27 DNA administration. Spleen cells from mice bearing IL-27-expressing Colon 26 tumours showed greater YAC-1-targeted cytotoxicity although CD3(-)/DX5(+) natural killer (NK) cell numbers remained unchanged. Recombinant IL-27 enhanced YAC-1-targeted cytotoxicity of IL-2-primed splenic NK cells and augmented a phosphorylation of signal transducer and activator of transcription 3 and an expression of perforin. These data collectively indicate that IL-27 DNA administration activates NK cells and augments vaccination effects of DNA encoding a tumour antigen through non-adaptive immune responses.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/uso terapéutico , ADN/uso terapéutico , Interleucina-27/genética , Neoplasias/terapia , Vacunas de ADN/uso terapéutico , beta-Galactosidasa/inmunología , Animales , Anticuerpos/inmunología , Antígenos de Neoplasias/genética , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/genética , Cardiotoxinas/administración & dosificación , ADN/administración & dosificación , ADN/genética , Interleucina-12/genética , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Perforina/biosíntesis , Fosforilación , Factor de Transcripción STAT3/metabolismo , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , beta-Galactosidasa/genética
15.
Nat Commun ; 6: 6771, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25857745

RESUMEN

Lymphatic endothelial cells (LECs) directly express peripheral tissue antigens and induce CD8 T-cell deletional tolerance. LECs express MHC-II molecules, suggesting they might also tolerize CD4 T cells. We demonstrate that when ß-galactosidase (ß-gal) is expressed in LECs, ß-gal-specific CD8 T cells undergo deletion via the PD-1/PD-L1 and LAG-3/MHC-II pathways. In contrast, LECs do not present endogenous ß-gal in the context of MHC-II molecules to ß-gal-specific CD4 T cells. Lack of presentation is independent of antigen localization, as membrane-bound haemagglutinin and I-Eα are also not presented by MHC-II molecules. LECs express invariant chain and cathepsin L, but not H2-M, suggesting that they cannot load endogenous antigenic peptides onto MHC-II molecules. Importantly, LECs transfer ß-gal to dendritic cells, which subsequently present it to induce CD4 T-cell anergy. Therefore, LECs serve as an antigen reservoir for CD4 T-cell tolerance, and MHC-II molecules on LECs are used to induce CD8 T-cell tolerance via LAG-3.


Asunto(s)
Presentación de Antígeno/genética , Antígenos CD/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Células Endoteliales/inmunología , Tolerancia Periférica/genética , Animales , Antígenos/genética , Antígenos/inmunología , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Diferenciación de Linfocitos B/inmunología , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Catepsina L/genética , Catepsina L/inmunología , Anergia Clonal/genética , Células Dendríticas/citología , Células Endoteliales/citología , Expresión Génica , Hemaglutininas/genética , Hemaglutininas/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Cultivo Primario de Células , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Transducción de Señal , beta-Galactosidasa/genética , beta-Galactosidasa/inmunología , Proteína del Gen 3 de Activación de Linfocitos
16.
Vaccine ; 33(18): 2118-24, 2015 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-25804706

RESUMEN

BACKGROUND: Vaccine strategies represent one of the fighting answers against multiresistant bacteria in a number of clinical settings like cystic fibrosis (CF). Mycobacterium abscessus, an emerging CF pathogen, raises difficult therapeutic problems due to its intrinsic antibiotic multiresistance. METHODS: By reverse vaccinology, we identified M. abscessus phospholipase C (MA-PLC) as a potential vaccine target. We deciphered here the protective response generated by vaccination with plasmid DNA encoding the MA-PLC formulated with a tetra functional block copolymer 704, in CF (ΔF508) mice. Protection was tested against aerosolized smooth and rough (hypervirulent) variants of M. abscessus. RESULTS: MA-PLC DNA vaccination (days 0, 21, 42) elicited a strong antibody response. A significant protective effect was obtained against aerosolized M. abscessus (S variant) in ΔF508 mice, but not in wild-type FVB littermates; similar results were observed when: (i) challenging mice with the "hypervirulent" R variant, and; (ii) immunizing mice with purified MA-PLC protein. High IgG titers against MA-PLC protein were measured in CF patients with M. abscessus infection; interestingly, significant titers were also detected in CF patients positive for Pseudomonas aeruginosa versus P. aeruginosa-negative controls. CONCLUSIONS: MA-PLC DNA- and PLC protein-vaccinated mice cleared more rapidly M. abscessus than ß-galactosidase DNA- or PBS- vaccinated mice in the context of CF. PLCs could constitute interesting vaccine targets against common PLC-producing CF pathogens like P. aeruginosa.


Asunto(s)
Vacunas Bacterianas/inmunología , Fibrosis Quística/complicaciones , Infecciones por Mycobacterium no Tuberculosas/prevención & control , Micobacterias no Tuberculosas/inmunología , Fosfolipasas de Tipo C/genética , Fosfolipasas de Tipo C/inmunología , Vacunas de ADN/inmunología , Animales , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Antibacterianos/sangre , Vacunas Bacterianas/administración & dosificación , Fibrosis Quística/inmunología , Fibrosis Quística/microbiología , Femenino , Humanos , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Masculino , Ratones , Infecciones por Mycobacterium no Tuberculosas/complicaciones , Micobacterias no Tuberculosas/enzimología , Infecciones por Pseudomonas/complicaciones , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Fosfolipasas de Tipo C/administración & dosificación , Vacunas de ADN/administración & dosificación , beta-Galactosidasa/genética , beta-Galactosidasa/inmunología
17.
Eur J Immunol ; 44(11): 3240-51, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25103845

RESUMEN

Paraneoplastic neurologic diseases (PND) involving immune responses directed toward intracellular antigens are poorly understood. Here, we examine immunity to the PND antigen Nova2, which is expressed exclusively in central nervous system (CNS) neurons. We hypothesized that ectopic expression of neuronal antigen in the periphery could incite PND. In our C57BL/6 mouse model, CNS antigen expression limits antigen-specific CD4+ and CD8+ T-cell expansion. Chimera experiments demonstrate that this tolerance is mediated by antigen expression in nonhematopoietic cells. CNS antigen expression does not limit tumor rejection by adoptively transferred transgenic T cells but does limit the generation of a memory population that can be expanded upon secondary challenge in vivo. Despite mediating cancer rejection, adoptively transferred transgenic T cells do not lead to paraneoplastic neuronal targeting. Preliminary experiments suggest an additional requirement for humoral activation to induce CNS autoimmunity. This work provides evidence that the requirements for cancer immunity and neuronal autoimmunity are uncoupled. Since humoral immunity was not required for tumor rejection, B-cell targeting therapy, such as rituximab, may be a rational treatment option for PND that does not hamper tumor immunity.


Asunto(s)
Autoinmunidad , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Sistema Nervioso Central/inmunología , Síndromes Paraneoplásicos del Sistema Nervioso/inmunología , Traslado Adoptivo , Animales , Anticuerpos Monoclonales de Origen Murino/farmacología , Antígenos de Neoplasias/inmunología , Antineoplásicos/farmacología , Linfocitos B/inmunología , Sistema Nervioso Central/citología , Tolerancia Inmunológica , Inmunización , Factores Inmunológicos/farmacología , Memoria Inmunológica , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Antígeno Ventral Neuro-Oncológico , Neuronas/inmunología , Proteínas de Unión al ARN/inmunología , Rituximab , beta-Galactosidasa/genética , beta-Galactosidasa/inmunología
18.
PLoS One ; 9(7): e102280, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25025197

RESUMEN

The type of immune response is critical for successful protection and typically determined by pathogen-associated danger molecules. In contrast, protein antigens are usually regarded as passive target structures. Here, we provide evidence that the structure of the antigen can profoundly influence the type of response that is elicited under else identical conditions. In mice, gene gun vaccines induce predominantly Th2-biased immune reactions against most antigens. One exception is E. coli beta-galactosidase (ßGal) that induces a balanced Th1/Th2 response. Because both, the delivered material (plasmid DNA-coated gold particles) as well as the procedure (biolistic delivery to the skin surface) is the same as for other antigens we hypothesized that Th1 induction could be a function of ßGal protein expressed in transfected cells. To test this we examined gene gun vaccines encoding structural or functional variants of the antigen. Employing a series of gene gun vaccines encoding individual structural domains of ßGal, we found that neither of them induced IgG2a antibodies. Even disruption of the homo-tetramer association of the native protein by deletion of a few N-terminal amino acids was sufficient to abrogate IgG2a production. However, enzymatically inactive ßGal with only one point mutation in the catalytic center retained the ability to induce Th1 reactions. Thus, structural but not functional integrity of the antigen must be retained for Th1 induction. ßGal is not a Th1 adjuvant in the classical sense because neither were ßGal-transgenic ROSA26 mice particularly Th1-biased nor did co-administration of a ßGal-encoding plasmid induce IgG2a against other antigens. Despite this, gene gun vaccines elicited Th1 reactions to antigens fused to the open reading frame of ßGal. We interpret these findings as evidence that different skin-borne antigens may be differentially handled by the immune system and that the three-dimensional structure of an antigen is an important determinant for this.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de Escherichia coli/inmunología , Células TH1/inmunología , Vacunas de ADN/inmunología , beta-Galactosidasa/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/genética , Biolística/métodos , Línea Celular , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Femenino , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Ratones , Dominios y Motivos de Interacción de Proteínas/inmunología , Multimerización de Proteína/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Vacunas de ADN/administración & dosificación , beta-Galactosidasa/química , beta-Galactosidasa/genética
19.
Pharm Res ; 31(2): 516-26, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24072262

RESUMEN

PURPOSE: Antigen-Expressing Immunostimulatory Liposomes (AnExILs) represent a novel DNA vaccination platform based on the production of protein antigens from DNA templates inside liposomes mediated by an in vitro transcription and translation (IVTT) mix. The aim of this study was to analyze the effects of AnExILs on different dendritic cells (DCs) models and to better understand the role of the different components of this formulation on its adjuvanticity. METHODS: The effect of ß-galactosidase-expressing AnExILs on maturation and particle uptake by murine DC cell line, fresh human monocyte-derived DCs or human dermal DCs in skin explants was investigated and compared to the effects of either plain liposomes or IVTT mix alone. RESULTS: AnExILs induced efficient DC chemotaxis and promoted up-regulation of maturation markers on murine DCs, due to the presence of IVTT in the formulation. Furthermore, the amount of active ßGal associated with DCs was higher for AnExILs than for free ßGal expressed in IVTT or ßGal encapsulated into non-adjuvanted liposomes. Most interestingly, the same trend was observed with human DCs. CONCLUSIONS: Both IVTT mix and liposomal vehicles were shown to be key components of the AnExIL formulation responsible for its adjuvanticity. AnExILs combine antigen production, adjuvanticity and delivery in one system, and can efficiently activate both murine and human DCs.


Asunto(s)
Antígenos/inmunología , Quimiotaxis/inmunología , Células Dendríticas/inmunología , Liposomas/inmunología , Piel/inmunología , Animales , Antígenos/genética , Línea Celular , Química Farmacéutica/métodos , Quimiotaxis/genética , Células Dendríticas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Humanos , Inmunización , Liposomas/metabolismo , Ratones , Monocitos/inmunología , Monocitos/metabolismo , Piel/metabolismo , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , beta-Galactosidasa/genética , beta-Galactosidasa/inmunología
20.
Microbiol Immunol ; 57(11): 778-84, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24033444

RESUMEN

The present investigation aimed at identifying the abilities of three different species of probiotic lactobacilli to modulate cellular immune responses in mouse neutrophils and macrophages in vivo over a study period of 60 days. Neutrophil respiratory burst enzymes (cytochrome c reductase and MPO) showed remarkable increased activity (P ≤ 0.01) after consumption of milks fermented by different species of probiotics over 30 and 60 days of feeding trials. Enzyme activities (ß-galactosidase and ß-glucuronidase) and nitric oxide production also increased considerably (P ≤ 0.01) in macrophages, both in peritoneal fluid and in enriched cell cultures. The effects of enhanced enzyme activities were corroborated by simultaneous increases in the phagocytic activities of neutrophils and macrophages. The increases in cellular functions were invariably maximal during the first 30 days of study and were maintained, but did not increase, over the next 30 days. Further, Lactobacillus helveticus-fed groups were most effective at modulating neutrophil functions whereas Lactobacillus paracasei-fed groups were more potent at enhancing macrophage functions. Together, our results indicate that probiotics have strain specific effects on stimulating cellular functions while not causing excessive stimulation of the immune system over longer feeding periods, thereby resulting in maximum and stable health benefits.


Asunto(s)
Inmunidad Innata/efectos de los fármacos , Lactobacillus/metabolismo , Leche/microbiología , Probióticos/administración & dosificación , Animales , Células Cultivadas , Fermentación , Lactobacillus/química , Lactobacillus/clasificación , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Factores de Tiempo , beta-Galactosidasa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...