Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Respir Cell Mol Biol ; 64(1): 39-49, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32970964

RESUMEN

Aspergillus fumigatus is an opportunistic fungal pathogen responsible for a spectrum of clinical manifestations. Dendritic cells recognize pathogen-associated molecular patterns of Aspergillus via two main receptor families, Toll-like receptors (TLRs) and C-type lectin receptors (CLR). Here, the importance of TLR and CLR signaling in the regulation of T-helper cell type 2 (Th2) responses was analyzed using a mouse model based on the transfer of bone marrow-derived dendritic cells (BMDCs) pulsed with A. fumigatus conidia. BMDCs were generated from mice deficient in either MyD88 or MALT1 (mucosa-associated lymphoid tissue lymphoma translocation protein 1). Both the MyD88 and MALT1 signaling pathway in BMDCs contributed to the production of inflammatory cytokines induced by A. fumigatus conidia. Mice sensitized with MyD88-/- BMDCs pulsed in vitro with A. fumigatus conidia showed an exacerbated allergic inflammation, with stronger eosinophil recruitment in the BAL and higher Th2 cytokine production compared with mice sensitized with wild-type or MALT1-/- BMDCs. This exacerbation was not observed when MyD88-/- BMDCs were pulsed with Cladosporium sphaerospermum, a nonpathogenic mold. A lack of TLR2 signaling recapitulated the exacerbation of the A. fumigatus Th2 response observed in the absence of MyD88 signaling, whereas TLR2 agonist dampened the response induced with A. fumigatus and C. sphaerospermum conidia. IL-10 production by BMDCs in response to A. fumigatus was dependent on the expression of TLR2 and MyD88. IL-10-/- BMDCs exacerbated, whereas MyD88-/- BMDCs supplemented with exogenous IL-10 decreased the allergic pulmonary inflammation. These results indicate that TLR2/MyD88-specific recognition of PAMPs from A. fumigatus conidia can upregulate IL-10 production and downregulate lung eosinophilia and the development of a Th2 response.


Asunto(s)
Aspergillus fumigatus/inmunología , Células Dendríticas/inmunología , Inflamación/inmunología , Pulmón/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 2/inmunología , Animales , Aspergilosis/inmunología , Asma/inmunología , Células Cultivadas , Cladosporium/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Eosinófilos/inmunología , Lectinas Tipo C/inmunología , Ratones , Ratones Endogámicos C57BL , Eosinofilia Pulmonar/inmunología , Células Th2/inmunología
2.
Nat Commun ; 9(1): 3417, 2018 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-30143632

RESUMEN

Cancer immunotherapy can induce durable antitumor responses. However, many patients poorly respond to such therapies. Here we describe a generic antitumor therapy that is based on the intratumor delivery of mRNA that codes for the necroptosis executioner mixed lineage kinase domain-like (MLKL) protein. This intervention stalls primary tumor growth and protects against distal and disseminated tumor formation in syngeneic mouse melanoma and colon carcinoma models. Moreover, MLKL-mRNA treatment combined with immune checkpoint blockade further improves the antitumor activity. MLKL-mRNA treatment rapidly induces T cell responses directed against tumor neo-antigens and requires CD4+ and CD8+ T cells to prevent tumor growth. Type I interferon signaling and Batf3-dependent dendritic cells are essential for this mRNA treatment to elicit tumor antigen-specific T cell responses. Moreover, MLKL-mRNA treatment blunts the growth of human lymphoma in mice with a reconstituted human adaptive immune system. MLKL-based treatment can thus be exploited as an effective antitumor immunotherapy.


Asunto(s)
Inmunoterapia/métodos , Necrosis/metabolismo , Neoplasias/terapia , Proteínas Quinasas/genética , ARN Mensajero/genética , Animales , Apoptosis/genética , Apoptosis/fisiología , Línea Celular , Femenino , Humanos , Linfoma/inmunología , Linfoma/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Necrosis/genética , Neoplasias/genética , Modelos de Riesgos Proporcionales , Transducción de Señal/genética , Transducción de Señal/fisiología
3.
J Control Release ; 284: 73-83, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-29908893

RESUMEN

Potent adjuvants are highly demanded for most protein and peptides based vaccine candidates in clinical development. Recognition of viral single stranded (ss)RNA by innate toll-like receptors 7/8 in dendritic cells results in a cytokine environment supportive to the establishment of long lasting antibody responses and Th1 oriented T cell immunity. To fully exploit the immunestimulatory properties of ssRNA, it needs to be adequately formulated to ensure its optimal delivery to dendritic cells in the vaccine draining lymph nodes. In the present paper, we report on the design of ssRNA nanocomplexes formed by complexation of the cationic poly(carbonic acid 2-dimethylamino-ethyl ester 1-methyl-2-(2-methacryloylamino)-ethyl ester) (pHPMA-DMAE) based polymeric carrier and ssRNA. The resulting ssRNA nanocomplexes were subsequently PEGylated through copper-free click chemistry using PEG-bicyclo[6.1.0]nonyne (PEG-BCN) and cross-linked via disulfide bonds to increase their stability. The obtained near-neutral charged PEGylated ssRNA nanocomplexes (~150 nm) combined ssRNA protection with highly efficient delivery of ssRNA to DCs in the vaccine draining lymph nodes after subcutanuously administration. When co-administrated with a model antigen (soluble ovalbumin (OVA)), ssRNA nanocomplexes were far more efficient at inducing CD8 cytolytic T cells when compared to OVA co-adminstarted with naked ssRNA. Furthermore, IgG2c antibody titers, indicative of Th1 skewed T cell responses, were >10 times increased by complexing ssRNA into the PEGylated nanocomplexes. This study highlights the potential of post-functionalizing ssRNA nanocomplexes by copper-free click chemistry and these findings indcate that this potent ssRNA adjuvant may profoundly improve the efficacy of a variety of vaccines requiring Th1-type immunity.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Portadores de Fármacos/química , Polietilenglicoles/química , ARN/administración & dosificación , Linfocitos T Citotóxicos/efectos de los fármacos , Adyuvantes Inmunológicos/farmacología , Animales , Formación de Anticuerpos/efectos de los fármacos , Reactivos de Enlaces Cruzados/química , Células Dendríticas/efectos de los fármacos , Femenino , Ganglios Linfáticos/efectos de los fármacos , Ratones Endogámicos C57BL , Ovalbúmina/administración & dosificación , Ovalbúmina/farmacología , ARN/farmacología
4.
Adv Healthc Mater ; 6(13)2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28436620

RESUMEN

To date, the mRNA delivery field has been heavily dominated by lipid-based systems. Reports on the use of nonlipid carriers for mRNA delivery in contrast are rare in the context of mRNA vaccination. This paper describes the potential of a cell-penetrating peptide containing the amphipathic RALA motif to deliver antigen-encoding mRNA to the immune system. RALA condenses mRNA into nanocomplexes that display acidic pH-dependent membrane disruptive properties. RALA mRNA nanocomplexes enable mRNA escape from endosomes and thereby allow expression of mRNA inside the dendritic cell cytosol. Strikingly, RALA mRNA nanocomplexes containing pseudouridine and 5-methylcytidine modified mRNA elicit potent cytolytic T cell responses against the antigenic mRNA cargo and show superior efficacy in doing so when compared to RALA mRNA nanocomplexes containing unmodified mRNA. RALA's unique sequence and structural organization are vital to act as mRNA vaccine vehicle, as arginine-rich peptide variants that lack the RALA motif show reduced mRNA complexation, impaired cellular uptake and lose the ability to transfect dendritic cells in vitro and to evoke T cell immunity in vivo.


Asunto(s)
Antígenos , Linfocitos T CD8-positivos/inmunología , Péptidos de Penetración Celular , Sistemas de Liberación de Medicamentos , Nanoestructuras/química , ARN Mensajero , Secuencias de Aminoácidos , Animales , Antígenos/genética , Antígenos/farmacología , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/farmacocinética , Péptidos de Penetración Celular/farmacología , Citoplasma/inmunología , Endosomas/inmunología , Femenino , Ratones , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/farmacología
5.
J Lipid Res ; 58(4): 709-718, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28193630

RESUMEN

The differentiation of macrophages into lipid-filled foam cells is a hallmark of the lung granuloma that forms in patients with active tuberculosis (TB). Mycolic acids (MAs), the abundant lipid virulence factors in the cell wall of Mycobacterium tuberculosis (Mtb), can induce this foam phenotype possibly as a way to perturb host cell lipid homeostasis to support the infection. It is not exactly clear how MAs allow differentiation of foam cells during Mtb infection. Here we investigated how chemically synthetic MAs, each with a defined stereochemistry similar to natural Mtb-associated mycolates, influence cell foamy phenotype and mycobacterial proliferation in murine host macrophages. Using light and laser-scanning-confocal microscopy, we assessed the influence of MA structure first on the induction of granuloma cell types, second on intracellular cholesterol accumulation, and finally on mycobacterial growth. While methoxy-MAs (mMAs) effected multi-vacuolar giant cell formation, keto-MAs (kMAs) induced abundant intracellular lipid droplets that were packed with esterified cholesterol. Macrophages from mice treated with kMA were permissive to mycobacterial growth, whereas cells from mMA treatment were not. This suggests a separate yet key involvement of oxygenated MAs in manipulating host cell lipid homeostasis to establish the state of TB.


Asunto(s)
Colesterol/metabolismo , Granuloma/tratamiento farmacológico , Macrófagos/metabolismo , Ácidos Micólicos/administración & dosificación , Tuberculosis/tratamiento farmacológico , Animales , Diferenciación Celular/efectos de los fármacos , Células Espumosas/efectos de los fármacos , Células Espumosas/metabolismo , Granuloma/metabolismo , Granuloma/patología , Humanos , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Pulmón/metabolismo , Pulmón/patología , Macrófagos/efectos de los fármacos , Ratones , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/patogenicidad , Ácidos Micólicos/síntesis química , Tuberculosis/metabolismo , Tuberculosis/microbiología , Tuberculosis/patología
6.
Trends Mol Med ; 23(3): 216-226, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28185789

RESUMEN

mRNA vaccines have emerged as potent tools to elicit antitumor T cell immunity. They are characterized by a strong induction of type I interferons (IFNs), potent inflammatory cytokines affecting T cell differentiation and survival. Recent reports have attributed opposing roles for type I IFNs in modulating CD8+ T cell immunity to mRNA vaccines, from profoundly stimulatory to strongly inhibitory. The mechanisms behind this duality are unclear. Disentangling the factors governing the beneficial or detrimental impact of type I IFNs on CD8+ T cell responses is vital to the design of mRNA vaccines of increased potency. In light of recent advancements regarding the complex role of type I IFNs in regulating CD8+ T cell immunity to infectious diseases, we posit that the dual outcome of type I IFNs on CD8+ T cell responses to mRNA vaccination is determined by the timing and intensity of type I IFN induction relative to T cell receptor (TCR) activation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad Celular , Interferón Tipo I/inmunología , ARN Mensajero/inmunología , Vacunas/inmunología , Animales , Células Dendríticas/inmunología , Humanos , Inmunidad Innata , Activación de Linfocitos
7.
Immun Inflamm Dis ; 4(3): 350-61, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27621817

RESUMEN

INTRODUCTION: Liver X receptors (LXRs) are nuclear receptors that function as cholesterol sensors and regulate cholesterol homeostasis. High cholesterol has been recognized as a risk factor in asthma; however, the mechanism of this linkage is not known. METHODS: To explore the importance of cholesterol homeostasis for asthma, we investigated the contribution of LXR activity in an ovalbumin- and a house dust mite-driven eosinophilic asthma mouse model. RESULTS: In both models, airway inflammation, airway hyper-reactivity, and goblet cell hyperplasia were reduced in mice deficient for both LXRα and LXRß isoforms (LXRα(-/-)ß(-/-)) as compared to wild-type mice. Inversely, treatment with the LXR agonist GW3965 showed increased eosinophilic airway inflammation. LXR activity contributed to airway inflammation through promotion of type 2 cytokine production as LXRα(-/-)ß(-/-) mice showed strongly reduced protein levels of IL-5 and IL-13 in the lungs as well as reduced expression of these cytokines by CD4(+) lung cells and lung-draining lymph node cells. In line herewith, LXR activation resulted in increased type 2 cytokine production by the lung-draining lymph node cells. CONCLUSIONS: In conclusion, our study demonstrates that the cholesterol regulator LXR acts as a positive regulator of eosinophilic asthma in mice, contributing to airway inflammation through regulation of type 2 cytokine production.

8.
Mol Ther ; 24(11): 2012-2020, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27506450

RESUMEN

Given their high potential to evoke cytolytic T cell responses, tumor antigen-encoding messenger RNA (mRNA) vaccines are now being intensively explored as therapeutic cancer vaccines. mRNA vaccines clearly benefit from wrapping the mRNA into nano-sized carriers such as lipoplexes that protect the mRNA from degradation and increase its uptake by dendritic cells in vivo. Nevertheless, the early innate host factors that regulate the induction of cytolytic T cells to mRNA lipoplex vaccines have remained unresolved. Here, we demonstrate that mRNA lipoplexes induce a potent type I interferon (IFN) response upon subcutaneous, intradermal and intranodal injection. Regardless of the route of immunization applied, these type I IFNs interfered with the generation of potent cytolytic T cell responses. Most importantly, blocking type I IFN signaling at the site of immunization through the use of an IFNAR blocking antibody greatly enhanced the prophylactic and therapeutic antitumor efficacy of mRNA lipoplexes in the highly aggressive B16 melanoma model. As type I IFN induction appears to be inherent to the mRNA itself rather than to unique properties of the mRNA lipoplex formulation, preventing type I IFN induction and/or IFNAR signaling at the site of immunization might constitute a widely applicable strategy to improve the potency of mRNA vaccination.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Interferón Tipo I/metabolismo , Melanoma Experimental/tratamiento farmacológico , ARN Mensajero/administración & dosificación , Linfocitos T Citotóxicos/metabolismo , Animales , Anticuerpos/administración & dosificación , Vacunas contra el Cáncer/inmunología , Humanos , Inyecciones Intradérmicas , Inyecciones Subcutáneas , Liposomas , Melanoma Experimental/inmunología , Ratones , ARN Mensajero/inmunología , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Resultado del Tratamiento
9.
Mol Ther ; 24(9): 1686-96, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27434590

RESUMEN

DNA vaccination holds great promise for the prevention and treatment of cancer and infectious diseases. However, the clinical ability of DNA vaccines is still controversial due to the limited immune response initially observed in humans. We hypothesized that electroporation of a plasmid encoding the HIV-1 Gag viral capsid protein would enhance cancer DNA vaccine potency. DNA electroporation used to deliver plasmids in vivo, induced type I interferons, thereby supporting the activation of innate immunity. The coadministration of ovalbumin (OVA) and HIV-1 Gag encoding plasmids modulated the adaptive immune response. This strategy favored antigen-specific Th1 immunity, delayed B16F10-OVA tumor growth and improved mouse survival in both prophylactic and therapeutic vaccination approaches. Similarly, a prophylactic DNA immunization against the melanoma-associated antigen gp100 was enhanced by the codelivery of the HIV-1 Gag plasmid. The adjuvant effect was not driven by the formation of HIV-1 Gag virus-like particles. This work highlights the ability of both electroporation and the HIV-1 Gag plasmid to stimulate innate immunity for enhancing cancer DNA vaccine immunogenicity and demonstrates interesting tracks for the design of new translational genetic adjuvants to overcome the current limitations of DNA vaccines in humans.


Asunto(s)
Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Plásmidos/genética , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología , Animales , Línea Celular , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Interferón Tipo I/biosíntesis , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Plásmidos/administración & dosificación , Modelos de Riesgos Proporcionales , Células TH1/inmunología , Células TH1/metabolismo , Transfección , Resultado del Tratamiento , Carga Tumoral , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/inmunología , Vacunas de Partículas Similares a Virus/ultraestructura
10.
Proc Natl Acad Sci U S A ; 113(29): 8098-103, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27382168

RESUMEN

Agonists of Toll-like receptors (TLRs) are potent activators of the innate immune system and hold promise as vaccine adjuvant and for anticancer immunotherapy. Unfortunately, in soluble form they readily enter systemic circulation and cause systemic inflammatory toxicity. Here we demonstrate that by covalent ligation of a small-molecule imidazoquinoline-based TLR7/8 agonist to 50-nm-sized degradable polymeric nanogels the potency of the agonist to activate TLR7/8 in in vitro cultured dendritic cells is largely retained. Importantly, imidazoquinoline-ligated nanogels focused the in vivo immune activation on the draining lymph nodes while dramatically reducing systemic inflammation. Mechanistic studies revealed a prevalent passive diffusion of the nanogels to the draining lymph node. Moreover, immunization studies in mice have shown that relative to soluble TLR7/8 agonist, imidazoquinoline-ligated nanogels induce superior antibody and T-cell responses against a tuberculosis antigen. This approach opens possibilities to enhance the therapeutic benefit of small-molecule TLR agonist for a variety of applications.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Imidazoles/administración & dosificación , Nanoestructuras/administración & dosificación , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacología , Animales , Antígenos Bacterianos/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Geles , Concentración de Iones de Hidrógeno , Imidazoles/química , Imidazoles/farmacología , Inmunización , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Glicoproteínas de Membrana/agonistas , Ratones , Ratones Endogámicos C57BL , Nanoestructuras/química , Linfocitos T/inmunología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas
11.
Eur J Immunol ; 46(9): 2149-54, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27349218

RESUMEN

Mycolic acids (MAs) are highly hydrophobic long-chain α-alkyl ß-hydroxy fatty acids present in the cell wall of Mycobacterium tuberculosis (Mtb) as a complex mixture of molecules with a common general structure but with variable functional groups in the meromycolate chain. In this study, we addressed the relationship between the MA molecular structure and their contribution to the development of T-cell immune responses. Hereto, we used the model antigen ovalbumin and single synthetic MAs, differing in oxygenation class and cis versus trans proximal cyclopropane configuration, as immune stimulatory agents. Subcutaneous delivery of liposome-formulated MAs with a proximal cis cyclopropane elicited antigen-specific Th1 and cytotoxic T-cell immune responses, whereas intratracheal immunization elicited pulmonary Th17 responses. These immune stimulatory activities depended not only on the cis versus trans proximal cyclopropane configuration but also on the MA oxygenation class. Our study thus shows that both the presence and nature of the functional groups in the meromycolate chain affect the immune stimulatory adjuvant activity of Mtb mycolates and suggests that Mtb bacilli may impact on the host protective immune response by modulating the cis versus trans stereochemistry of its mycolates as well as by altering the oxygenation class of the meromycolate functional group.


Asunto(s)
Adyuvantes Inmunológicos , Mycobacterium tuberculosis/inmunología , Ácidos Micólicos/inmunología , Tuberculosis/inmunología , Animales , Citocinas/biosíntesis , Femenino , Inmunización , Inmunoglobulina G/inmunología , Factores Inmunológicos , Inmunomodulación , Inyección Intratimpánica , Inyecciones Subcutáneas , Interferón gamma/biosíntesis , Liposomas/química , Activación de Linfocitos , Ratones , Mycobacterium tuberculosis/metabolismo , Ácidos Micólicos/administración & dosificación , Ácidos Micólicos/química , Fosfatidilcolinas/química , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Tuberculosis/prevención & control , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas contra la Tuberculosis/inmunología
12.
Biomacromolecules ; 17(7): 2479-88, 2016 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-27231045

RESUMEN

We report on the design of glycosylated nanogels via core-cross-linking of amphiphilic non-water-soluble block copolymers composed of an acetylated glycosylated block and a pentafluorophenyl (PFP) activated ester block prepared by reversible addition-fragmentation (RAFT) polymerization. Self-assembly, pH-sensitive core-cross-linking, and removal of remaining PFP esters and protecting groups are achieved in one pot and yield fully hydrated sub-100 nm nanogels. Using cell subsets that exhibit high and low expression of the mannose receptor (MR) under conditions that suppress active endocytosis, we show that mannosylated but not galactosylated nanogels can efficiently target the MR that is expressed on the cell surface of primary dendritic cells (DCs). These nanogels hold promise for immunological applications involving DCs and macrophage subsets.


Asunto(s)
Células Dendríticas/citología , Células Dendríticas/metabolismo , Manosa/química , Polietilenglicoles/química , Polietileneimina/química , Polímeros/química , Animales , Células Cultivadas , Concentración de Iones de Hidrógeno , Lectinas Tipo C/metabolismo , Manosa/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Nanogeles , Polimerizacion , Receptores de Superficie Celular/metabolismo
13.
Cell Rep ; 15(2): 274-87, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-27050509

RESUMEN

Successful immunogenic apoptosis in experimental cancer therapy depends on the induction of strong host anti-tumor responses. Given that tumors are often resistant to apoptosis, it is important to identify alternative molecular mechanisms that elicit immunogenic cell death. We have developed a genetic model in which direct dimerization of FADD combined with inducible expression of RIPK3 promotes necroptosis. We report that necroptotic cancer cells release damage-associated molecular patterns and promote maturation of dendritic cells, the cross-priming of cytotoxic T cells, and the production of IFN-γ in response to tumor antigen stimulation. Using both FADD-dependent and FADD-independent RIPK3 induction systems, we demonstrate the efficient vaccination potential of immunogenic necroptotic cells. Our study broadens the current concept of immunogenic cell death and opens doors for the development of new strategies in cancer therapy.


Asunto(s)
Antineoplásicos/inmunología , Apoptosis , Inmunidad , Neoplasias/inmunología , Vacunación , Alarminas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocinas/metabolismo , Reactividad Cruzada/efectos de los fármacos , Reactividad Cruzada/inmunología , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Inmunidad/efectos de los fármacos , Ligandos , Ratones , Modelos Biológicos , FN-kappa B/metabolismo , Necrosis , Fagocitosis/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Tetraciclina/farmacología
14.
J Allergy Clin Immunol ; 137(3): 700-9.e9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26560044

RESUMEN

BACKGROUND: Human respiratory syncytial virus (RSV) is a frequent cause of asthma exacerbations, yet the susceptibility of asthmatic patients to RSV is poorly understood. OBJECTIVE: We sought to address the contribution of resident alveolar macrophages (rAMs) to susceptibility to RSV infection in mice that recovered from allergic airway eosinophilia. METHODS: Mice were infected with RSV virus after clearance of allergic airway inflammation (AAI). The contribution of post-AAI rAMs was studied in vivo by means of clodronate liposome-mediated depletion, adoptive transfer, and treatment with recombinant cytokines before RSV infection. RESULTS: After clearing the allergic bronchial inflammation, post-AAI mice had bronchial hyperreactivity and increased inflammatory cell influx when infected with RSV compared with nonallergic mice, whereas viral clearance was comparable in both mouse groups. Post-AAI rAMs were necessary and sufficient for mediating these proinflammatory effects. In post-AAI mice the residing CD11c(hi) autofluorescent rAM population did not upregulate the terminal differentiation marker sialic acid-binding immunoglobulin-like lectin F and overproduced TNF and IL-6 through increased nuclear factor κB nuclear translocation. In line with these results, post-AAI lungs had reduced levels of the rAM maturation cytokine GM-CSF. Intratracheal administration of GM-CSF induced final rAM maturation in post-AAI mice and prevented the increased susceptibility to RSV-induced hyperreactivity and inflammation. CONCLUSION: Defective production of GM-CSF leads to insufficient post-AAI rAM maturation in mice that recovered from an AAI, causing increased susceptibility to RSV-induced immunopathology. Promoting the differentiation of post-AAI rAMs might be a therapeutic option for preventing RSV-induced exacerbations in human asthmatic patients.


Asunto(s)
Asma/complicaciones , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Macrófagos Alveolares/efectos de los fármacos , Infecciones por Virus Sincitial Respiratorio/complicaciones , Virus Sincitial Respiratorio Humano , Traslado Adoptivo , Alérgenos/inmunología , Animales , Asma/inmunología , Asma/metabolismo , Asma/patología , Asma/terapia , Líquido del Lavado Bronquioalveolar , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Inmunofenotipificación , Mediadores de Inflamación/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Ratones , FN-kappa B/metabolismo , Fenotipo , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/patología
15.
Angew Chem Int Ed Engl ; 55(4): 1334-9, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26666207

RESUMEN

The induction of antigen-specific adaptive immunity exclusively occurs in lymphoid organs. As a consequence, the efficacy by which vaccines reach these tissues strongly affects the efficacy of the vaccine. Here, we report the design of polymer hydrogel nanoparticles that efficiently target multiple immune cell subsets in the draining lymph nodes. Nanoparticles are fabricated by infiltrating mesoporous silica particles (ca. 200 nm) with poly(methacrylic acid) followed by disulfide-based crosslinking and template removal. PEGylation of these nanoparticles does not affect their cellular association in vitro, but dramatically improves their lymphatic drainage in vivo. The functional relevance of these observations is further illustrated by the increased priming of antigen-specific T cells. Our findings highlight the potential of engineered hydrogel nanoparticles for the lymphatic delivery of antigens and immune-modulating compounds.


Asunto(s)
Hidrogeles , Ganglios Linfáticos/metabolismo , Nanopartículas , Polímeros/química , Vacunas/administración & dosificación , Animales , Antígenos CD/inmunología , Ganglios Linfáticos/inmunología , Ratones , Microscopía Electrónica de Transmisión
16.
ACS Appl Mater Interfaces ; 8(2): 1147-55, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26694764

RESUMEN

Nanomaterials hold potential of altering the interaction between therapeutic molecules and target cells or tissues. High aspect ratio nanomaterials in particular have been reported to possess unprecedented properties and are intensively investigated for their interaction with biological systems. Graphene oxide (GOx) is a water-soluble graphene derivative that combines high aspect ratio dimension with functional groups that can be exploited for bioconjugation. Here, we demonstrate that GOx nanosheets can spontaneously adsorb proteins by a combination of interactions. This property is then explored for intracellular protein vaccine delivery, in view of the potential of GOx nanosheets to destabilize lipid membranes such as those of intracellular vesicles. Using a series of in vitro experiments, we show that GOx nanosheet adsorbed proteins are efficiently internalized by dendritic cells (DCs: the most potent class of antigen presenting cells of the immune system) and promote antigen cross-presentation to CD8 T cells. The latter is a hallmark in the induction of potent cellular antigen-specific immune responses against intracellular pathogens and cancer.


Asunto(s)
Sistemas de Liberación de Medicamentos , Grafito/química , Proteínas/química , Vacunas/química , Adsorción , Células Dendríticas/efectos de los fármacos , Grafito/uso terapéutico , Humanos , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Óxidos , Proteínas/uso terapéutico , Linfocitos T/efectos de los fármacos , Vacunas/uso terapéutico
17.
J Control Release ; 217: 53-63, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26307350

RESUMEN

The local delivery of small interfering RNA (siRNA) to the lungs may provide a therapeutic solution to a range of pulmonary disorders. Resident alveolar macrophages (rAM) in the bronchoalveolar lumen play a critical role in lung inflammatory responses and therefore constitute a particularly attractive target for siRNA therapeutics. However, achieving efficient gene silencing in the lung while avoiding pulmonary toxicity requires appropriate formulation of siRNA in functional nanocarriers. In this study, we evaluated pulmonary surfactant-coated dextran nanogels for the delivery of siRNA to rAM upon pharyngeal aspiration in BALB/c mice. Both the surfactant-coated and uncoated nanogels achieved high levels of siRNA uptake in rAM, yet only the surfactant-coated formulation could significantly reduce gene expression on the protein level. Surfactant-coated nanogels induced a profound downregulation of target mRNA levels, reaching 70% knockdown with ~1mgkg(-1) siRNA dose. In addition, only mild acute pro-inflammatory cytokine and chemokine responses were detected one day after nanoparticle aspiration, accompanied by a moderate neutrophil infiltration in the bronchoalveolar lumen. The latter could be substantially reduced by removal of excess surfactant from the formulation. Overall, our hybrid core-shell nanoparticles have demonstrated safe and effective siRNA delivery to rAM, providing a new therapeutic approach for treatment of inflammatory pathologies in the lung.


Asunto(s)
Antígenos Comunes de Leucocito/genética , Macrófagos Alveolares/metabolismo , Nanopartículas/administración & dosificación , Surfactantes Pulmonares/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Recuento de Células , Citocinas/metabolismo , Dextranos/química , Femenino , Geles , Silenciador del Gen , Antígenos Comunes de Leucocito/metabolismo , Ratones Endogámicos BALB C , Nanopartículas/química , Surfactantes Pulmonares/química
18.
Sci Rep ; 5: 12599, 2015 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-26226941

RESUMEN

IL-4/IL-13-induced alternatively activated macrophages (M(IL-4/IL-13), AAMs or M2) are known to express E-cadherin, enabling them to engage in heterotypic cellular interactions and IL-4-driven macrophage fusion in vitro. Here we show that E-cadherin overexpression in Raw 264.7 macrophages inhibits their inflammatory response to LPS stimulation, as demonstrated by a reduced secretion of inflammatory mediators like interleukin (IL)-6, tumor necrosis factor (TNF) and nitric oxide (NO). To study the function of E-cadherin in M(IL-4/IL-13) macrophages in vivo, we generated macrophage-specific E-cadherin-deficient C57BL/6 mice. Using this new tool, we analyzed immunological parameters during two typical AAM-associated Th2-driven diseases and assessed Th2-associated granuloma formation. Although E-cadherin is strongly induced in AAMs during Taenia crassiceps helminth infections and allergic airway inflammation, its deletion in macrophages does not affect the course of both Th2 cytokine-driven diseases. Moreover, macrophage E-cadherin expression is largely redundant for granuloma formation around Schistosoma mansoni ova. Overall, we conclude that E-cadherin is a valuable AAM marker which suppresses the inflammatory response when overexpressed. Yet E-cadherin deletion in macrophages does not affect M(LPS+IFNγ) and M(IL-4) polarization in vitro, nor in vivo macrophage function, at least in the conditions tested.


Asunto(s)
Cadherinas/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Macrófagos/fisiología , Animales , Linfocitos B/patología , Lavado Broncoalveolar , Cadherinas/genética , Cisticercosis/inmunología , Cisticercosis/patología , Inflamación/genética , Inflamación/patología , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Macrófagos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Esquistosomiasis mansoni/metabolismo , Esquistosomiasis mansoni/patología , Células Th2/inmunología
19.
Vaccine ; 33(28): 3179-85, 2015 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-25980430

RESUMEN

We investigated the influence of the site of administration of DNA vaccine on the induced immune response. DNA vaccines were administered by electroporation at three different sites: tibial cranial muscle, abdominal skin and ear pinna. Aiming to draw general conclusions about DNA vaccine delivery, we successively used several plasmids encoding either luciferase and ovalbumin as models or gp160 and P1A as vaccines against HIV and P815 mastocytoma, respectively. Low levels and duration of luciferase transgene expression were observed after electroporation of the abdominal skin, partly explaining its lower immunogenic performance as compared to the other sites of administration. Analyses of OT-I CD8+ and OT-II CD4+ T cell responses highlighted the differential impact of the delivery site on the elicited immune response. Muscle electroporation induced the strongest humoral immune response and both muscle and ear pinna sites induced cellular immunity against gp160. Ear pinna delivery generated the highest level of CTL responses against P1A but electroporation of muscle and ear pinna were equally efficient in delaying P815 growth and improving mice survival. The present study demonstrated that the site of administration is a key factor to be tested in the development of DNA vaccine.


Asunto(s)
Electroporación , Vacunación/métodos , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/inmunología , Abdomen , Administración Cutánea , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Pabellón Auricular , Femenino , Anticuerpos Anti-VIH/sangre , Anticuerpos Anti-VIH/inmunología , Proteínas gp160 de Envoltorio del VIH/genética , VIH-1/inmunología , Miembro Posterior , Inmunidad Celular , Inmunidad Humoral , Luciferasas/genética , Ratones Endogámicos C57BL , Músculo Esquelético , Plásmidos , Piel/inmunología
20.
J Control Release ; 195: 99-109, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25078552

RESUMEN

In this paper we report on the design, characterization and immuno-biological evaluation of nanoporous polyelectrolyte microparticles as vaccine carrier. Relative to soluble antigen, formulation of antigen as a sub-10 µm particle can strongly enhance antigen-specific cellular immune responses. The latter is crucial to confer protective immunity against intracellular pathogens and for anti-cancer vaccines. However, a major bottleneck in microparticulate vaccine formulation is the development of generic strategies that afford antigen encapsulation under benign and scalable conditions. Our strategy is based on spray drying of a dilute aqueous solution of antigen, oppositely charged polyelectrolytes and mannitol as a pore-forming component. The obtained solid microparticles can be redispersed in aqueous medium, leading to leaching out of the mannitol, thereby creating a highly porous internal structure. This porous structure enhances enzymatic processing of encapsulated proteins. After optimizing the conditions to process these microparticles we demonstrate that they strongly enhance cross-presentation in vitro by dendritic cells to CD8 T cells. In vivo experiments in mice confirm that this vaccine formulation technology is capable of enhancing cellular immune responses.


Asunto(s)
Sulfato de Dextran/química , Portadores de Fármacos/química , Manitol/química , Ovalbúmina/química , Péptidos/química , Vacunas/química , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Portadores de Fármacos/administración & dosificación , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovalbúmina/administración & dosificación , Porosidad , Vacunas/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA