Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Immunol ; 11: 606805, 2020.
Article in English | MEDLINE | ID: mdl-33519816

ABSTRACT

The Adjuvant System AS01 contains monophosphoryl lipid A (MPL) and the saponin QS-21 in a liposomal formulation. AS01 is included in recently developed vaccines against malaria and varicella zoster virus. Like for many other adjuvants, induction of adaptive immunity by AS01 is highly dependent on the ability to recruit and activate dendritic cells (DCs) that migrate to the draining lymph node for T and B cell stimulation. The objective of this study was to more precisely address the contribution of the different conventional (cDC) and monocyte-derived DC (MC) subsets in the orchestration of the adaptive immune response after immunization with AS01 adjuvanted vaccine. The combination of MPL and QS-21 in AS01 induced strong recruitment of CD26+XCR1+ cDC1s, CD26+CD172+ cDC2s and a recently defined CCR2-dependent CD64-expressing inflammatory cDC2 (inf-cDC2) subset to the draining lymph node compared to antigen alone, while CD26-CD64+CD88+ MCs were barely detectable. At 24 h post-vaccination, cDC2s and inf-cDC2s were superior amongst the different subsets in priming antigen-specific CD4+ T cells, while simultaneously presenting antigen to CD8+ T cells. Diphtheria toxin (DT) mediated depletion of all DCs prior to vaccination completely abolished adaptive immune responses, while depletion 24 h after vaccination mainly affected CD8+ T cell responses. Vaccinated mice lacking Flt3 or the chemokine receptor CCR2 showed a marked deficit in inf-cDC2 recruitment and failed to raise proper antibody and T cell responses. Thus, the adjuvant activity of AS01 is associated with the potent activation of subsets of cDC2s, including the newly described inf-cDC2s.


Subject(s)
Adaptive Immunity/drug effects , Adjuvants, Immunologic/pharmacology , Dendritic Cells/drug effects , Herpes Zoster Vaccine/pharmacology , Lipid A/analogs & derivatives , Receptors, CCR2/metabolism , Saponins/pharmacology , Viral Envelope Proteins/pharmacology , fms-Like Tyrosine Kinase 3/metabolism , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Immunization , Lipid A/pharmacology , Liposomes , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/pharmacology , Receptors, CCR2/genetics , Signal Transduction , fms-Like Tyrosine Kinase 3/genetics
2.
J Allergy Clin Immunol ; 140(1): 76-88.e7, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27746238

ABSTRACT

BACKGROUND: Allergic asthma is a CD4 TH2-lymphocyte driven disease characterized by airway hyperresponsiveness and eosinophilia. B cells can present antigens to CD4 T cells and produce IgE immunoglobulins that arm effector cells; however, mouse models are inconclusive on whether B cells are necessary for asthma development. OBJECTIVES: We sought to address the role of B cells in a house dust mite (HDM)-driven TH2-high asthma mouse model. METHODS: Wild-type and B cell-deficient muMT mice were sensitized and challenged through the airways with HDM extracts. The antigen-presenting capacities of B cells were studied by using new T-cell receptor transgenic 1-DER mice specific for the Der p 1 allergen. RESULTS: In vitro-activated B cells from HDM-exposed mice presented antigen to 1-DER T cells and induced a TH2 phenotype. In vivo B cells were dispensable for activation of naive 1-DER T cells but necessary for full expansion of primed 1-DER T cells. At high HDM challenge doses, B cells were not required for development of pulmonary asthmatic features yet contributed to TH2 expansion in the mediastinal lymph nodes but not in the lungs. When the amount of challenge allergen was decreased, muMT mice had reduced asthma features. Under these limiting conditions, B cells contributed also to expansion of TH2 effector cells in the lungs and central memory T cells in the mediastinal lymph nodes. CONCLUSION: B cells are a major part of the adaptive immune response to inhaled HDM allergen, particularly when the amount of inhaled allergen is low, by expanding allergen-specific T cells.


Subject(s)
Antigens, Dermatophagoides/immunology , Asthma/immunology , B-Lymphocytes/immunology , Th2 Cells/immunology , Animals , Antigen Presentation , Cytokines/immunology , Lymph Nodes/cytology , Mice, Inbred C57BL , Mice, Transgenic , Pyroglyphidae/immunology , Spleen/cytology
3.
Respir Res ; 17: 35, 2016 Apr 02.
Article in English | MEDLINE | ID: mdl-27039089

ABSTRACT

ß-(1,3)-Glucan is present in mould cell walls and frequently detected in house dust mite (HDM) faeces. ß-Glucan exposure is thought to be associated with pulmonary allergic inflammation in mouse and man, although the published data are inconsistent. Here, we show that highly purified ß-glucan exacerbates HDM-induced eosinophilic, T helper 2 type airway responses by acting as an adjuvant, promoting activation, proliferation and polarisation of HDM-specific T cells (1-Derß T cells). We therefore provide definitive evidence that ß-glucan can influence allergic pulmonary inflammation.


Subject(s)
Antigens, Dermatophagoides/immunology , Antigens, Dermatophagoides/toxicity , Asthma/chemically induced , Asthma/immunology , beta-Glucans/immunology , beta-Glucans/toxicity , Animals , Drug Synergism , Environmental Exposure/adverse effects , Mice , Mice, Inbred C57BL , Mycotoxins/immunology , Mycotoxins/toxicity
4.
Biomacromolecules ; 17(3): 874-81, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26812240

ABSTRACT

Although the field of cancer immunotherapy is intensively investigated, there is still a need for generic strategies that allow easy, mild and efficient formulation of vaccine antigens. Here we report on a generic polymer-protein ligation strategy to formulate protein antigens into reversible polymeric conjugates for enhanced uptake by dendritic cells and presentation to CD8 T-cells. A N-hydroxypropylmethacrylamide (HPMA)-based copolymer was synthesized via RAFT polymerization followed by introduction of pyridyldisulfide moieties. To enhance ligation efficiency to ovalbumin, which is used as a model protein antigen, protected thiols were introduced onto lysine residues and deprotected in situ in the presence of the polymer. The ligation efficiency was compared for both the thiol-modified versus unmodified ovalbumin, and the reversibility was confirmed. Furthermore, the obtained nanoconjugates were tested in vitro for their interaction and association with dendritic cells, showing enhanced cellular uptake and antigen cross-presentation to CD8 T-cells.


Subject(s)
Antigens/chemistry , Methacrylates/chemistry , Nanoconjugates/chemistry , Ovalbumin/chemistry , Vaccines, Conjugate/chemistry , Animals , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Dendritic Cells/immunology , Mice , Ovalbumin/immunology , Vaccines, Conjugate/immunology
5.
ACS Appl Mater Interfaces ; 8(2): 1147-55, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26694764

ABSTRACT

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.


Subject(s)
Drug Delivery Systems , Graphite/chemistry , Proteins/chemistry , Vaccines/chemistry , Adsorption , Dendritic Cells/drug effects , Graphite/therapeutic use , Humans , Nanostructures/chemistry , Nanostructures/therapeutic use , Oxides , Proteins/therapeutic use , T-Lymphocytes/drug effects , Vaccines/therapeutic use
7.
PLoS One ; 10(8): e0134219, 2015.
Article in English | MEDLINE | ID: mdl-26261989

ABSTRACT

Asthma is a heterogeneous disease whose etiology is poorly understood but is likely to involve innate responses to inhaled microbial components that are found in allergens. The influence of these components on pulmonary inflammation has been largely studied in the context of individual agonists, despite knowledge that they can have synergistic effects when used in combination. Here we have explored the effects of LPS and ß-glucan, two commonly-encountered microbial agonists, on the pathogenesis of allergic and non-allergic respiratory responses to house dust mite allergen. Notably, sensitization with these microbial components in combination acted synergistically to promote robust neutrophilic inflammation, which involved both Dectin-1 and TLR-4. This pulmonary neutrophilic inflammation was corticosteroid-refractory, resembling that found in patients with severe asthma. Thus our results provide key new insights into how microbial components influence the development of respiratory pathology.


Subject(s)
Asthma/etiology , Lipopolysaccharides/immunology , Neutrophils/immunology , beta-Glucans/immunology , Animals , Asthma/drug therapy , Asthma/metabolism , Asthma/pathology , Disease Models, Animal , Drug Resistance , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Mice, Knockout , Neutrophil Infiltration , Neutrophils/metabolism , Neutrophils/pathology , Pyroglyphidae/immunology , Steroids/administration & dosage , Steroids/pharmacology , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
8.
J Control Release ; 195: 99-109, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25078552

ABSTRACT

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.


Subject(s)
Dextran Sulfate/chemistry , Drug Carriers/chemistry , Mannitol/chemistry , Ovalbumin/chemistry , Peptides/chemistry , Vaccines/chemistry , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Drug Carriers/administration & dosage , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/administration & dosage , Porosity , Vaccines/administration & dosage
9.
J Immunol ; 193(4): 1920-30, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25024381

ABSTRACT

Adjuvant System AS01 is a liposome-based vaccine adjuvant containing 3-O-desacyl-4'-monophosphoryl lipid A and the saponin QS-21. AS01 has been selected for the clinical development of several candidate vaccines including the RTS,S malaria vaccine and the subunit glycoprotein E varicella zoster vaccine (both currently in phase III). Given the known immunostimulatory properties of MPL and QS-21, the objective of this study was to describe the early immune response parameters after immunization with an AS01-adjuvanted vaccine and to identify relationships with the vaccine-specific adaptive immune response. Cytokine production and innate immune cell recruitment occurred rapidly and transiently at the muscle injection site and draining lymph node postinjection, consistent with the rapid drainage of the vaccine components to the draining lymph node. The induction of Ag-specific Ab and T cell responses was dependent on the Ag being injected at the same time or within 24 h after AS01, suggesting that the early events occurring postinjection were required for these elevated adaptive responses. In the draining lymph node, after 24 h, the numbers of activated and Ag-loaded monocytes and MHCII(high) dendritic cells were higher after the injection of the AS01-adjuvanted vaccine than after Ag alone. However, only MHCII(high) dendritic cells appeared efficient at and necessary for direct Ag presentation to T cells. These data suggest that the ability of AS01 to improve adaptive immune responses, as has been demonstrated in clinical trials, is linked to a transient stimulation of the innate immune system leading to the generation of high number of efficient Ag-presenting dendritic cells.


Subject(s)
Adaptive Immunity/immunology , Adjuvants, Immunologic/administration & dosage , Dendritic Cells/immunology , Lipid A/analogs & derivatives , Saponins/administration & dosage , Saponins/immunology , Vaccines/immunology , Animals , Antigen Presentation/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Drug Combinations , Female , Histocompatibility Antigens Class II/immunology , Humans , Lipid A/administration & dosage , Lipid A/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Vaccination
10.
J Exp Med ; 211(6): 1019-25, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24799501

ABSTRACT

There is currently no paradigm in immunology that enables an accurate prediction of how the immune system will respond to any given agent. Here we show that the immunological responses induced by members of a broad class of inorganic crystalline materials are controlled purely by their physicochemical properties in a highly predictable manner. We show that structurally and chemically homogeneous layered double hydroxides (LDHs) can elicit diverse human dendritic cell responses in vitro. Using a systems vaccinology approach, we find that every measured response can be modeled using a subset of just three physical and chemical properties for all compounds tested. This correlation can be reduced to a simple linear equation that enables the immunological responses stimulated by newly synthesized LDHs to be predicted in advance from these three parameters alone. We also show that mouse antigen-specific antibody responses in vivo and human macrophage responses in vitro are controlled by the same properties, suggesting they may control diverse responses at both individual component and global levels of immunity. This study demonstrates that immunity can be determined purely by chemistry and opens the possibility of rational manipulation of immunity for therapeutic purposes.


Subject(s)
Antibody Formation/immunology , Dendritic Cells/immunology , Hydroxides/immunology , Macrophages/immunology , Animals , Antibodies/blood , Antibodies/immunology , Cells, Cultured , Crystallization , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Hydroxides/chemistry , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Multivariate Analysis , Ovalbumin/immunology
11.
PLoS Biol ; 12(1): e1001762, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24453940

ABSTRACT

Mast cells are implicated in the pathogenesis of inflammatory and autoimmune diseases. However, this notion based on studies in mast cell-deficient mice is controversial. We therefore established an in vivo model for hyperactive mast cells by specifically ablating the NF-κB negative feedback regulator A20. While A20 deficiency did not affect mast cell degranulation, it resulted in amplified pro-inflammatory responses downstream of IgE/FcεRI, TLRs, IL-1R, and IL-33R. As a consequence house dust mite- and IL-33-driven lung inflammation, late phase cutaneous anaphylaxis, and collagen-induced arthritis were aggravated, in contrast to experimental autoimmune encephalomyelitis and immediate anaphylaxis. Our results provide in vivo evidence that hyperactive mast cells can exacerbate inflammatory disorders and define diseases that might benefit from therapeutic intervention with mast cell function.


Subject(s)
Anaphylaxis/immunology , Arthritis, Experimental/immunology , DNA-Binding Proteins/deficiency , Encephalomyelitis, Autoimmune, Experimental/immunology , Intracellular Signaling Peptides and Proteins/deficiency , Mast Cells/immunology , Ubiquitin-Protein Ligases/deficiency , Anaphylaxis/chemically induced , Anaphylaxis/metabolism , Anaphylaxis/pathology , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Collagen Type II/administration & dosage , Cysteine Endopeptidases , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Dinitrophenols/administration & dosage , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/genetics , Interleukins/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Mast Cells/metabolism , Mast Cells/pathology , Mice , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , NF-kappa B/genetics , NF-kappa B/immunology , Peptide Fragments/administration & dosage , Pneumonia/chemically induced , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , Pyroglyphidae/immunology , Receptors, IgE/genetics , Receptors, IgE/immunology , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Serum Albumin, Bovine/administration & dosage , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
12.
Curr Pharm Des ; 18(16): 2305-13, 2012.
Article in English | MEDLINE | ID: mdl-22390693

ABSTRACT

Vaccinations have been given to people for at least 100 years of which 80 years formulated in adjuvants. Currently, aluminium- containing formulas and the oil-based substance called MF59 are the two adjuvants licensed for clinical use. Despite the massive use of aluminium adjuvant, its mechanism of action has been only recently addressed. In this review, we will discuss both the cellular and molecular mode of action of aluminium-containing adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum/pharmacology , Adjuvants, Immunologic/chemistry , Aluminum/analysis , Animals , Humans
13.
J Med Microbiol ; 61(Pt 7): 927-934, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22174375

ABSTRACT

Alum has been the most widely used adjuvant for over 80 years. Although there have been searches for alternative adjuvants, aluminium-containing adjuvants will continue to be used for many years due to their good track record of safety, low cost and adjuvanticity with a variety of antigens. For infections that can be prevented by induction of serum antibodies, aluminium-containing adjuvants formulated under optimal conditions are the adjuvants of choice. There are also some limitations of aluminium-containing adjuvants, which include local reactions, augmentation of IgE antibody responses, ineffectiveness for some antigens and inability to augment cell-mediated immune responses, especially cytotoxic T-cell responses. In this review, we describe the current knowledge regarding the mechanisms (both cellular and molecular) by which alum employs its adjuvant effect, although the final mechanism is not yet well-defined. Furthermore, we discuss how alum's adjuvanticity could be improved.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Alum Compounds/administration & dosage , Alum Compounds/pharmacology , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/economics , Alum Compounds/adverse effects , Alum Compounds/economics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...