Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Immunol ; 5(48)2020 06 26.
Article in English | MEDLINE | ID: mdl-32591409

ABSTRACT

Liver resident-memory CD8+ T cells (TRM cells) can kill liver-stage Plasmodium-infected cells and prevent malaria, but simple vaccines for generating this important immune population are lacking. Here, we report the development of a fully synthetic self-adjuvanting glycolipid-peptide conjugate vaccine designed to efficiently induce liver TRM cells. Upon cleavage in vivo, the glycolipid-peptide conjugate vaccine releases an MHC I-restricted peptide epitope (to stimulate Plasmodium-specific CD8+ T cells) and an adjuvant component, the NKT cell agonist α-galactosylceramide (α-GalCer). A single dose of this vaccine in mice induced substantial numbers of intrahepatic malaria-specific CD8+ T cells expressing canonical markers of liver TRM cells (CD69, CXCR6, and CD101), and these cells could be further increased in number upon vaccine boosting. We show that modifications to the peptide, such as addition of proteasomal-cleavage sequences or epitope-flanking sequences, or the use of alternative conjugation methods to link the peptide to the glycolipid improved liver TRM cell generation and led to the development of a vaccine able to induce sterile protection in C57BL/6 mice against Plasmodium berghei sporozoite challenge after a single dose. Furthermore, this vaccine induced endogenous liver TRM cells that were long-lived (half-life of ~425 days) and were able to maintain >90% sterile protection to day 200. Our findings describe an ideal synthetic vaccine platform for generating large numbers of liver TRM cells for effective control of liver-stage malaria and, potentially, a variety of other hepatotropic infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Glycolipids/immunology , Liver/immunology , Malaria Vaccines/immunology , Malaria/immunology , Peptides/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Liver/pathology , Malaria/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Vaccination
2.
Org Biomol Chem ; 17(5): 1225-1237, 2019 01 31.
Article in English | MEDLINE | ID: mdl-30656346

ABSTRACT

Activated NKT cells can stimulate antigen-presenting cells leading to enhanced peptide antigen-specific immunity. However, administration of potent NKT cell agonists like α-galactosylceramide (α-GalCer) can be associated with release of high levels of cytokines, and in some situations, hepatotoxicity. Here we show that it is possible to provoke sufficient NKT cell activity to stimulate strong antigen-specific T cell responses without these unwanted effects. This was achieved by chemically conjugating antigenic peptides to α-galactosylphytosphingosine (α-GalPhs), an NKT cell agonist with very weak activity based on structural characterisation and biological assays. Conjugation improved delivery to antigen-presenting cells in vivo, while use of a cathepsin-sensitive linker to release the α-GalPhs and peptide within the same cell promoted strong T cell activation and therapeutic anti-tumour responses in mice. The conjugates activated human NKT cells and enhanced human T cell responses to a viral peptide in vitro. Accordingly, we have demonstrated a means to safely exploit the immunostimulatory properties of NKT cells to enhance T cell activation for virus- and tumour-specific immunity.


Subject(s)
Antigen-Presenting Cells/immunology , Cancer Vaccines/administration & dosage , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Neoplasms, Experimental/immunology , Peptides/administration & dosage , Adjuvants, Immunologic , Animals , Antigens, CD1d/chemistry , Cancer Vaccines/immunology , Chemical and Drug Induced Liver Injury/prevention & control , Epitopes/chemistry , Glycolipids/chemistry , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/drug therapy , Peptides/chemistry , Peptides/immunology
3.
ACS Chem Biol ; 12(11): 2898-2905, 2017 11 17.
Article in English | MEDLINE | ID: mdl-29043774

ABSTRACT

The development of a universal vaccine for influenza A virus (IAV) that does not require seasonal modification is a long-standing health goal, particularly in the context of the increasing threat of new global pandemics. Vaccines that specifically induce T cell responses are of considerable interest because they can target viral proteins that are more likely to be shared between different virus strains and subtypes and hence provide effective cross-reactive IAV immunity. From a practical perspective, such vaccines should induce T cell responses with long-lasting memory, while also being simple to manufacture and cost-effective. Here we describe the synthesis and evaluation of a vaccine platform based on solid phase peptide synthesis and bio-orthogonal conjugation methodologies. The chemical approach involves covalently attaching synthetic long peptides from a virus-associated protein to a powerful adjuvant molecule, α-galactosylceramide (α-GalCer). Strain-promoted azide-alkyne cycloaddition is used as a simple and efficient method for conjugation, and pseudoproline methodology is used to increase the efficiency of the peptide synthesis. α-GalCer is a glycolipid that stimulates NKT cells, a population of lymphoid-resident immune cells that can provide potent stimulatory signals to antigen-presenting cells engaged in driving proliferation and differentiation of peptide-specific T cells. When used in mice, the vaccine induced T cell responses that provided effective prophylactic protection against IAV infection, with the speed of viral clearance greater than that seen from previous viral exposure. These findings are significant because the vaccines are highly defined, quick to synthesize, and easily characterized and are therefore appropriate for large scale affordable manufacture.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Galactosylceramides/therapeutic use , Influenza A virus/immunology , Influenza Vaccines/therapeutic use , Orthomyxoviridae Infections/prevention & control , Peptides/therapeutic use , Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/pharmacology , Animals , CD8-Positive T-Lymphocytes/immunology , Cycloaddition Reaction , Female , Galactosylceramides/chemical synthesis , Galactosylceramides/immunology , Humans , Influenza A virus/chemistry , Influenza Vaccines/chemical synthesis , Influenza, Human/immunology , Influenza, Human/prevention & control , Mice, Inbred C57BL , Natural Killer T-Cells/immunology , Orthomyxoviridae Infections/immunology , Peptides/chemical synthesis , Peptides/immunology , Solid-Phase Synthesis Techniques
4.
Org Lett ; 17(24): 5954-7, 2015 Dec 18.
Article in English | MEDLINE | ID: mdl-26606283

ABSTRACT

A major challenge in the development of highly defined synthetic vaccines is the codelivery of vaccine components (i.e., antigen and adjuvant) to secondary lymphoid tissue to induce optimal immune responses. This problem can be addressed by synthesizing vaccines that comprise peptide antigens covalently attached to glycolipid adjuvants through biologically cleavable linkers. Toward this, a strategy utilizing previously unreported 6″-deoxy-6″-thio analogues of α-GalCer that can undergo chemoselective conjugation with peptide antigens is described. Administration of these conjugate vaccines leads to enhanced priming of antigen specific T cells. This simple vaccine design is broadly applicable to multiple disease indications such as cancer and infectious disease.


Subject(s)
Galactosylceramides/chemical synthesis , Macrocyclic Compounds/chemical synthesis , Peptides/chemical synthesis , Cesium/analysis , Galactosylceramides/chemistry , Humans , Macrocyclic Compounds/chemistry , Maleimides/chemical synthesis , Maleimides/chemistry , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry
5.
J Immunother ; 38(8): 321-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26325376

ABSTRACT

Circulating antigens released from tumor cells can drain into the spleen and be acquired by resident antigen-presenting cells (APCs). Here, we examined the ability of splenic dendritic cells to cross-present tumor antigens to CD8+ T cells and investigated the effects that this has on T-cell therapy in a murine model of lymphoma. In the presence of established lymphoma, langerin (CD207)-expressing CD8α+ dendritic cells acquired, processed, and cross-presented tumor antigens to naive CD8+ T cells. Although this resulted in initial T-cell proliferation, the T-cell population failed to expand measurably over the following days, and tumor-free survival was actually improved when langerin-expressing cells were depleted. In contrast, following adoptive T-cell therapy with in vitro-activated CD8+ T cells, marked antitumor activity was observed and associated with accumulation of activated antigen-specific CD8+ T cells in the spleen and blood, whereas tumor protection and T-cell accumulation were significantly reduced in animals depleted of langerin-expressing cells. Therefore, although resident APCs that acquire tumor antigens may induce tolerance in naive cells in the absence of further stimuli, they can play an important role in promoting antitumor immunity during the course of T-cell therapy. It is possible that further therapeutic benefit will result from improving the activation status of these APCs.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive , Animals , Antigens, Surface/immunology , Cell Line, Tumor , Lectins, C-Type/immunology , Lymphoma/therapy , Mannose-Binding Lectins/immunology , Mice, Inbred C57BL , Mice, Transgenic , Spleen/cytology , Spleen/immunology
6.
J Immunol ; 195(3): 821-31, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26078270

ABSTRACT

The function of dendritic cells (DCs) can be modulated through multiple signals, including recognition of pathogen-associated molecular patterns, as well as signals provided by rapidly activated leukocytes in the local environment, such as innate-like T cells. In this article, we addressed the possibility that the roles of different murine DC subsets in cross-priming CD8(+) T cells can change with the nature and timing of activatory stimuli. We show that CD8α(+) DCs play a critical role in cross-priming CD8(+) T cell responses to circulating proteins that enter the spleen in close temporal association with ligands for TLRs and/or compounds that activate NKT cells. However, if NKT cells are activated first, then CD8α(-) DCs become conditioned to respond more vigorously to TLR ligation, and if triggered directly, these cells can also contribute to priming of CD8(+) T cell responses. In fact, the initial activation of NKT cells can condition multiple DC subsets to respond more effectively to TLR ligation, with plasmacytoid DCs making more IFN-α and both CD8α(+) and CD8α(-) DCs manufacturing more IL-12. These results suggest that different DC subsets can contribute to T cell priming if provided appropriately phased activatory stimuli, an observation that could be factored into the design of more effective vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Animals , Antigen Presentation/immunology , Antigens, Surface/genetics , Interferon-alpha/biosynthesis , Interferon-alpha/immunology , Interleukin-12/biosynthesis , Lectins, C-Type/genetics , Mannose-Binding Lectins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spleen/immunology , Toll-Like Receptors/immunology
7.
J Leukoc Biol ; 96(6): 1001-10, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25170118

ABSTRACT

This study tests the hypothesis that CD8α(+) DCs in the spleen of mice contain an immature precursor for functionally mature, "classical" cross-presenting CD8α(+) DCs. The lymphoid tissues contain a network of phenotypically distinct DCs with unique roles in surveillance and immunity. Splenic CD8α(+) DCs have been shown to exhibit a heightened capacity for phagocytosis of cellular material, secretion of IL-12, and cross-priming of CD8(+) T cells. However, this population can be subdivided further on the basis of expression of both langerin/CD207 and CX(3)CR1. We therefore evaluated the functional capacities of these different subsets. The CX(3)CR1(+) CD8α(+) DC subset does not express langerin and does not exhibit the classical features above. The CX(3)CR1(-) CD8α(+) DC can be divided into langerin-positive and negative populations, both of which express DEC205, Clec9A, and high basal levels of CD86. However, the langerin(+) CX(3)CR1(-) CD8α(+) subset has a superior capacity for acquiring cellular material and producing IL-12 and is more susceptible to activation-induced cell death. Significantly, following purification and adoptive transfer into new hosts, the langerin(-) CX(3)CR1(-) CD8α(+) subset survives longer, up-regulates expression of langerin, and becomes more susceptible to activation-induced cell death. Last, in contrast to langerin(+) CX(3)CR1(-) CD8α(+), the langerin(-) CX(3)CR1(-) CD8α(+) are still present in Batf3(-/-) mice. We conclude that the classical attributes of CD8α(+) DC are confined primarily to the langerin(+) CX(3)CR1(-) CD8α(+) DC population and that the langerin(-) CX(3)CR1(-) subset represents a Batf3-independent precursor to this mature population.


Subject(s)
Adaptive Immunity , Antigens, Differentiation/analysis , Dendritic Cells/classification , Adoptive Transfer , Animals , Antigen Presentation , Antigens, Surface/analysis , Antigens, Surface/biosynthesis , Antigens, Surface/genetics , Basic-Leucine Zipper Transcription Factors/analysis , CD8 Antigens/analysis , CD8-Positive T-Lymphocytes/immunology , CX3C Chemokine Receptor 1 , Cell Differentiation , Cell Lineage , Cells, Cultured , Cellular Senescence , Crosses, Genetic , Dendritic Cells/chemistry , Dendritic Cells/immunology , Female , Galactosylceramides/immunology , Histocompatibility Antigens Class I/immunology , Immune Tolerance/immunology , Immunophenotyping , Interleukin-12 Subunit p40/biosynthesis , Lectins, C-Type/analysis , Lectins, C-Type/biosynthesis , Lectins, C-Type/genetics , Male , Mannose-Binding Lectins/analysis , Mannose-Binding Lectins/biosynthesis , Mannose-Binding Lectins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phagocytosis/immunology , Receptors, Chemokine/analysis , Repressor Proteins/analysis , Spleen/cytology , Spleen/immunology
8.
J Immunol Methods ; 406: 104-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24556588

ABSTRACT

Splenic langerin(+) CD8α(+) dendritic cells (DCs) have exhibited a critical role in cross-priming CD8(+) T cell responses. To further study the roles of this DC subset, a protocol for the continuous depletion of langerin(+) CD8α(+) DCs was established using the pre-existing lang-DTREGFP mouse model. Due to the fast turnover rate of splenic CD8α(+) DCs, maintaining the depletion of langerin(+) CD8α(+) DCs required multiple diphtheria toxin (DT) treatments. We found that prolonged treatment with DT did not cause weight loss, or neutrophilia, as reported in some DT-based depletion models. Therefore, the in vivo depletion of murine langerin(+) CD8α(+) DCs can be maintained over time to analyse their function during the full course of an immune response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Diphtheria Toxin/immunology , Lymphocyte Depletion , Animals , Antigen Presentation/immunology , Antigens, Surface/immunology , CD8 Antigens/immunology , Diphtheria Toxin/administration & dosage , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , Mannose-Binding Lectins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spleen/cytology , Spleen/immunology
9.
Int J Mol Med ; 25(6): 883-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20428792

ABSTRACT

The transmembrane glycoprotein CD133 is a marker commonly used for isolation and analysis of putative cancer stem-like cells. However, analysis of CD133 expression is potentially confounded by the fact that two of the commonly used anti-CD133 antibodies, AC133 and 293C, only recognize CD133 that has undergone glycosylation. Therefore, our aim was to thoroughly examine antibody recognition and mRNA expression of CD133 in glioblastoma multiforme. Glioblastoma cell lines and primary cultures obtained from resected tumor tissue were analyzed by real-time PCR, Western blot analysis, and flow cytometry for CD133, and immunofluorescence was used to determine cellular localization. The AC133 and 293C antibodies did not detect any CD133 on the surface of the glioblastoma cells despite the fact that a protein was detected using C24B9, an anti-CD133 antibody that recognizes an unglycosylated epitope. This CD133 variant was truncated ( approximately 16 kDa) and, unlike typical expression of full-length CD133 protein, was found throughout the cytoplasm instead of localized to the plasma membrane. Levels of mRNA and protein for the variant increased with stress, indicating potential for it to be a functional molecule. Because AC133 and 293C antibodies do not detect all CD133 variants in glioblastoma cells, alternate detection methods need to be utilized for complete analysis of CD133 expression and for accurately determining the relationship between CD133 and cancer stem-like cells.


Subject(s)
Antigens, CD/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Glycoproteins/metabolism , Mutant Proteins/metabolism , Peptides/metabolism , AC133 Antigen , Antibodies, Monoclonal/immunology , Antigens, CD/chemistry , Antigens, CD/genetics , Cell Line, Tumor , Epitopes/immunology , Gene Expression Regulation , Glycoproteins/chemistry , Glycoproteins/genetics , Glycosylation , Humans , Microscopy, Confocal , Peptides/chemistry , Peptides/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stress, Physiological
SELECTION OF CITATIONS
SEARCH DETAIL
...