Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 36
1.
Int J Mol Sci ; 23(23)2022 Nov 25.
Article En | MEDLINE | ID: mdl-36499067

Human milk oligosaccharides (HMOs) and their most abundant component, 2'-Fucosyllactose (2'-FL), are known to be immunomodulatory. Previously, it was shown that HMOs and 2'-FL bind to the C-type lectin receptor DC-SIGN. Here we show, using a ligand-receptor competition assay, that a whole mixture of HMOs from pooled human milk (HMOS) and 2'-FL inhibit the binding of the carbohydrate-binding receptor DC-SIGN to its prototypical ligands, fucose and the oligosaccharide Lewis-B, (Leb) in a dose-dependent way. Interestingly, such inhibition by HMOS and 2'-FL was not detected for another C-type lectin, langerin, which is evolutionarily similar to DC-SIGN. The cell-ligand competition assay using DC-SIGN expressing cells confirmed that 2'-FL inhibits the binding of DC-SIGN to Leb. Molecular dynamic (MD) simulations show that 2'-FL exists in a preorganized bioactive conformation before binding to DC-SIGN and this conformation is retained after binding to DC-SIGN. Leb has more flexible conformations and utilizes two binding modes, which operate one at a time via its two fucoses to bind to DC-SIGN. Our hypothesis is that 2'-FL may have a reduced entropic penalty due to its preorganized state, compared to Leb, and it has a lower binding enthalpy, suggesting a better binding to DC-SIGN. Thus, due to the better binding to DC-SIGN, 2'-FL may replace Leb from its binding pocket in DC-SIGN. The MD simulations also showed that 2'-FL does not bind to langerin. Our studies confirm 2'-FL as a specific ligand for DC-SIGN and suggest that 2'-FL can replace other DC-SIGN ligands from its binding pocket during the ligand-receptor interactions in possible immunomodulatory processes.


Lectins, C-Type , Milk, Human , Trisaccharides , Humans , Fucose/analysis , Lectins, C-Type/metabolism , Ligands , Milk, Human/metabolism , Receptors, Cell Surface/metabolism , Trisaccharides/pharmacology
2.
Commun Biol ; 5(1): 41, 2022 01 11.
Article En | MEDLINE | ID: mdl-35017635

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most aggressive malignancies with a 5-year survival rate of only 9%. Despite the fact that changes in glycosylation patterns during tumour progression have been reported, no systematic approach has been conducted to evaluate its potential for patient stratification. By analysing publicly available transcriptomic data of patient samples and cell lines, we identified here two specific glycan profiles in PDAC that correlated with progression, clinical outcome and epithelial to mesenchymal transition (EMT) status. These different glycan profiles, confirmed by glycomics, can be distinguished by the expression of O-glycan fucosylated structures, present only in epithelial cells and regulated by the expression of GALNT3. Moreover, these fucosylated glycans can serve as ligands for DC-SIGN positive tumour-associated macrophages, modulating their activation and inducing the production of IL-10. Our results show mechanisms by which the glyco-code contributes to the tolerogenic microenvironment in PDAC.


Carcinoma, Pancreatic Ductal , Glycoproteins , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/immunology , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/immunology , Glycoproteins/metabolism , Glycosylation , Humans , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Polysaccharides/chemistry , Polysaccharides/genetics , Polysaccharides/immunology , Polysaccharides/metabolism
3.
Front Immunol ; 12: 702359, 2021.
Article En | MEDLINE | ID: mdl-34276695

It was previously shown that secretion of PE-PGRS and PPE-MPTR proteins is abolished in clinical M. tuberculosis isolates with a deletion in the ppe38-71 operon, which is associated with increased virulence. Here we investigate the proteins dependent on PPE38 for their secretion and their role in the innate immune response using temporal proteomics and protein turnover analysis in a macrophage infection model. A decreased pro-inflammatory response was observed in macrophages infected with PPE38-deficient M. tuberculosis CDC1551 as compared to wild type bacteria. We could show that dampening of the pro-inflammatory response is associated with activation of a RelB/p50 pathway, while the canonical inflammatory pathway is active during infection with wild type M. tuberculosis CDC1551. These results indicate a molecular mechanism by which M. tuberculosis PE/PPE proteins controlled by PPE38 have an effect on modulating macrophage responses through NF-kB signalling.


Antigens, Bacterial/immunology , Macrophages/immunology , NF-kappa B/immunology , Tuberculosis/immunology , Virulence Factors/immunology , Humans , Inflammation/immunology , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Signal Transduction/immunology , THP-1 Cells , Virulence/immunology
4.
Article En | MEDLINE | ID: mdl-33839052

Snakebite is classified as a priority Neglected Tropical Disease by the World Health Organization. Understanding the pathology of individual snake venom toxins is of great importance when developing more effective snakebite therapies. Snake venoms may induce a range of pathologies, including haemolytic activity. Although snake venom-induced erythrocyte lysis is not the primary cause of mortality, haemolytic activity can greatly debilitate victims and contributes to systemic haemotoxicity. Current assays designed for studying haemolytic activity are not suitable for rapid screening of large numbers of toxic compounds. Consequently, in this study, a high-throughput haemolytic assay was developed that allows profiling of erythrocyte lysis, and was validated using venom from a number of medically important snake species (Calloselasma rhodostoma, Daboia russelii, Naja mossambica, Naja nigricollis and Naja pallida). The assay was developed in a format enabling direct integration into nanofractionation analytics, which involves liquid chromatographic separation of venom followed by high-resolution fractionation and subsequent bioassaying (and optional proteomics analysis), and parallel mass spectrometric detection. Analysis of the five snake venoms via this nanofractionation approach involving haemolytic assaying provided venom-cytotoxicity profiles and enabled identification of the toxins responsible for haemolytic activity. Our results show that the elapid snake venoms (Naja spp.) contained both direct and indirect lytic toxins, while the viperid venoms (C. rhodostoma and D. russelii) only showed indirect lytic activities, which required the addition of phospholipids to exert cytotoxicity on erythrocytes. The haemolytic venom toxins identified were mainly phospholipase A2s and cytotoxic three finger toxins. Finally, the applicability of this new analytical method was demonstrated using a conventional snakebite antivenom treatment and a small-molecule drug candidate to assess neutralisation of venom cytotoxins.


Erythrocytes/drug effects , Hemolysis/drug effects , High-Throughput Screening Assays/methods , Nanotechnology/methods , Snake Venoms , Animals , Chemical Fractionation , Chromatography, Liquid , Humans , Mass Spectrometry , Phospholipases A2 , Snake Venoms/chemistry , Snake Venoms/toxicity , Snakes
5.
Nat Commun ; 12(1): 1270, 2021 02 24.
Article En | MEDLINE | ID: mdl-33627655

Changes in glycosylation during tumour progression are a key hallmark of cancer. One of the glycan moieties generally overexpressed in cancer are sialic acids, which can induce immunomodulatory properties via binding to Siglec receptors. We here show that Pancreatic Ductal Adenocarcinoma (PDAC) tumour cells present an increased sialylation that can be recognized by Siglec-7 and Siglec-9 on myeloid cells. We identified the expression of the α2,3 sialyltransferases ST3GAL1 and ST3GAL4 as main contributor to the synthesis of ligands for Siglec-7 and Siglec-9 in tumour cells. Analysing the myeloid composition in PDAC, using single cell and bulk transcriptomics data, we identified monocyte-derived macrophages as contributors to the poor clinical outcome. Tumour-derived sialic acids dictate monocyte to macrophage differentiation via signalling through Siglec-7 and Siglec-9. Moreover, triggering of Siglec-9 in macrophages reduce inflammatory programmes, while increasing PD-L1 and IL-10 expression, illustrating that sialic acids modulate different myeloid cells. This work highlights a critical role for sialylated glycans in controlling immune suppression and provides new potential targets for cancer immunotherapy in PDAC.


Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Lectins/metabolism , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , Monocytes/metabolism , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Sialic Acids/pharmacology , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Cell Differentiation/drug effects , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Lectins/genetics , Macrophages/drug effects , Monocytes/drug effects , Phosphorylation/drug effects , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Pancreatic Neoplasms
6.
Immunother Adv ; 1(1): ltab012, 2021 Jan.
Article En | MEDLINE | ID: mdl-35919745

Dendritic cells (DCs) are key in the initiation of the adaptive T cell responses to tailor adequate immunity that corresponds to the type of pathogen encountered. Oppositely, DCs control the resolution phase of inflammation and are able to induce tolerance after receiving anti-inflammatory cytokines or upon encounter of self-associated molecular patterns, such as α2-3 linked sialic acid (α2-3sia). OBJECTIVE: We here investigated whether α2-3sia, that bind immune inhibitory Siglec receptors, would alter signaling and reprogramming of LPS-stimulated human monocyte-derived DCs (moDCs). METHODS AND RESULTS: Transcriptomic analysis of moDCs stimulated with α2-3sia-conjugated dendrimers revealed differentially expressed genes related to metabolic pathways, cytokines, and T cell differentiation. An increase in genes involved in ATPase regulator activity, oxidoreductase activity, and glycogen metabolic processes was detected. Metabolic extracellular flux analysis confirmed a more energetic moDC phenotype upon α2-3sia binding as evidenced by an increase in both glycolysis and mitochondrial oxidative phosphorylation. TH1 differentiation promoting genes IFNL and IL27, were significantly downregulated in the presence of α2-3sia. Functional assays confirmed that α2-3sia binding to moDCs induced phosphorylation of Siglec-9, reduced production of inflammatory cytokines IL-12 and IL-6, and increased IL-10. Surprisingly, α2-3sia-differentiated moDCs promoted FoxP3+CD25+/-CD127- regulatory T cell differentiation and decreased FoxP3-CD25-CD127- effector T cell proliferation. CONCLUSIONS: In conclusion, we demonstrate that α2-3sia binding to moDCs, phosphorylates Siglec-9, alters metabolic pathways, cytokine signaling, and T cell differentiation processes in moDCs and promotes regulatory T cells. The sialic acid-Siglec axis on DCs is therefore, a novel target to induce tolerance and to explore for immunotherapeutic interventions aimed to restore inflammatory processes.

7.
Chemistry ; 27(8): 2742-2752, 2021 Feb 05.
Article En | MEDLINE | ID: mdl-33090600

The protein myelin oligodendrocyte glycoprotein (MOG) is a key component of myelin and an autoantigen in the disease multiple sclerosis (MS). Post-translational N-glycosylation of Asn31 of MOG seems to play a key role in modulating the immune response towards myelin. This is mediated by the interaction of Lewis-type glycan structures in the N-glycan of MOG with the DC-SIGN receptor on dendritic cells (DCs). Here, we report the synthesis of an unnatural Lewis X (LeX )-containing Fmoc-SPPS-compatible asparagine building block (SPPS=solid-phase peptide synthesis), as well as asparagine building blocks containing two LeX -derived oligosaccharides: LacNAc and Fucα1-3GlcNAc. These building blocks were used for the glycosylation of the immunodominant portion of MOG (MOG31-55 ) and analyzed with respect to their ability to bind to DC-SIGN in different biological setups, as well as their ability to inhibit the citrullination-induced aggregation of MOG31-55 . Finally, a cytokine secretion assay was carried out on human monocyte-derived DCs, which showed the ability of the neoglycopeptide decorated with a single LeX to alter the balance of pro- and anti-inflammatory cytokines, inducing a tolerogenic response.


Asparagine/metabolism , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Immunomodulation , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein/chemistry , Myelin-Oligodendrocyte Glycoprotein/metabolism , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Asparagine/chemistry , Cell Adhesion Molecules/genetics , Humans , Lectins, C-Type/genetics , Ligands , Multiple Sclerosis/metabolism , Myelin-Oligodendrocyte Glycoprotein/immunology , Receptors, Cell Surface/genetics
8.
ACS Cent Sci ; 6(9): 1602-1616, 2020 Sep 23.
Article En | MEDLINE | ID: mdl-32999936

The gut microbiota guide the development of the host immune system by setting a systemic threshold for immune activation. Lipopolysaccharides (LPSs) from gut bacteria are able to trigger systemic and local proinflammatory and immunomodulatory responses, and this capability strongly relies on their fine structures. Up to now, only a few LPS structures from gut commensals have been elucidated; therefore, the molecular motifs that may be important for LPS-mammalian cell interactions at the gut level are still obscure. Here, we report on the full structure of the LPS isolated from one of the prominent species of the genus Bacteroides, Bacteroides vulgatus. The LPS turned out to consist of a particular chemical structure based on hypoacylated and mono-phosphorylated lipid A and with a galactofuranose-containing core oligosaccharide and an O-antigen built up of mannose and rhamnose. The evaluation of the immunological properties of this LPS on human in vitro models revealed a very interesting capability to produce anti-inflammatory cytokines and to induce a synergistic action of MD-2/TLR4- and TLR2-mediated signaling pathways.

9.
Front Cell Dev Biol ; 8: 556, 2020.
Article En | MEDLINE | ID: mdl-32760719

Langerhans cells (LCs) are antigen-presenting cells that reside in the skin. They uniquely express high levels of the C-type lectin receptor Langerin (CD207), which is an attractive target for antigen delivery in immunotherapeutic vaccination strategies against cancer. We here assess a library of 20 synthetic, well-defined mannoside clusters, built up from one, two, and three of six monomannosides, dimannosides, or trimannosides, appended to an oligopeptide backbone, for binding with Langerin using surface plasmon resonance and flow cytometric quantification. It is found that Langerin binding affinity increases with increasing number of mannosides. Hexavalent presentation of the mannosides resulted in binding affinities ranging from 3 to 12 µM. Trivalent presentation of the dimannosides and trimannosides led to Langerin affinity in the same range. The model melanoma gp100 antigenic peptide was subsequently equipped with a hexavalent cluster of the dimannosides and trimannosides as targeting moieties. Surprisingly, although the bifunctional conjugates were taken up in LCs in a Langerin-dependent manner, limited antigen presentation to cytotoxic T cells was observed. These results indicate that targeting glycan moieties on immunotherapeutic vaccines should not only be validated for target binding, but also on the continued effects on biology, such as antigen presentation to both CD8+ and CD4+ T cells.

10.
Front Immunol ; 11: 990, 2020.
Article En | MEDLINE | ID: mdl-32536918

In this study we developed a liposome-based vaccine containing palmitoylated synthetic long peptides (SLP) and alpha galactosylceramide (αGC) to specifically target dendritic cells (DC) for activation of both innate (invariant natural killer T-cells [iNKT]) and adaptive (CD8+ T-cells) players of the immune system. Combination of model tumor specific antigens (gp100/MART-1) formulated as a SLP and αGC in one liposome results in strong activation of CD8+ and iNKT, as measured by IFNγ secretion. Moreover, addition of lipo-Lewis Y (LeY) to the liposomes for C-type lectin targeting increased not only uptake by monocyte-derived dendritic cells (moDC), dermal dendritic cells and Langerhans cells but also enhanced gp100-specific CD8+ T- and iNKT cell activation by human skin-emigrated antigen presenting cells in an ex vivo explant model. Loading of moDC with liposomes containing LeY also showed priming of MART-126-35L specific CD8+ T-cells. In conclusion, chemically linking a lipid tail to a glycan-based targeting moiety and SLP combined with αGC in one liposome allows for easy generation of vaccine formulations that target multiple skin DC subsets and induce tumor antigen specific CD8+ T- and iNKT cells. These liposomes present a new vaccination strategy against tumors.


CD8-Positive T-Lymphocytes/drug effects , Cancer Vaccines/pharmacology , Dendritic Cells/drug effects , Galactosylceramides/pharmacology , Lewis Blood Group Antigens/pharmacology , Melanoma/drug therapy , Natural Killer T-Cells/drug effects , Peptides/pharmacology , Skin Neoplasms/drug therapy , Adaptive Immunity/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Galactosylceramides/immunology , Humans , Immunity, Innate/drug effects , Lewis Blood Group Antigens/immunology , Liposomes , Lymphocyte Activation/drug effects , Melanoma/immunology , Melanoma/metabolism , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Peptides/immunology , Skin/drug effects , Skin/immunology , Skin/metabolism , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Tissue Culture Techniques
11.
Front Immunol ; 11: 305, 2020.
Article En | MEDLINE | ID: mdl-32161592

Many tumors display alterations in the biosynthetic pathways of glycosylation, resulting in increased expression of specific tumor-associated glycan structures. Expression of these altered glycan structures is associated with metastasis and poor prognosis. Antigen presenting cells can recognize tumor-associated glycan structures, including the truncated O-glycan Tn antigen, via specific glycan receptors. Tn antigen-mediated activation of the C-type lectin MGL on dendritic cells induces regulatory T cells via the enhanced secretion of IL-10. Although these findings indicate that MGL engagement by glycan ligands can modulate immune responses, the impact of MGL ligation on dendritic cells is still not completely understood. Therefore, we employed RNA sequencing, GO term enrichment and pathway analysis on human monocyte-derived dendritic cells stimulated with two different MGL glycan ligands. Our analyses revealed a reduced expression of genes coding for key enzymes involved in the glycolysis pathway, TCA cycle, and oxidative phosphorylation. In concordance with this, extracellular flux analysis confirmed the decrease in glycolytic activity upon MGL triggering in human dendritic cells. To our knowledge, we are the first to report a diminished glycolytic activity of human dendritic cells upon C-type lectin stimulation. Overall, our findings highlight the impact of tumor-associated glycans on dendritic cell biology and metabolism and will increase our understanding on how glycans can shape immunity.


Acetylgalactosamine/metabolism , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Acetylgalactosamine/immunology , Dendritic Cells/immunology , Glycolysis , Glycosylation , Healthy Volunteers , Humans , Lectins, C-Type/immunology , Ligands , Oxidative Phosphorylation , Primary Cell Culture
12.
ACS Chem Biol ; 15(3): 728-739, 2020 03 20.
Article En | MEDLINE | ID: mdl-32045202

Dendritic cells (DCs) are armed with a multitude of Pattern Recognition Receptors (PRRs) to recognize pathogens and initiate pathogen-tailored T cell responses. In these responses, the maturation of DCs is key, as well as the production of cytokines that help to accomplish T cell responses. DC-SIGN is a frequently exploited PRR that can effectively be targeted with mannosylated antigens to enhance the induction of antigen-specific T cells. The natural O-mannosidic linkage is susceptible to enzymatic degradation, and its chemical sensitivity complicates the synthesis of mannosylated antigens. For this reason, (oligo)mannosides are generally introduced in a late stage of the antigen synthesis, requiring orthogonal conjugation handles for their attachment. To increase the stability of the mannosides and streamline the synthesis of mannosylated peptide antigens, we here describe the development of an acid-stable C-mannosyl lysine, which allows for the inline introduction of mannosides during solid-phase peptide synthesis (SPPS). The developed amino acid has been successfully used for the assembly of both small ligands and peptide antigen conjugates comprising an epitope of the gp100 melanoma-associated antigen and a TLR7 agonist for DC activation. The ligands showed similar internalization capacities and binding affinities as the O-mannosyl analogs. Moreover, the antigen conjugates were capable of inducing maturation, stimulating the secretion of pro-inflammatory cytokines, and providing enhanced gp100 presentation to CD8+ and CD4+ T cells, similar to their O-mannosyl counterparts. Our results demonstrate that the C-mannose lysine is a valuable building block for the generation of anticancer peptide-conjugate vaccine modalities.


Antineoplastic Agents/chemical synthesis , Cancer Vaccines/chemical synthesis , Glycopeptides/chemistry , Lysine/chemistry , Mannose/chemistry , Vaccines, Conjugate/chemistry , Antigens, Neoplasm/metabolism , Antineoplastic Agents/metabolism , Cancer Vaccines/metabolism , Cell Culture Techniques , Cytokines/metabolism , Dendritic Cells , Epitopes/chemistry , Epitopes/metabolism , Fluorescent Dyes/chemistry , Humans , Optical Imaging , T-Lymphocytes , Toll-Like Receptor 7/metabolism , Vaccines, Conjugate/metabolism , Vaccines, Synthetic/chemistry , gp100 Melanoma Antigen/metabolism
13.
Front Chem ; 7: 650, 2019.
Article En | MEDLINE | ID: mdl-31637232

Dendritic cells (DCs) are important initiators of adaptive immunity, and they possess a multitude of Pattern Recognition Receptors (PRR) to generate an adequate T cell mediated immunity against invading pathogens. PRR ligands are frequently conjugated to tumor-associated antigens in a vaccination strategy to enhance the immune response toward such antigens. One of these PPRs, DC-SIGN, a member of the C-type lectin receptor (CLR) family, has been extensively targeted with Lewis structures and mannose glycans, often presented in multivalent fashion. We synthesized a library of well-defined mannosides (mono-, di-, and tri-mannosides), based on known "high mannose" structures, that we presented in a systematically increasing number of copies (n = 1, 2, 3, or 6), allowing us to simultaneously study the effect of mannoside configuration and multivalency on DC-SIGN binding via Surface Plasmon Resonance (SPR) and flow cytometry. Hexavalent presentation of the clusters showed the highest binding affinity, with the hexa-α1,2-di-mannoside being the most potent ligand. The four highest binding hexavalent mannoside structures were conjugated to a model melanoma gp100-peptide antigen and further equipped with a Toll-like receptor 7 (TLR7)-agonist as adjuvant for DC maturation, creating a trifunctional vaccine conjugate. Interestingly, DC-SIGN affinity of the mannoside clusters did not directly correlate with antigen presentation enhancing properties and the α1,2-di-mannoside cluster with the highest binding affinity in our library even hampered T cell activation. Overall, this systematic study has demonstrated that multivalent glycan presentation can improve DC-SIGN binding but enhanced binding cannot be directly translated into enhanced antigen presentation and the sole assessment of binding affinity is thus insufficient to determine further functional biological activity. Furthermore, we show that well-defined antigen conjugates combining two different PRR ligands can be generated in a modular fashion to increase the effectiveness of vaccine constructs.

14.
Cell Mol Immunol ; 14(4): 360-370, 2017 Apr.
Article En | MEDLINE | ID: mdl-26456691

The potential of the skin immune system to generate immune responses is well established, and the skin is actively exploited as a vaccination site. Human skin contains several antigen-presenting cell subsets with specialized functions. In particular, the capacity to cross-present exogenous antigens to CD8+ T cells is of interest for the design of effective immunotherapies against viruses or cancer. Here, we show that primary human Langerhans cells (LCs) were able to cross-present a synthetic long peptide (SLP) to CD8+ T cells. In addition, modification of this SLP using antibodies against the receptor langerin, but not dectin-1, further enhanced the cross-presenting capacity of LCs through routing of internalized antigens to less proteolytic early endosome antigen 1+ early endosomes. The potency of LCs to enhance CD8+ T-cell responses could be further increased through activation of LCs with the toll-like receptor 3 ligand polyinosinic:polycytidylic acid (pI:C). Altogether, the data provide evidence that human LCs are able to cross-present antigens after langerin-mediated internalization. Furthermore, the potential for antigen modification to target LCs specifically provides a rationale for generating effective anti-tumor or anti-viral cytotoxic T lymphocyte responses.


Antigens, CD/metabolism , Antigens/metabolism , Cross-Priming/immunology , Endocytosis , Endosomes/metabolism , Langerhans Cells/metabolism , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Peptides/metabolism , Antibodies/metabolism , Cell Compartmentation , Cell Differentiation/drug effects , Cross-Priming/drug effects , Endocytosis/drug effects , Endosomes/drug effects , Humans , Langerhans Cells/cytology , Langerhans Cells/drug effects , Ligands , Poly I-C/pharmacology , Skin/metabolism , Toll-Like Receptors/metabolism
15.
Immunol Cell Biol ; 94(10): 907-916, 2016 11.
Article En | MEDLINE | ID: mdl-27550748

Interleukin (IL)-1ß has proven to be crucial in the differentiation of human and mouse Th17 cells. Although it has become evident that IL-1ß has potent IL-17-inducing effects on CD4+ T cells directly, it has not yet been explored whether IL-1ß can also prime dendritic cells (DCs) for a Th17 instruction program. Here, we show that human immature DCs exposed to IL-1ß promote IL-17 production in human memory CD4+ T cells. IL-1ß-primed DCs express high levels of CD14 that mediate IL-17 production through direct interaction with T cells. Moreover, culturing human CD4+CD45RO+ memory T cells with soluble CD14 is sufficient for the upregulation of retinoic acid-related orphan receptor-γ thymus and IL-17 production. In addition, in a human in situ model using tissue-resident skin DCs, upregulation of CD14 expression induced by IL-1ß on skin residents DCs promotes IL-17 production in memory T cells; strongly suggesting the in vivo relevance of this mechanism. Our findings uncover new roles for IL-1ß and CD14, and may therefore have important consequences for the development of new therapies for Th17-mediated autoimmune diseases and bacterial and fungal pathogenic infections.


CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/metabolism , Immunologic Memory , Inflammation/pathology , Interleukin-17/biosynthesis , Interleukin-1beta/metabolism , Lipopolysaccharide Receptors/metabolism , CD4-Positive T-Lymphocytes/drug effects , Cell Movement/drug effects , Dendritic Cells/drug effects , Humans , Immunologic Memory/drug effects , Monocytes/cytology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Peptidoglycan/pharmacology , Phenotype , Skin/pathology , Solubility , Th17 Cells/drug effects , Th17 Cells/immunology , Up-Regulation/drug effects
16.
PLoS One ; 10(11): e0143519, 2015.
Article En | MEDLINE | ID: mdl-26605924

Cutaneous antigen presenting cells (APCs) are critical for the induction and regulation of skin immune responses. The human skin contains phenotypically and functionally distinct APCs subsets that are present at two separated locations. While CD1ahigh LCs form a dense network in the epidermis, the CD14+ and CD1a+ APCs reside in the dermal compartment. A better understanding of the biology of human skin APC subsets is necessary for the improvement of vaccine strategies that use the skin as administration route. In particular, progress in the characterization of uptake and activatory receptors will certainly improve APC-targeting strategies in vaccination. Here we performed a detailed analysis of the expression and function of glycan-binding and pattern-recognition receptors in skin APC subsets. The results demonstrate that under steady state conditions human CD1a+ dermal dendritic cells (DCs) were phenotypically most mature as measured by the expression of CD83 and CD86, whereas the CD14+ cells showed a higher expression of the CLRs DC-SIGN, mannose receptor and DCIR and had potent antigen uptake capacity. Furthermore, steady state LCs showed superior antigen cross-presentation as compared to the dermal APC subsets. Our results also demonstrate that the TLR3 ligand polyribosinic-polyribocytidylic acid (pI:C) was the most potent stimulator of cytokine production by both LCs and dDCs. These studies warrant further exploration of human CD1a+ dDCs and LCs as target cells for cancer vaccination to induce anti-tumor immune responses.


Antigen-Presenting Cells/metabolism , Antigens, CD1/metabolism , Epidermal Cells , Epidermis/physiology , Langerhans Cells/metabolism , Lipopolysaccharide Receptors/metabolism , Phenotype , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Biomarkers , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Calcitonin Receptor-Like Protein/genetics , Calcitonin Receptor-Like Protein/metabolism , Cluster Analysis , Cross-Priming/immunology , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression , Gene Expression Profiling , Humans , Immunophenotyping , Inflammation Mediators , Langerhans Cells/immunology , Ligands , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
17.
J Control Release ; 216: 37-46, 2015 Oct 28.
Article En | MEDLINE | ID: mdl-26151293

Dendritic cells (DC) are attractive targets for cancer immunotherapy as they initiate strong and long-lived tumour-specific T cell responses. DC can be effectively targeted in vivo with tumour antigens by using nanocarriers such as liposomes. Cross-presentation of tumour antigens is enhanced with strong adjuvants such as TLR ligands. However, often these adjuvants have off-target effects, and would benefit from a DC-specific targeting strategy, similar to the tumour antigen. The goal of this study was to develop a strategy for specifically targeting DC with tumour antigen and adjuvant by using glycoliposomes. We have generated liposomes containing the glycan Lewis(Le)(X) which is highly specific for the C-type lectin receptor DC-SIGN expressed by DC. Le(X)-modified liposomes were taken up by human monocyte-derived DC in a DC-SIGN-specific manner. As adjuvants we incorporated the TLR ligands Pam3CySK4, Poly I:C, MPLA and R848 into liposomes and compared their adjuvant capacity on DC. Incorporation of the TLR4 ligand MPLA into glycoliposomes induced DC maturation and production of pro-inflammatory cytokines, in a DC-SIGN-specific manner, and DC activation was comparable to administration of soluble MPLA. Incorporation of MPLA into glycoliposomes significantly enhanced antigen cross-presentation of the melanoma tumour antigen gp100280-288 peptide to CD8(+) T cells compared to non-glycosylated MPLA liposomes. Importantly, antigen cross-presentation of the gp100280-288 peptide was significantly higher using MPLA glycoliposomes compared to the co-administration of soluble MPLA with glycoliposomes. Taken together, our data demonstrates that specific targeting of a gp100 tumour antigen and the adjuvant MPLA to DC-SIGN-expressing DC enhances the uptake of peptide-containing liposomes, the activation of DC, and induces tumour antigen-specific CD8(+) T cell responses. These data demonstrate that adjuvant-containing glycoliposome-based vaccines targeting DC-SIGN(+) DC represent a powerful new approach for CD8(+) T cell activation.


Dendritic Cells/drug effects , Liposomes/chemistry , Nucleic Acid Amplification Techniques/methods , T-Lymphocytes, Cytotoxic/drug effects , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/genetics , Antigen Presentation/drug effects , Antigens, Neoplasm/chemistry , CD8-Positive T-Lymphocytes/drug effects , Cytokines/biosynthesis , Drug Delivery Systems , Humans , Macrophages/drug effects , Melanoma, Experimental/genetics , Toll-Like Receptor 4/drug effects , gp100 Melanoma Antigen/drug effects
18.
J Invest Dermatol ; 135(11): 2697-2704, 2015 Nov.
Article En | MEDLINE | ID: mdl-26083554

Dendritic cells (DCs) have an important role in tumor control via the induction of tumor-specific T-cell responses and are therefore an ideal target for immunotherapy. The human skin is an attractive site for tumor vaccination as it contains various DC subsets. The simultaneous delivery of tumor antigen with an adjuvant is beneficial for cross-presentation and the induction of tumor-specific T-cell responses. We therefore developed liposomes that contain the melanoma-associated antigen glycoprotein 100280-288 peptide and Toll-like receptor 4 (TLR4) ligand monophosphoryl lipid A (MPLA) as adjuvant. These liposomes are efficiently taken up by monocyte-derived DCs, and antigen presentation to CD8(+) T cells was significantly higher with MPLA-modified liposomes as compared with non-modified liposomes or the co-administration of soluble MPLA. We used a human skin explant model to evaluate the efficiency of intradermal delivery of liposomes. Liposomes were efficiently taken up by CD1a(+) and especially CD14(+) dermal DCs. Induction of CD8(+) T-cell responses by emigrated dermal DCs was significantly higher when MPLA was incorporated into the liposomes as compared with non-modified liposomes or co-administration of soluble MPLA. Thus, the modification of antigen-carrying liposomes with TLR ligand MPLA significantly enhances tumor-specific T-cell responses by dermal DCs and is an attractive vaccination strategy in human skin.


Antigen Presentation/immunology , Antigens, Neoplasm/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Skin/drug effects , Biopsy, Needle , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/pharmacology , Cells, Cultured , Cross-Priming , Flow Cytometry , Humans , Immunohistochemistry , Immunotherapy/methods , Liposomes/pharmacology , Skin/cytology , Skin/pathology , Toll-Like Receptors/immunology
19.
J Invest Dermatol ; 135(9): 2228-2236, 2015 Sep.
Article En | MEDLINE | ID: mdl-25885805

CD14(+) dendritic cells (DCs) present in the dermis of human skin represent a large subset of dermal DCs (dDCs) that are considered macrophage-like cells with poor antigen (cross)-presenting capacity and limited migratory potential to the lymph nodes. CD14(+) dDC highly express DC-specific ICAM-3-grabbing non-integrin (DC-SIGN), a receptor containing potent endocytic capacity, facilitating intracellular routing of antigens to major histocompatibility complex I and II (MHC-I andII) loading compartments for the presentation to antigen-specific CD8(+) and CD4(+) T cells. Here we show using a human skin explant model that the in situ targeting of antigens to DC-SIGN using glycan-modified liposomes enhances the antigen-presenting capacity of CD14(+) dDCs. Intradermal vaccination of liposomes modified with the DC-SIGN-targeting glycan Lewis(X), containing melanoma antigens (MART-1 or Gp100), accumulated in CD14(+) dDCs and resulted in enhanced Gp100- or MART-1-specific CD8(+) T-cell responses. Simultaneous intradermal injection of the cytokines GM-CSF and IL-4 as adjuvant enhanced the migration of the skin DCs and increased the expression of DC-SIGN on the CD14(+) and CD1a(+) dDCs. These data demonstrate that human CD14(+) dDCs exhibit potent cross-presenting capacity when targeted in situ through DC-SIGN.


Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/immunology , Lectins, C-Type/immunology , Lipopolysaccharide Receptors/immunology , Receptors, Cell Surface/immunology , Analysis of Variance , Cell Movement , Cells, Cultured , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Lipopolysaccharide Receptors/metabolism , Liposomes/immunology , Liposomes/metabolism , Polysaccharides/immunology , Polysaccharides/metabolism , Sampling Studies
20.
J Control Release ; 203: 67-76, 2015 Apr 10.
Article En | MEDLINE | ID: mdl-25656175

Dendritic cells (DCs) and Langerhans cells (LC) are professional antigen presenting cells (APCs) that initiate humoral and cellular immune responses. Targeted delivery of antigen towards DC- or LC-specific receptors enhances vaccine efficacy. In this study, we compared the efficiency of glycan-based antigen targeting to both the human DC-specific C-type lectin receptor (CLR) DC-SIGN and the LC-specific CLR langerin. Since DC-SIGN and langerin are able to recognize the difucosylated oligosaccharide Lewis Y (Le(Y)), we prepared neoglycoconjugates bearing this glycan epitope to allow targeting of both lectins. Le(Y)-modified liposomes, with an approximate diameter of 200nm, were significantly endocytosed by DC-SIGN(+) DCs and mediated efficient antigen presentation to CD4(+) and CD8(+) T cells. Surprisingly, although langerin bound to Le(Y)-modified liposomes, LCs exposed to Le(Y)-modified liposomes could not endocytose liposomes nor mediate antigen presentation to T cells. However, LCs mediated an enhanced cross-presentation when antigen was delivered through langerin using Le(Y)-modified synthetic long peptides. In contrast, Le(Y)-modified synthetic long peptides were recognized by DC-SIGN, but did not trigger antigen internalization nor antigen cross-presentation. These data demonstrate that langerin and DC-SIGN have different size requirements for antigen uptake. Although using glycans remains an interesting option in the design of anti-cancer vaccines targeting multiple CLRs, aspects such as molecule size and conformation need to be taken in consideration.


Antigens, CD/immunology , Antigens/immunology , Cell Adhesion Molecules/immunology , Cross-Priming , Glycoconjugates/immunology , Lectins, C-Type/immunology , Liposomes/immunology , Mannose-Binding Lectins/immunology , Polysaccharides/immunology , Receptors, Cell Surface/immunology , Amino Acid Sequence , Antigen Presentation , Antigens/chemistry , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Carbohydrate Sequence , Dendritic Cells/immunology , Drug Delivery Systems , Glycoconjugates/chemistry , Glycosphingolipids/chemistry , Glycosphingolipids/immunology , Humans , Langerhans Cells/immunology , Liposomes/chemistry , Molecular Sequence Data , Peptides/chemistry , Peptides/immunology , Polysaccharides/chemistry
...