Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Nat Immunol ; 10(2): 203-13, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19122653

ABSTRACT

The C-type lectin dectin-1 activates the transcription factor NF-kappaB through a Syk kinase-dependent signaling pathway to induce antifungal immunity. Here we show that dectin-1 expressed on human dendritic cells activates not only the Syk-dependent canonical NF-kappaB subunits p65 and c-Rel, but also the noncanonical NF-kappaB subunit RelB. Dectin-1, when stimulated by the beta-glucan curdlan or by Candida albicans, induced a second signaling pathway mediated by the serine-threonine kinase Raf-1, which integrated with the Syk pathway at the point of NF-kappaB activation. Raf-1 antagonized Syk-induced RelB activation by promoting sequestration of RelB into inactive p65-RelB dimers, thereby altering T helper cell differentiation. Thus, dectin-1 activates two independent signaling pathways, one through Syk and one through Raf-1, to induce immune responses.


Subject(s)
Cell Differentiation/immunology , Enzyme Activation/immunology , Membrane Proteins/metabolism , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/cytology , Acetylation , Candida albicans/immunology , Cytokines/biosynthesis , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/immunology , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Lectins, C-Type , Membrane Proteins/immunology , Mycoses/immunology , NF-kappa B/immunology , Nerve Tissue Proteins/immunology , Phosphorylation , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-raf/immunology , Proto-Oncogene Proteins c-raf/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Syk Kinase , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
2.
Chemistry ; 27(8): 2742-2752, 2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33090600

ABSTRACT

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.


Subject(s)
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
3.
Immunol Cell Biol ; 94(10): 907-916, 2016 11.
Article in English | MEDLINE | ID: mdl-27550748

ABSTRACT

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.


Subject(s)
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
4.
Eur J Immunol ; 44(8): 2415-24, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24825342

ABSTRACT

Toll-like receptor (TLR) ligands are attractive candidate adjuvants for therapeutic cancer vaccines, since TLR signaling stimulates and tunes both humoral and cellular immune responses induced by dendritic cells (DCs). Given that human skin contains a dense network of DCs, which are easily accessible via (intra-)dermal delivery of vaccines, skin is actively explored as an antitumor vaccination site. Here we used a human skin explant model to explore the potential of TLR ligands as adjuvants for DC activation in their complex microenvironment. We show that topical application of Aldara skin cream, 5% of which comprises the TLR7 agonist imiquimod, significantly enhanced DC migration as compared with that resulting from intradermal injection of the TLR7/8 ligand R848 or the soluble form of imiquimod. Moreover, Aldara-treated DCs showed highest levels of the costimulatory molecules CD86, CD83, CD40, and CD70. Topical Aldara induced the highest production of pro-inflammatory cytokines in skin biopsies. When combined with intradermal peptide vaccination, Aldara-stimulated DCs showed enhanced cross-presentation of the melanoma antigen MART-1, which resulted in increased priming and activation of MART-1-specific CD8(+) T cells. These results point to advantageous effects of combining the topical application of Aldara with antitumor peptide vaccination.


Subject(s)
Aminoquinolines/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Toll-Like Receptor 7/immunology , Administration, Topical , Aminoquinolines/immunology , CD8-Positive T-Lymphocytes/drug effects , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Cross-Priming/immunology , Cytokines/immunology , Dendritic Cells/drug effects , Humans , Imidazoles/immunology , Imidazoles/pharmacology , Imiquimod , Injections, Intradermal , Ligands , MART-1 Antigen/immunology , Melanoma/immunology , Skin/immunology , Toll-Like Receptor 7/agonists
5.
J Immunol ; 190(7): 3338-45, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23467931

ABSTRACT

TLR agonists are attractive candidate adjuvants for therapeutic cancer vaccines as they can induce a balanced humoral and T cell-mediated immune response. With a dense network of dendritic cells (DCs) and draining lymphatics, the skin provides an ideal portal for vaccine delivery. Beside direct DC activation, TLR agonists may also induce DC activation through triggering the release of inflammatory mediators by accessory cells in the skin microenvironment. Therefore, a human skin explant model was used to explore the in vivo potential of intradermally delivered TLR agonists to stimulate Langerhans cells and dermal DCs in their natural complex tissue environment. The skin-emigrated DCs were phenotyped and analyzed for T cell stimulatory capacity. We report that, of six tested TLR-agonists, the TLR2 and -3 agonists peptidoglycan (PGN) and polyribosinic-polyribocytidylic acid (Poly I:C) were uniquely able to enhance the T cell-priming ability of skin-emigrated DCs, which, in the case of PGN, was accompanied by Th1 polarization. The enhanced priming capacity of Poly I:C-stimulated DCs was associated with a strong upregulation of appropriate costimulatory molecules, including CD70, whereas that of PGN-stimulated DCs was associated with the release of a broad array of proinflammatory cytokines. Transcriptional profiling further supported the notion that the PGN- and Poly I:C-induced effects were mediated through binding to TLR2/nucleotide-binding oligomerization domain 2 and TLR3/MDA5, respectively. These data warrant further exploration of PGN and Poly I:C, alone or in combination, as DC-targeted adjuvants for intradermal cancer vaccines.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/immunology , Peptidoglycan/administration & dosage , Poly I-C/administration & dosage , Skin/drug effects , Skin/immunology , Toll-Like Receptors/agonists , Cell Movement/drug effects , Cell Movement/immunology , Cells, Cultured , Cytokines/biosynthesis , Dendritic Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Profiling , Humans , Inflammation Mediators/metabolism , Injections, Intradermal , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/metabolism , Langerhans Cells/drug effects , Langerhans Cells/immunology , Langerhans Cells/metabolism , Ligands , Phenotype , Phosphorylation/drug effects , Skin/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Toll-Like Receptors/metabolism
6.
Commun Biol ; 5(1): 41, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35017635

ABSTRACT

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.


Subject(s)
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
7.
Infect Immun ; 79(7): 2681-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21502591

ABSTRACT

Carbohydrate mimicry between Campylobacter jejuni lipooligosaccharides (LOS) and host neural gangliosides plays a crucial role in the pathogenesis of Guillain-Barré syndrome (GBS). Campylobacter jejuni LOS may mimic various gangliosides, which affects the immunogenicity and the type of neurological deficits in GBS patients. Previous studies have shown the interaction of LOS with sialic acid-specific siglec receptors, although the functional consequences remain unknown. Cells that express high levels of siglecs include dendritic cells (DCs), which are crucial for initiation and differentiation of immune responses. We confirm that α2,3-sialylated GD1a/GM1a mimic and α2,8-sialylated GD1c mimic LOS structures interact with recombinant Sn and siglec-7, respectively. Although the linkage of the terminal sialic acid of LOS did not regulate expression of DC maturation markers, it displayed clear opposite expression levels of interleukin-12 (IL-12) and OX40L, molecules involved in DC-mediated Th cell differentiation. Accordingly, targeting DC-expressed siglec-7 with α2,8-linked sialylated LOS resulted in Th1 responses, whereas Th2 responses were induced by targeting with LOS containing α2,3-linked sialic acid. Thus, our data demonstrate for the first time that depending on the sialylated composition of Campylobacter jejuni LOS, specific Th differentiation programs are initiated, possibly through targeting of distinct DC-expressed siglecs.


Subject(s)
Campylobacter jejuni/immunology , Dendritic Cells/immunology , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , T-Lymphocytes/immunology , Campylobacter jejuni/chemistry , Campylobacter jejuni/metabolism , Carbohydrate Conformation , Cell Differentiation , Cell Line , Cell Polarity , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , G(M1) Ganglioside/chemistry , Gangliosides/chemistry , Gangliosides/immunology , Gangliosides/metabolism , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/microbiology , HEK293 Cells , Humans , Interleukin-12/biosynthesis , Interleukin-12/genetics , Interleukin-12/immunology , Lectins/metabolism , Lipopolysaccharides/chemistry , Molecular Mimicry , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/immunology , N-Acetylneuraminic Acid/metabolism , OX40 Ligand/biosynthesis , OX40 Ligand/genetics , Polymerase Chain Reaction , Sialic Acid Binding Immunoglobulin-like Lectins , T-Lymphocytes/metabolism , Th1 Cells/immunology , Th2 Cells/immunology
8.
PLoS Pathog ; 5(10): e1000625, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19834553

ABSTRACT

Gonorrhea is one of the most prevalent sexually transmitted diseases in the world. A naturally occurring variation of the terminal carbohydrates on the lipooligosaccharide (LOS) molecule correlates with altered disease states. Here, we investigated the interaction of different stable gonoccocal LOS phenotypes with human dendritic cells and demonstrate that each variant targets a different set of receptors on the dendritic cell, including the C-type lectins MGL and DC-SIGN. Neisseria gonorrhoeae LOS phenotype C constitutes the first bacterial ligand to be described for the human C-type lectin receptor MGL. Both MGL and DC-SIGN are locally expressed at the male and female genital area, the primary site of N. gonorrhoeae infection. We show that targeting of different C-type lectins with the N. gonorrhoeae LOS variants results in alterations in dendritic cell cytokine secretion profiles and the induction of distinct adaptive CD4(+) T helper responses. Whereas N. gonorrhoeae variant A with a terminal N-acetylglucosamine on its LOS was recognized by DC-SIGN and induced significantly more IL-10 production, phenotype C, carrying a terminal N-acetylgalactosamine, primarily interacted with MGL and skewed immunity towards the T helper 2 lineage. Together, our results indicate that N. gonorrhoeae LOS variation allows for selective manipulation of dendritic cell function, thereby shifting subsequent immune responses in favor of bacterial survival.


Subject(s)
Antigenic Variation/immunology , Dendritic Cells/immunology , Lipopolysaccharides/immunology , Lymphocyte Activation/immunology , Neisseria gonorrhoeae/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CHO Cells , Carbohydrate Sequence , Cells, Cultured , Cricetinae , Cricetulus , Female , Glycosylation , Humans , Immunity, Cellular/physiology , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Male , Models, Biological , Molecular Sequence Data , Phenotype
9.
J Immunol ; 183(5): 2984-94, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19648269

ABSTRACT

Tolerogenic dendritic cells (TDC) offer a promising therapeutic potential to ameliorate autoimmune diseases. Reported to inhibit adaptive immune responses, little is known about their innate immunity receptor repertoire. In this study, we compared three types of human TDC (IL-10-DC, dexamethasone (DX)-DC, and 1,25(OH)(2)D(3)-DC) by their TLR expression and response to a set of TLR ligands. TDC are endowed with the same TLR set as standard monocyte-derived dendritic cells but respond differentially to the TLR stimuli Pam3CSK4, polyinosinic-polycytidylic acid, LPS, and flagellin. TDC expressed low or no IL-12-related cytokines and remarkably elevated IL-10 levels. Interestingly, only TDC up-regulated the expression of TLR2 upon stimulation. This boosted the tolerogenic potential of these cells, because IL-10 production was up-regulated in TLR2-stimulated, LPS-primed DX-DC, whereas IL-12 and TNF-alpha secretion remained low. When comparing the TDC subsets, DX-DC and 1,25(OH)(2)D(3)-DC up-regulated TLR2 irrespective of the TLR triggered, whereas in IL-10-DC this effect was only mediated by LPS. Likewise, DX-DC and 1,25(OH)(2)D(3)-DC exhibited impaired ability to mature, reduced allostimulatory properties, and hampered capacity to induce Th1 differentiation. Therefore, both DX-DC and 1,25(OH)(2)D(3)-DC display the strongest tolerogenic and anti-inflammatory features and might be most suitable tools for the treatment of autoimmune diseases.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Immune Tolerance , Inflammation Mediators/antagonists & inhibitors , Toll-Like Receptor 2/biosynthesis , Toll-Like Receptor 2/blood , Up-Regulation/immunology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/classification , Down-Regulation/immunology , Feedback, Physiological/immunology , Flagellin/antagonists & inhibitors , Flagellin/metabolism , Humans , Inflammation Mediators/blood , Inflammation Mediators/physiology , Ligands , Lipopeptides/antagonists & inhibitors , Lipopeptides/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/metabolism , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Poly I-C/antagonists & inhibitors , Poly I-C/metabolism , Toll-Like Receptor 2/agonists
10.
Proc Natl Acad Sci U S A ; 105(49): 19474-9, 2008 Dec 09.
Article in English | MEDLINE | ID: mdl-19047644

ABSTRACT

Dendritic cells (DCs) are antigen-presenting cells that play an essential role in mucosal tolerance. They regularly encounter beneficial intestinal bacteria, but the nature of these cellular contacts and the immune responses elicited by the bacteria are not entirely elucidated. Here, we examined the interactions of Lactobacillus acidophilus NCFM and its cell surface compounds with DCs. L. acidophilus NCFM attached to DCs and induced a concentration-dependent production of IL-10, and low IL-12p70. We further demonstrated that the bacterium binds to DC-specific ICAM-3-grabbing nonintegrin (DC-SIGN), a DC- specific receptor. To identify the DC-SIGN ligand present on the bacterium, we took advantage of a generated array of L. acidophilus NCFM mutants. A knockout mutant of L. acidophilus NCFM lacking the surface (S) layer A protein (SlpA) was significantly reduced in binding to DC-SIGN. This mutant incurred a chromosomal inversion leading to dominant expression of a second S layer protein, SlpB. In the SlpB-dominant strain, the nature of the interaction of this bacterium with DCs changed dramatically. Higher concentrations of proinflammatory cytokines such as IL-12p70, TNFalpha, and IL-1beta were produced by DCs interacting with the SlpB-dominant strain compared with the parent NCFM strain. Unlike the SlpA-knockout mutant, T cells primed with L. acidophilus NCFM stimulated DCs produced more IL-4. The SlpA-DC-SIGN interaction was further confirmed as purified SlpA protein ligated directly to the DC-SIGN. In conclusion, the major S layer protein, SlpA, of L. acidophilus NCFM is the first probiotic bacterial DC-SIGN ligand identified that is functionally involved in the modulation of DCs and T cells functions.


Subject(s)
Bacterial Proteins/immunology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Lactobacillus acidophilus/immunology , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Differentiation/immunology , Cell Line , Cytokines/metabolism , Immune Tolerance/immunology , Kidney/cytology , Lactobacillus acidophilus/genetics , Lactobacillus acidophilus/metabolism , Macrophages/immunology , Macrophages/microbiology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mutagenesis , Probiotics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Transfection
11.
Immunother Adv ; 1(1): ltab012, 2021 Jan.
Article in English | MEDLINE | ID: mdl-35919745

ABSTRACT

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.

12.
Nat Commun ; 12(1): 1270, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33627655

ABSTRACT

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.


Subject(s)
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
13.
Front Immunol ; 11: 305, 2020.
Article in English | MEDLINE | ID: mdl-32161592

ABSTRACT

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.


Subject(s)
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
14.
ACS Cent Sci ; 6(9): 1602-1616, 2020 Sep 23.
Article in English | MEDLINE | ID: mdl-32999936

ABSTRACT

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.

15.
Front Cell Dev Biol ; 8: 556, 2020.
Article in English | MEDLINE | ID: mdl-32760719

ABSTRACT

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.

16.
ACS Chem Biol ; 15(3): 728-739, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32045202

ABSTRACT

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.


Subject(s)
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
17.
Infect Immun ; 77(2): 896-903, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19015250

ABSTRACT

P.69 pertactin (P.69 Prn), an adhesion molecule from the causative agent of pertussis, Bordetella pertussis, is present in cellular and most acellular vaccines that are currently used worldwide. Although both humoral immunity and cellular immunity directed against P.69 Prn have been implicated in protective immune mechanisms, the identities of CD4(+) T-cell epitopes on the P.69 Prn protein remain unknown. Here, a single I-A(d)-restricted B. pertussis conserved CD4(+) T-cell epitope at the N terminus of P.69 Prn was identified by using a BALB/c T-cell hybridoma. The epitope appeared immunodominant among four other minor strain-conserved P.69 Prn epitopes recognized after vaccination and B. pertussis infection, and it was capable of evoking a Th1/Th17-type cytokine response. B. pertussis P.69 Prn immune splenocytes did not cross-react with natural variants of the epitope as present in Bordetella parapertussis and Bordetella bronchiseptica. Finally, it was found that the immunodominant P.69 Prn epitope is broadly recognized in the human population by CD4(+) T cells in an HLA-DQ-restricted manner. During B. pertussis infection, the epitope was associated with a Th1-type CD4(+) T-cell response. Hence, this novel P.69 Prn epitope is involved in CD4(+) T-cell immunity after B. pertussis vaccination and infection in mice and, more importantly, in humans. Thus, it may provide a useful tool for the evaluation of the type, magnitude, and maintenance of B. pertussis-specific CD4(+) T-cell mechanisms in preclinical and clinical vaccine studies.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Bordetella pertussis/immunology , CD4-Positive T-Lymphocytes/immunology , Immunodominant Epitopes/immunology , Virulence Factors, Bordetella/immunology , Amino Acid Sequence , Animals , Bacterial Outer Membrane Proteins/chemistry , Cell Line , Cell Proliferation , Cytokines/metabolism , Female , HLA-DQ Antigens/immunology , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Specific Pathogen-Free Organisms , Virulence Factors, Bordetella/chemistry , Whooping Cough/immunology
18.
J Invest Dermatol ; 135(11): 2697-2704, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26083554

ABSTRACT

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.


Subject(s)
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.
PLoS One ; 10(11): e0143519, 2015.
Article in English | MEDLINE | ID: mdl-26605924

ABSTRACT

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.


Subject(s)
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
20.
J Control Release ; 203: 67-76, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25656175

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL