Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Viruses ; 16(5)2024 05 14.
Article En | MEDLINE | ID: mdl-38793659

Respiratory syncytial virus (RSV) is the most prevalent cause of acute lower respiratory infection in young children. Currently, the first RSV vaccines are approved by the FDA. Recently, N6-methyladenosine (m6A) RNA methylation has been implicated in the regulation of the viral life cycle and replication of many viruses, including RSV. m6A methylation of RSV RNA has been demonstrated to promote replication and prevent anti-viral immune responses by the host. Whether m6A is also involved in viral entry and whether m6A can also affect RSV infection via different mechanisms than methylation of viral RNA is poorly understood. Here, we identify m6A reader YTH domain-containing protein 1 (YTHDC1) as a novel negative regulator of RSV infection. We demonstrate that YTHDC1 abrogates RSV infection by reducing the expression of RSV entry receptor CX3C motif chemokine receptor 1 (CX3CR1) on the cell surface of lung epithelial cells. Altogether, these data reveal a novel role for m6A methylation and YTHDC1 in the viral entry of RSV. These findings may contribute to the development of novel treatment options to control RSV infection.


Adenosine , CX3C Chemokine Receptor 1 , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Virus Internalization , Humans , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/physiology , Adenosine/analogs & derivatives , Adenosine/metabolism , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Virus Replication , Methylation , Down-Regulation , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , Epithelial Cells/virology , Epithelial Cells/metabolism , Cell Line , A549 Cells , RNA, Viral/genetics , RNA, Viral/metabolism , Host-Pathogen Interactions , Nerve Tissue Proteins
2.
J Leukoc Biol ; 111(2): 367-377, 2022 02.
Article En | MEDLINE | ID: mdl-33884657

Inhibitory and activating immune receptors play a key role in modulating the amplitude and duration of immune responses during infection and in maintaining immune balance in homeostatic conditions. The CD200 Receptor (CD200R) gene family in humans encodes one inhibitory receptor, CD200R1, and one putative activating member, CD200R1 Like (CD200R1L). It is demonstrated that CD200R1L is endogenously expressed by human neutrophils and activates cellular functions such as reactive oxygen species (ROS) production via Syk, PI3Kß, PI3Kδ, and Rac GTPase signaling. Phylogenetic analysis shows that CD200R1L is present in many species among vertebrates, ranging from birds to primates, suggesting that evolutionary conservation of this receptor is critical for protection against co-evolving pathogens. The duplication event that generated CD200R1L from CD200R occurred several times throughout evolution, supporting convergent evolution of CD200R1L. In our phylogenetic trees, CD200R1L has longer branch lengths than CD200R1 in most species, suggesting that CD200R1L is evolving faster than CD200R1. It is proposed that CD200R1L represents a hitherto uncharacterized activating receptor on human neutrophils.


Evolution, Molecular , Neutrophils/metabolism , Orexin Receptors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Syk Kinase/metabolism , rac GTP-Binding Proteins/metabolism , Antibodies, Monoclonal/immunology , Humans , Interleukin-8/metabolism , Neutrophils/cytology , Neutrophils/immunology , Orexin Receptors/genetics , Phosphatidylinositol 3-Kinases/genetics , Phylogeny , Reactive Oxygen Species/metabolism , Syk Kinase/genetics , rac GTP-Binding Proteins/genetics
3.
Neuron ; 110(4): 613-626.e9, 2022 02 16.
Article En | MEDLINE | ID: mdl-34921782

The current paradigm is that inflammatory pain passively resolves following the cessation of inflammation. Yet, in a substantial proportion of patients with inflammatory diseases, resolution of inflammation is not sufficient to resolve pain, resulting in chronic pain. Mechanistic insight into how inflammatory pain is resolved is lacking. Here, we show that macrophages actively control resolution of inflammatory pain remotely from the site of inflammation by transferring mitochondria to sensory neurons. During resolution of inflammatory pain in mice, M2-like macrophages infiltrate the dorsal root ganglia that contain the somata of sensory neurons, concurrent with the recovery of oxidative phosphorylation in sensory neurons. The resolution of pain and the transfer of mitochondria requires expression of CD200 receptor (CD200R) on macrophages and the non-canonical CD200R-ligand iSec1 on sensory neurons. Our data reveal a novel mechanism for active resolution of inflammatory pain.


Macrophages , Sensory Receptor Cells , Animals , Ganglia, Spinal/metabolism , Humans , Macrophages/metabolism , Mice , Mitochondria , Pain/metabolism , Sensory Receptor Cells/metabolism
4.
Sci Signal ; 14(704): eabb4324, 2021 Oct 12.
Article En | MEDLINE | ID: mdl-34637328

CD200 receptor 1 (CD200R) is an inhibitory immunoreceptor that suppresses Toll-like receptor (TLR)­induced cytokine production through the adaptor protein Dok2 and the GTPase activating protein (GAP) p120-RasGAP, which can be cleaved during mild cellular stress. We found that in the presence of cleaved p120-RasGAP, CD200R lost its capacity to inhibit phosphorylation of ribosomal S6 protein (rpS6), suggesting the reduced activity of mammalian target of rapamycin complex 1 (mTORC1). Furthermore, treatment of human peripheral blood mononuclear cells (PBMC) with interferon-α (IFN-α) resulted in increased amounts of cleaved p120-RasGAP. Upon pretreatment of cells with increasing concentrations of IFN-α, CD200R switched from inhibiting to potentiating the TLR7- and TLR8-induced expression of the gene encoding IFN-γ, a cytokine that is important for innate and adaptive immunity and is implicated in systemic lupus erythematosus (SLE) pathogenesis. PBMC from patients with SLE, a prototypic type I IFN disease, had an increased abundance of cleaved p120-RasGAP compared to that in cells from healthy controls. In a subset of SLE patients, CD200R stopped functioning as an inhibitory receptor or potentiated TLR-induced IFNG mRNA expression. Thus, our data suggest that type I IFN rewires CD200R signaling to be proinflammatory, which could contribute to the perpetuation of inflammation in patients with SLE.


Interferon Type I , Leukocytes, Mononuclear , Adaptor Proteins, Signal Transducing/metabolism , Humans , Interferon Type I/genetics , Interferon-alpha , Leukocytes, Mononuclear/metabolism , Signal Transduction
5.
PLoS One ; 16(3): e0244770, 2021.
Article En | MEDLINE | ID: mdl-33780466

The inhibitory signaling of CD200 receptor 1 (CD200R) has been attributed to its NPxY signaling motif. However, NPxY-motifs are present in multiple protein families and are mostly known to mediate protein trafficking between subcellular locations rather than signaling. Therefore, we investigated whether additional motifs specify the inhibitory function of CD200R. We performed phylogenetic analysis of the intracellular domain of CD200R in mammals, birds, bony fish, amphibians and reptiles. Indeed, the tyrosine of the NPxY-motif is fully conserved across species, in line with its central role in CD200R signaling. In contrast, P295 of the NPxY-motif is not conserved. Instead, a conserved stretch of negatively charged amino acids, EEDE279, and two conserved residues P285 and K292 in the flanking region prior to the NPxY-motif are required for CD200R mediated inhibition of p-Erk, p-Akt308, p-Akt473, p-rpS6 and LPS-induced IL-8 secretion. Altogether, we show that instead of the more common NPxY-motif, CD200R signaling can be assigned to a unique signaling motif in mammals defined by: EEDExxPYxxYxxKxNxxY.


Orexin Receptors/metabolism , Signal Transduction , Amino Acid Motifs , Animals , Cell Line , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Mutagenesis, Site-Directed , Orexin Receptors/chemistry , Orexin Receptors/classification , Orexin Receptors/genetics , Phosphorylation , Phylogeny , Protein Domains , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Tyrosine/metabolism
6.
Cell Immunol ; 357: 104199, 2020 11.
Article En | MEDLINE | ID: mdl-32942189

Signal Inhibitory Receptor on Leukocytes-1 (SIRL-1) is expressed on human blood monocytes and granulocytes and inhibits myeloid effector functions. On monocytes, but not granulocytes, SIRL-1 expression is low or absent in individuals with the single nucleotide polymorphism (SNP) rs612529C. The expression of SIRL-1 in tissue and the influence of rs612529 hereon is currently unknown. Here, we used flow cytometry to determine SIRL-1 expression on immune cells in human blood and three barrier tissues; skin, colon and lung. SIRL-1 was expressed by virtually all neutrophils and eosinophils in these tissues. In contrast, SIRL-1 was not expressed by monocyte-derived cells in skin and colon, whereas it was highly expressed by lung classical monocytes. Lung monocytes from individuals with a rs612529C allele had decreased SIRL-1 expression, consistent with the genotype association in blood. Within the different monocyte subsets in blood and lung, SIRL-1 expression was highest in classical monocytes and lowest in nonclassical monocytes. SIRL-1 was not expressed by dendritic cells in blood and barrier tissues. Together, these results indicate that SIRL-1 is differentially expressed on phagocyte subsets in blood and barrier tissues, and that its expression on monocytes is genotype- and tissue-specific. Immune regulation of monocytes by SIRL-1 may be of particular importance in the lung.


Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Adult , Colon/cytology , Colon/metabolism , Eosinophils/immunology , Female , Flow Cytometry/methods , Humans , Leukocytes/immunology , Leukocytes, Mononuclear/immunology , Lung/cytology , Lung/metabolism , Male , Monocytes/immunology , Monocytes/metabolism , Mononuclear Phagocyte System/immunology , Neutrophils/immunology , Phagocytes/immunology , Phagocytes/metabolism , Skin/cytology , Skin/metabolism
7.
Sci Rep ; 7(1): 6529, 2017 07 26.
Article En | MEDLINE | ID: mdl-28747804

A wide variety of microbial and inflammatory factors induce DNA release from neutrophils as neutrophil extracellular traps (NETs). Consensus on the kinetics and mechanism of NET release has been hindered by the lack of distinctive methods to specifically quantify NET release in time. Here, we validate and refine a semi-automatic live imaging approach for quantification of NET release. Importantly, our approach is able to correct for neutrophil input and distinguishes NET release from neutrophil death by other means, aspects that are lacking in many NET quantification methods. Real time visualization shows that opsonized S. aureus rapidly induces cell death by toxins, while actual NET formation occurs after 90 minutes, similar to the kinetics of NET release by immune complexes and PMA. Inhibition of SYK, PI3K and mTORC2 attenuates NET release upon challenge with physiological stimuli but not with PMA. In contrast, neutrophils from chronic granulomatous disease patients show decreased NET release only in response to PMA. With this refined method, we conclude that NET release in primary human neutrophils is dependent on the SYK-PI3K-mTORC2 pathway and that PMA stimulation should be regarded as mechanistically distinct from NET formation induced by natural triggers.


Cytological Techniques/methods , Extracellular Traps/metabolism , Intravital Microscopy/methods , Neutrophils/metabolism , Humans , Neutrophils/immunology , Signal Transduction , Staphylococcus aureus/immunology , Time Factors
8.
Nat Rev Rheumatol ; 12(10): 593-604, 2016 10.
Article En | MEDLINE | ID: mdl-27539666

The recent success of immune checkpoint blockade in cancer therapy illustrates the importance of the inhibitory receptors cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) and programmed cell death protein 1 (PD1) in the regulation of antitumour immune responses. However, blocking signalling by these inhibitory immune checkpoint receptors is also associated with substantial inflammatory effects that can resemble autoimmune responses, which is consistent with the role of these receptors in protecting the host from excessive inflammation. The human genome encodes over 300 inhibitory receptors, which represent as many opportunities to modulate inflammation in a disease-specific and tissue-specific manner. We argue that rheumatologists and oncologists should join forces to study these inhibitory immune molecules. An improved understanding of these immune checkpoints will enable both fields to make progress in exploiting inhibitory immune receptors therapeutically. In this Review, we discuss data from studies reporting the adverse inflammatory effects of cancer therapies that target immune checkpoints. We discuss the potential implications of these findings on the biological understanding of autoimmune rheumatic diseases and highlight therapeutic strategies that could be used to target inhibitory receptors for the treatment of these conditions.


Antirheumatic Agents/therapeutic use , CTLA-4 Antigen/antagonists & inhibitors , Cell Cycle Checkpoints/drug effects , Immunotherapy , Neoplasms , Rheumatic Diseases/therapy , Antirheumatic Agents/immunology , CTLA-4 Antigen/immunology , Cell Cycle Checkpoints/immunology , Evidence-Based Medicine , Humans , Immunotherapy/methods , Inflammation/drug therapy , Molecular Targeted Therapy/methods , Neoplasms/therapy , Programmed Cell Death 1 Receptor/drug effects , Rheumatic Diseases/immunology , Treatment Outcome
9.
Nat Commun ; 5: 5074, 2014 Oct 03.
Article En | MEDLINE | ID: mdl-25278262

Dendritic cells (DCs) orchestrate antibody-mediated responses to combat extracellular pathogens including parasites by initiating T helper cell differentiation. Here we demonstrate that carbohydrate-specific signalling by DC-SIGN drives follicular T helper cell (TFH) differentiation via IL-27 expression. Fucose, but not mannose, engagement of DC-SIGN results in activation of IKKε, which collaborates with type I IFNR signalling to induce formation and activation of transcription factor ISGF3. Notably, ISGF3 induces expression of IL-27 subunit p28, and subsequent IL-27 secreted by DC-SIGN-primed DCs is pivotal for the induction of Bcl-6(+)CXCR5(+)PD-1(hi)Foxp1(lo) TFH cells, IL-21 secretion by TFH cells and T-cell-dependent IgG production by B cells. Thus, we have identified an essential role for DC-SIGN-induced ISGF3 by fucose-based PAMPs in driving IL-27 and subsequent TFH polarization, which might be harnessed for vaccination design.


Cell Adhesion Molecules/metabolism , Dendritic Cells/cytology , Fucose/chemistry , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interleukin-27/metabolism , Lectins, C-Type/metabolism , Receptors, Cell Surface/metabolism , T-Lymphocytes, Helper-Inducer/cytology , Amino Acid Motifs , B-Lymphocytes/cytology , Cell Differentiation , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Dimerization , Flow Cytometry , Humans , Immunoglobulin G/chemistry , Interferon Regulatory Factor-7/metabolism , Leukocytes, Mononuclear/cytology , Lymphocyte Activation/immunology , Mannose/chemistry , Proto-Oncogene Proteins c-bcl-6 , RNA Interference , Signal Transduction
10.
Retrovirology ; 11: 52, 2014 Jul 02.
Article En | MEDLINE | ID: mdl-24990163

BACKGROUND: Sexual transmission is the main route of HIV-1 infection and the CCR5-using (R5) HIV-1 is predominantly transmitted, even though CXCR4-using (X4) HIV-1 is often abundant in chronic HIV-1 patients. The mechanisms underlying this tropism selection are unclear. Mucosal Langerhans cells (LCs) are the first immune cells to encounter HIV-1 and here we investigated the role of LCs in selection of R5 HIV-1 using an ex vivo epidermal and vaginal transmission models. RESULTS: Immature LCs were productively infected by X4 as well as R5 HIV-1. However, only R5 but not X4 viruses were selectively transmitted by immature LCs to T cells. Transmission of HIV-1 was depended on de novo production of HIV-1 in LCs, since it could be inhibited by CCR5 fusion inhibitors as well as reverse transcription inhibitors. Notably, the activation state of LCs affected the restriction in X4 HIV-1 transmission; immune activation by TNF facilitated transmission of X4 as well as R5 HIV-1. CONCLUSIONS: These data suggest that LCs play a crucial role in R5 selection and that immature LCs effectively restrict X4 at the level of transmission.


HIV Infections/transmission , HIV-1/physiology , Langerhans Cells/physiology , Receptors, CXCR4/physiology , Humans , Langerhans Cells/virology , Receptors, CXCR4/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Virus Replication
11.
Nat Immunol ; 13(3): 246-54, 2012 Jan 22.
Article En | MEDLINE | ID: mdl-22267217

Production of the proinflammatory cytokine interleukin 1ß (IL-1ß) by dendritic cells is crucial in host defense. Here we identify a previously unknown role for dectin-1 in the activation of a noncanonical caspase-8 inflammasome in response to fungi and mycobacteria. Dectin-1 induced both the production and maturation of IL-1ß through signaling routes mediated by the kinase Syk. Whereas the CARD9-Bcl-10-MALT1 scaffold directed IL1B transcription, the recruitment of MALT1-caspase-8 and ASC into this scaffold was crucial for processing of pro-IL-1ß by caspase-8. In contrast to activation of the canonical caspase-1 inflammasome, which requires additional activation of cytosolic receptors, activation of the noncanonical caspase-8 inflammasome was independent of pathogen internalization. Thus, dectin-1 acted as an extracellular sensor for pathogens that induced both IL-1ß production and maturation through a noncanonical caspase-8-dependent inflammasome for protective immunity.


Caspase 8/immunology , Inflammasomes/immunology , Interleukin-1beta/immunology , Lectins, C-Type/immunology , Candida albicans/immunology , Enzyme Activation , Extracellular Space/immunology , Humans , Lectins, C-Type/metabolism , Mycobacterium/immunology , Signal Transduction
12.
Eur J Immunol ; 41(9): 2619-31, 2011 Sep.
Article En | MEDLINE | ID: mdl-21739428

Langerhans cells (LCs) are a subset of DCs that reside in the upper respiratory tract and are ideally suited to sense respiratory virus infections. Measles virus (MV) is a highly infectious lymphotropic and myelotropic virus that enters the host via the respiratory tract. Here, we show that human primary LCs are capable of capturing MV through the C-type lectin Langerin. Both immature and mature LCs presented MV-derived antigens in the context of HLA class II to MV-specific CD4(+) T cells. Immature LCs were not susceptible to productive infection by MV and did not present endogenous viral antigens in the context of HLA class I. In contrast, mature LCs could be infected by MV and presented de novo synthesized viral antigens to MV-specific CD8(+) T cells. Notably, neither immature nor mature LCs were able to cross-present exogenous UV-inactivated MV or MV-infected apoptotic cells. The lack of direct infection of immature LCs, and the inability of both immature and mature LCs to cross-present MV antigens, suggest that human LCs may not be directly involved in priming MV-specific CD8(+) T cells. Immune activation of LCs seems a prerequisite for MV infection of LCs and subsequent CD8(+) T-cell priming via the endogenous antigen presentation pathway.


Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Langerhans Cells/metabolism , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Measles virus/immunology , Measles/immunology , Receptors, Virus/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cell Line , Cross-Priming/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Langerhans Cells/immunology , Langerhans Cells/pathology , Langerhans Cells/virology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lymphocyte Activation , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/immunology , Measles/virology , Measles virus/pathogenicity , Mice , Receptors, Virus/genetics , Receptors, Virus/immunology , Respiratory System/pathology , Transgenes/genetics
13.
Curr Opin HIV AIDS ; 6(5): 348-52, 2011 Sep.
Article En | MEDLINE | ID: mdl-21743322

PURPOSE OF REVIEW: This review summarizes the current knowledge of innate signaling events that are involved in HIV-1 infection. We here focus on dendritic cells, which are among the first cells that encounter HIV-1 after exposure. RECENT FINDINGS: HIV-1 triggers multiple pattern recognition receptors on dendritic cells that facilitate infection and transmission to T cells. Triggering of the C-type lectin DC-SIGN induces signals that promote HIV-1 replication in dendritic cells and transmission to T cells. Similarly, dendritic cell immunoreceptor has been shown to bind HIV-1 and facilitate transmission to T cells. The cytosolic sensors TRIM5 and cyclophilin A recognize capsid proteins and activate antiviral responses to prevent HIV-1 infection. Moreover, activation of mammalian target of rapamycin (mTOR) by HIV downregulates autophagy preventing adaptive immune responses. SUMMARY: Dendritic cells express an array of pattern recognition receptors that are involved in HIV-1 infection. However, HIV-1 dampens signaling by these receptors leading to suppressed responses or takes advantage of their signaling for its own benefit.


Dendritic Cells/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , HIV-1/pathogenicity , Immunity, Innate , Signal Transduction , Humans , Receptors, Immunologic/immunology
14.
Nat Immunol ; 11(5): 419-26, 2010 May.
Article En | MEDLINE | ID: mdl-20364151

Pattern-recognition receptors (PRRs) elicit antiviral immune responses to human immunodeficiency virus type 1 (HIV-1). Here we show that HIV-1 required signaling by the PRRs Toll-like receptor 8 (TLR8) and DC-SIGN for replication in dendritic cells (DCs). HIV-1 activated the transcription factor NF-kappaB through TLR8 to initiate the transcription of integrated provirus by RNA polymerase II (RNAPII). However, DC-SIGN signaling was required for the generation of full-length viral transcripts. Binding of the HIV-1 envelope glycoprotein gp120 to DC-SIGN induced kinase Raf-1-dependent phosphorylation of the NF-kappaB subunit p65 at Ser276, which recruited the transcription-elongation factor pTEF-b to nascent transcripts. Transcription elongation and generation of full-length viral transcripts was dependent on pTEF-b-mediated phosphorylation of RNAPII at Ser2. Inhibition of either pathway abrogated replication and prevented HIV-1 transmission. Thus, HIV-1 subverts crucial components of the immune system for replication that might be targeted to prevent infection and dissemination.


Dendritic Cells/metabolism , HIV Infections/immunology , HIV-1/physiology , Immunity, Innate , Toll-Like Receptor 8/metabolism , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/virology , HIV-1/pathogenicity , Humans , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , Positive Transcriptional Elongation Factor B/metabolism , Protein Binding/genetics , Protein Engineering , Proto-Oncogene Proteins c-raf/metabolism , RNA Polymerase II/metabolism , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Second Messenger Systems/genetics , Second Messenger Systems/immunology , Sequence Deletion/genetics , Toll-Like Receptor 8/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology , Virus Replication/drug effects , Virus Replication/genetics , Virus Replication/immunology
15.
Immunol Cell Biol ; 88(4): 410-5, 2010.
Article En | MEDLINE | ID: mdl-20309013

Langerhans cells (LC) are the first dendritic cells (DC) to encounter pathogens entering the body via mucosa or skin. Equipped with pattern recognition receptors (PRR), LC are able to detect and respond to these pathogens. An important PRR, in human exclusively expressed by LC, is the C-type lectin Langerin. Langerin forms a protective barrier against HIV-1 infection by binding and degradation of this virus. In addition, antigens targeted to Langerin are presented to T cells to induce an adaptive immune response. Therefore Langerin functions as an innate anti-viral defense mechanism and an antigen receptor involved in adaptive immune responses. Here, we review the role of Langerin in antiviral protection, its structural similarity to other C-type lectins, and how its function and polymorphisms influence the function of Langerhans cells.


Antigens, CD/metabolism , Langerhans Cells/immunology , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Receptors, Virus/metabolism , Antigens, CD/pharmacology , Antiviral Agents/pharmacology , HIV-1/drug effects , Humans , Mannose-Binding Lectins/pharmacology , Receptors, HIV/metabolism
16.
Nat Immunol ; 10(10): 1081-8, 2009 Oct.
Article En | MEDLINE | ID: mdl-19718030

Cooperation between different innate signaling pathways induced by pattern-recognition receptors (PRRs) on dendritic cells (DCs) is crucial for tailoring adaptive immunity to pathogens. Here we show that carbohydrate-specific signaling through the C-type lectin DC-SIGN tailored cytokine production in response to distinct pathogens. DC-SIGN was constitutively associated with a signalosome complex consisting of the scaffold proteins LSP1, KSR1 and CNK and the kinase Raf-1. Mannose-expressing Mycobacterium tuberculosis and human immunodeficiency virus type 1 (HIV-1) induced the recruitment of effector proteins to the DC-SIGN signalosome to activate Raf-1, whereas fucose-expressing pathogens such as Helicobacter pylori actively dissociated the KSR1-CNK-Raf-1 complex from the DC-SIGN signalosome. This dynamic regulation of the signalosome by mannose- and fucose-expressing pathogens led to the enhancement or suppression of proinflammatory responses, respectively. Our study reveals another level of plasticity in tailoring adaptive immunity to pathogens.


Carbohydrates/immunology , Cell Adhesion Molecules/immunology , HIV-1/immunology , Helicobacter pylori/immunology , Lectins, C-Type/immunology , Mycobacterium tuberculosis/immunology , Receptors, Cell Surface/immunology , Signal Transduction/immunology , Blotting, Western , Cell Adhesion Molecules/metabolism , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fucose/immunology , Humans , Lectins, C-Type/metabolism , Mannose/immunology , Microfilament Proteins/immunology , Microfilament Proteins/metabolism , Protein Kinases/immunology , Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf/immunology , Proto-Oncogene Proteins c-raf/metabolism , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
Nat Immunol ; 10(2): 203-13, 2009 Feb.
Article En | MEDLINE | ID: mdl-19122653

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.


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
18.
PLoS Pathog ; 4(4): e1000049, 2008 Apr 18.
Article En | MEDLINE | ID: mdl-18421379

Measles virus (MV) is among the most infectious viruses that affect humans and is transmitted via the respiratory route. In macaques, MV primarily infects lymphocytes and dendritic cells (DCs). Little is known about the initial target cell for MV infection. Since DCs bridge the peripheral mucosal tissues with lymphoid tissues, we hypothesize that DCs are the initial target cells that capture MV in the respiratory tract and transport the virus to the lymphoid tissues where MV is transmitted to lymphocytes. Recently, we have demonstrated that the C-type lectin DC-SIGN interacts with MV and enhances infection of DCs in cis. Using immunofluorescence microscopy, we demonstrate that DC-SIGN+ DCs are abundantly present just below the epithelia of the respiratory tract. DC-SIGN+ DCs efficiently present MV-derived antigens to CD4+ T-lymphocytes after antigen uptake via either CD150 or DC-SIGN in vitro. However, DC-SIGN+ DCs also mediate transmission of MV to CD4+ and CD8+ T-lymphocytes. We distinguished two different transmission routes that were either dependent or independent on direct DC infection. DC-SIGN and CD150 are both involved in direct DC infection and subsequent transmission of de novo synthesized virus. However, DC-SIGN, but not CD150, mediates trans-infection of MV to T-lymphocytes independent of DC infection. Together these data suggest a prominent role for DCs during the initiation, dissemination, and clearance of MV infection.


Antigens, CD/physiology , CD4-Positive T-Lymphocytes/virology , Cell Adhesion Molecules/physiology , Dendritic Cells/virology , Lectins, C-Type/physiology , Measles virus/physiology , Nerve Tissue Proteins/physiology , Receptors, Cell Surface/physiology , Animals , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Chlorocebus aethiops , Dendritic Cells/immunology , Disease Models, Animal , Host-Pathogen Interactions , Humans , Monocytes , Signaling Lymphocytic Activation Molecule Family Member 1 , Vero Cells
...