Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
MAbs ; 15(1): 2281763, 2023.
Article in English | MEDLINE | ID: mdl-38031350

ABSTRACT

Neutrophil extracellular traps (NETs) contribute to the pathophysiology of multiple inflammatory and autoimmune diseases. Targeting the NETosis pathway has demonstrated significant therapeutic potency in various disease models. Here, we describe a first-in-class monoclonal antibody (CIT-013) with high affinity for citrullinated histones H2A and H4, which inhibits NETosis and reduces tissue NET burden in vivo with significant anti-inflammatory consequences. We provide a detailed understanding of the epitope selectivity of CIT-013. Detection of CIT-013 epitopes in rheumatoid arthritis (RA) synovium provides evidence that RA is an autoimmune disease with excessive citrullinated NETs that can be targeted by CIT-013. We show that CIT-013 acts upon the final stage of NETosis, binding to its chromatin epitopes when plasma membrane integrity is compromised to prevent NET release. Bivalency of CIT-013 is necessary for NETosis inhibition. In addition, we show that CIT-013 binding to NETs and netting neutrophils enhance their phagocytosis by macrophages in an Fc-dependent manner. This is confirmed using a murine neutrophilic airway inflammation model where a mouse variant of CIT-013 reduced tissue NET burden with significant anti-inflammatory consequences. CIT-013's therapeutic activity provides new insights for the development of NET antagonists and indicates the importance of a new emerging therapy for NET-driven diseases with unmet therapeutic needs.


Subject(s)
Antibodies, Monoclonal , Arthritis, Rheumatoid , Autoimmune Diseases , Extracellular Traps , Animals , Mice , Anti-Inflammatory Agents , Antibodies, Monoclonal/pharmacology , Arthritis, Rheumatoid/drug therapy , Autoimmune Diseases/drug therapy , Epitopes/metabolism , Histones/metabolism , Neutrophils , Anti-Citrullinated Protein Antibodies/pharmacology
2.
J Immunother Cancer ; 11(4)2023 04.
Article in English | MEDLINE | ID: mdl-37068796

ABSTRACT

BACKGROUND: Preclinical studies have firmly established the CD47-signal-regulatory protein (SIRP)α axis as a myeloid immune checkpoint in cancer, and this is corroborated by available evidence from the first clinical studies with CD47 blockers. However, CD47 is ubiquitously expressed and mediates functional interactions with other ligands as well, and therefore targeting of the primarily myeloid cell-restricted inhibitory immunoreceptor SIRPα may represent a better strategy. METHOD: We generated BYON4228, a novel SIRPα-directed antibody. An extensive preclinical characterization was performed, including direct comparisons to previously reported anti-SIRPα antibodies. RESULTS: BYON4228 is an antibody directed against SIRPα that recognizes both allelic variants of SIRPα in the human population, thereby maximizing its potential clinical applicability. Notably, BYON4228 does not recognize the closely related T-cell expressed SIRPγ that mediates interactions with CD47 as well, which are known to be instrumental in T-cell extravasation and activation. BYON4228 binds to the N-terminal Ig-like domain of SIRPα and its epitope largely overlaps with the CD47-binding site. BYON4228 blocks binding of CD47 to SIRPα and inhibits signaling through the CD47-SIRPα axis. Functional studies show that BYON4228 potentiates macrophage-mediated and neutrophil-mediated killing of hematologic and solid cancer cells in vitro in the presence of a variety of tumor-targeting antibodies, including trastuzumab, rituximab, daratumumab and cetuximab. The silenced Fc region of BYON4228 precludes immune cell-mediated elimination of SIRPα-positive myeloid cells, implying anticipated preservation of myeloid immune effector cells in patients. The unique profile of BYON4228 clearly distinguishes it from previously reported antibodies representative of agents in clinical development, which either lack recognition of one of the two SIRPα polymorphic variants (HEFLB), or cross-react with SIRPγ and inhibit CD47-SIRPγ interactions (SIRPAB-11-K322A, 1H9), and/or have functional Fc regions thereby displaying myeloid cell depletion activity (SIRPAB-11-K322A). In vivo, BYON4228 increases the antitumor activity of rituximab in a B-cell Raji xenograft model in human SIRPαBIT transgenic mice. Finally, BYON4228 shows a favorable safety profile in cynomolgus monkeys. CONCLUSIONS: Collectively, this defines BYON4228 as a preclinically highly differentiating pan-allelic SIRPα antibody without T-cell SIRPγ recognition that promotes the destruction of antibody-opsonized cancer cells. Clinical studies are planned to start in 2023.


Subject(s)
CD47 Antigen , Neoplasms , Mice , Animals , Humans , T-Lymphocytes/metabolism , Rituximab , Macrophages , Neoplasms/drug therapy , Antibodies, Neoplasm
3.
J Immunother Cancer ; 7(1): 340, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31801627

ABSTRACT

BACKGROUND: Accumulating preclinical data indicate that targeting the SIRPα/CD47 axis alone or in combination with existing targeted therapies or immune checkpoint inhibitors enhances tumor rejection. Although several CD47-targeting agents are currently in phase I clinical trials and demonstrate activity in combination therapy, high and frequent dosing was required and safety signals (acute anemia, thrombocytopenia) were recorded frequently as adverse events. Based on the restricted expression pattern of SIRPα we hypothesized that antibodies targeting SIRPα might avoid some of the concerns noted for CD47-targeting agents. METHODS: SIRPα-targeting antibodies were generated and characterized for binding to human SIRPα alleles and blockade of the interaction with CD47. Functional activity was established in vitro using human macrophages or neutrophils co-cultured with human Burkitt's lymphoma cell lines. The effect of SIRPα versus CD47 targeting on human T-cell activation was studied using an allogeneic mixed lymphocyte reaction and a Staphylococcus enterotoxin B-induced T-cell proliferation assay. Potential safety concerns of the selected SIRPα-targeting antibody were addressed in vitro using a hemagglutination assay and a whole blood cytokine release assay, and in vivo in a single-dose toxicity study in cynomolgus monkeys. RESULTS: The humanized monoclonal IgG2 antibody ADU-1805 binds to all known human SIRPα alleles, showing minimal binding to SIRPß1, while cross-reacting with SIRPγ, and potently blocking the interaction of SIRPα with CD47. Reduced FcγR binding proved critical to retaining its function towards phagocyte activation. In vitro characterization demonstrated that ADU-1805 promotes macrophage phagocytosis, with similar potency to anti-CD47 antibodies, and enhances neutrophil trogocytosis. Unlike CD47-targeting agents, ADU-1805 does not interfere with T-cell activation and is not expected to require frequent and extensive dosing due to the restricted expression of SIRPα to cells of the myeloid lineage. ADU-1805 is cross-reactive to cynomolgus monkey SIRPα and upon single-dose intravenous administration in these non-human primates (NHPs) did not show any signs of anemia, thrombocytopenia or other toxicities. CONCLUSIONS: Blocking the SIRPα-CD47 interaction via SIRPα, while similarly efficacious in vitro, differentiates ADU-1805 from CD47-targeting agents with respect to safety and absence of inhibition of T-cell activation. The data presented herein support further advancement of ADU-1805 towards clinical development.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , CD47 Antigen/antagonists & inhibitors , Immunity, Innate/drug effects , Immunomodulation/drug effects , Receptors, Immunologic/antagonists & inhibitors , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, Differentiation , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/pharmacokinetics , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Flow Cytometry , Humans , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Models, Biological , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Phagocytosis/drug effects , Phagocytosis/immunology
4.
Oncotarget ; 8(40): 67439-67456, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28978044

ABSTRACT

Immunotherapy for metastatic melanoma offers great promise but, to date, only a subset of patients have responded. There is an urgent need to identify ways of allocating patients to the most beneficial therapy, to increase survival and decrease therapy-associated morbidity and costs. Blood-based biomarkers are of particular interest because of their straightforward implementation in routine clinical care. We sought to identify markers for dendritic cell (DC) vaccine-based immunotherapy against metastatic melanoma through gene expression analysis of peripheral blood mononuclear cells. A large-scale microarray analysis of 74 samples from two treatment centers, taken directly after the first round of DC vaccination, was performed. We found that phosphatidylethanolamine binding protein 1 (PEBP1)/Raf Kinase inhibitory protein (RKIP) expression can be used to identify a significant proportion of patients who performed poorly after DC vaccination. This result was validated by q-PCR analysis on blood samples from a second cohort of 95 patients treated with DC vaccination in four different centers. We conclude that low PEBP1 expression correlates with poor overall survival after DC vaccination. Intriguingly, this was only the case for expression of PEBP1 after, but not prior to, DC vaccination. Moreover, the change in PEBP1 expression upon vaccination correlated well with survival. Further analyses revealed that PEBP1 expression positively correlated with genes involved in T cell responses but inversely correlated with genes associated with myeloid cells and aberrant inflammation including STAT3, NOTCH1, and MAPK1. Concordantly, PEBP1 inversely correlated with the myeloid/lymphoid-ratio and was suppressed in patients suffering from chronic inflammatory disease.

5.
Sci Rep ; 7(1): 6713, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28751750

ABSTRACT

Glycan-protein lateral interactions have gained increased attention as important modulators of receptor function, by regulating surface residence time and endocytosis of membrane glycoproteins. The pathogen-recognition receptor DC-SIGN is highly expressed at the membrane of antigen-presenting dendritic cells, where it is organized in nanoclusters and binds to different viruses, bacteria and fungi. We recently demonstrated that DC-SIGN N-glycans spatially restrict receptor diffusion within the plasma membrane, favoring its internalization through clathrin-coated pits. Here, we investigated the involvement of the N-glycans of DC-SIGN expressing cells on pathogen binding strengthening when interacting with Candida fungal cells by using atomic force microscope (AFM)-assisted single cell-pathogen adhesion measurements. The use of DC-SIGN mutants lacking the N-glycans as well as blocking glycan-mediated lateral interactions strongly impaired cell stiffening during pathogen binding. Our findings demonstrate for the first time the direct involvement of the cell membrane glycans in strengthening cell-pathogen interactions. This study, therefore, puts forward a possible role for the glycocalyx as extracellular cytoskeleton contributing, possibly in connection with the intracellular actin cytoskeleton, to optimize strengthening of cell-pathogen interactions in the presence of mechanical forces.


Subject(s)
Candida albicans/chemistry , Cell Adhesion Molecules/chemistry , Dendritic Cells/microbiology , Glycocalyx/chemistry , Host-Pathogen Interactions , Lectins, C-Type/chemistry , Polysaccharides/chemistry , Receptors, Cell Surface/chemistry , Animals , Binding Sites , Biomechanical Phenomena , CHO Cells , Candida albicans/metabolism , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cricetulus , Dendritic Cells/metabolism , Dendritic Cells/ultrastructure , Gene Expression , Glycocalyx/metabolism , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Microscopy, Atomic Force , Mutation , Polysaccharides/metabolism , Primary Cell Culture , Protein Domains , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Transgenes
6.
Oncotarget ; 8(12): 19879-19893, 2017 Mar 21.
Article in English | MEDLINE | ID: mdl-28186996

ABSTRACT

Effective stimulation of immune cells is crucial for the success of cancer immunotherapies. Current approaches to evaluate the efficiency of stimuli are mainly defined by known flow cytometry-based cell activation or cell maturation markers. This method however does not give a complete overview of the achieved activation state and may leave important side effects unnoticed. Here, we used an unbiased RNA sequencing (RNA-seq)-based approach to compare the capacity of four clinical-grade dendritic cell (DC) activation stimuli used to prepare DC-vaccines composed of various types of DC subsets; the already clinically applied GM-CSF and Frühsommer meningoencephalitis (FSME) prophylactic vaccine and the novel clinical grade adjuvants protamine-RNA complexes (pRNA) and CpG-P. We found that GM-CSF and pRNA had similar effects on their target cells, whereas pRNA and CpG-P induced stronger type I interferon (IFN) expression than FSME. In general, the pathways most affected by all stimuli were related to immune activity and cell migration. GM-CSF stimulation, however, also induced a significant increase of genes related to nonsense-mediated decay, indicating a possible deleterious effect of this stimulus. Taken together, the two novel stimuli appear to be promising alternatives. Our study demonstrates how RNA-seq based investigation of changes in a large number of genes and gene groups can be exploited for fast and unbiased, global evaluation of clinical-grade stimuli, as opposed to the general limited evaluation of a pre-specified set of genes, by which one might miss important biological effects that are detrimental for vaccine efficacy.


Subject(s)
Dendritic Cells/immunology , Immunotherapy/methods , Sequence Analysis, RNA/methods , Vaccines/immunology , Adjuvants, Immunologic/pharmacology , Antigens, CD1/immunology , Antigens, CD1/metabolism , Cells, Cultured , Cluster Analysis , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Flow Cytometry , Gene Expression Profiling/methods , Gene Ontology , Glycoproteins/immunology , Glycoproteins/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interferon Type I/biosynthesis , Interferon Type I/genetics , Interferon Type I/immunology , Oligodeoxyribonucleotides/immunology , Oligodeoxyribonucleotides/pharmacology , Protamines/immunology , Protamines/pharmacology , RNA/immunology , RNA/pharmacology , Transcriptome/drug effects , Transcriptome/immunology , Vaccines/therapeutic use , Viral Vaccines/immunology , Viral Vaccines/pharmacology
7.
Sci Rep ; 6: 20693, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26869100

ABSTRACT

Sphingolipids are essential constituents of the plasma membrane (PM) and play an important role in signal transduction by modulating clustering and dynamics of membrane receptors. Changes in lipid composition are therefore likely to influence receptor organisation and function, but how this precisely occurs is difficult to address given the intricacy of the PM lipid-network. Here, we combined biochemical assays and single molecule dynamic approaches to demonstrate that the local lipid environment regulates adhesion of integrin receptors by impacting on their lateral mobility. Induction of sphingomyelinase (SMase) activity reduced sphingomyelin (SM) levels by conversion to ceramide (Cer), resulting in impaired integrin adhesion and reduced integrin mobility. Dual-colour imaging of cortical actin in combination with single molecule tracking of integrins showed that this reduced mobility results from increased coupling to the actin cytoskeleton brought about by Cer formation. As such, our data emphasizes a critical role for the PM local lipid composition in regulating the lateral mobility of integrins and their ability to dynamically increase receptor density for efficient ligand binding in the process of cell adhesion.


Subject(s)
Cell Membrane/metabolism , Integrins/metabolism , Nanoparticles/chemistry , Sphingolipids/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Cell Adhesion , Cell Line , Ceramides/metabolism , Diffusion , G(M1) Ganglioside/metabolism , Humans , Lymphocyte Function-Associated Antigen-1/metabolism , Monocytes/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/metabolism
8.
PLoS One ; 11(2): e0149637, 2016.
Article in English | MEDLINE | ID: mdl-26889827

ABSTRACT

The ß2-integrin lymphocyte function-associated antigen 1 (LFA-1) plays an important role in the migration, adhesion and intercellular communication of dendritic cells (DCs). During the differentiation of human DCs from monocyte precursors, LFA-1 ligand binding capacity is completely lost, even though its expression levels were remained constant. Yet LFA-1-mediated adhesive capacity on DCs can be regained by exposing DCs to the chemokine CCL21, suggesting a high degree of regulation of LFA-1 activity during the course of DC differentiation. The molecular mechanisms underlying this regulation of LFA-1 function in DCs, however, remain elusive. To get more insight we attempted to identify specific LFA-1 binding partners that may play a role in regulating LFA-1 activity in DCs. We used highly sensitive label free quantitative mass-spectrometry to identify proteins co-immunoprecipitated (co-IP) with LFA-1 from ex vivo generated DCs. Among the potential binding partners we identified not only established components of integrin signalling pathways and cytoskeletal proteins, but also several novel LFA-1 binding partners including CD13, galectin-3, thrombospondin-1 and CD44. Further comparison to the LFA-1 interaction partners in monocytes indicated that DC differentiation was accompanied by an overall increase in LFA-1 associated proteins, in particular cytoskeletal, signalling and plasma membrane (PM) proteins. The here presented LFA-1 interactome composed of 78 proteins thus represents a valuable resource of potential regulators of LFA-1 function during the DC lifecycle.


Subject(s)
Dendritic Cells/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Protein Interaction Maps , Proteome/metabolism , Blotting, Western , CD13 Antigens/metabolism , Cell Membrane/metabolism , Computer Simulation , Galectin 3/metabolism , Humans , Hyaluronan Receptors/metabolism , Immunoprecipitation , Integrins/metabolism , Ligands , Mass Spectrometry , Monocytes/metabolism , Protein Binding , Reproducibility of Results
9.
J Immunol ; 196(1): 459-68, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26597008

ABSTRACT

Dendritic cell (DC) migration is essential for efficient host defense against pathogens and cancer, as well as for the efficacy of DC-based immunotherapies. However, the molecules that induce the migratory phenotype of DCs are poorly defined. Based on a large-scale proteome analysis of maturing DCs, we identified the GPI-anchored protein semaphorin 7A (Sema7A) as being highly expressed on activated primary myeloid and plasmacytoid DCs in human and mouse. We demonstrate that Sema7A deficiency results in impaired chemokine CCL21-driven DC migration in vivo. Impaired formation of actin-based protrusions, resulting in slower three-dimensional migration, was identified as the mechanism underlying the DC migration defect. Furthermore, we show, by atomic force microscopy, that Sema7A decreases adhesion strength to extracellular matrix while increasing the connectivity of adhesion receptors to the actin cytoskeleton. This study demonstrates that Sema7A controls the assembly of actin-based protrusions that drive DC migration in response to CCL21.


Subject(s)
Actin Cytoskeleton/metabolism , Antigens, CD/physiology , Cell Movement/physiology , Chemokine CCL21/metabolism , Dendritic Cells/physiology , Extracellular Matrix/metabolism , Semaphorins/physiology , Animals , Antigens, CD/genetics , Cell Adhesion , Cell Movement/genetics , Cells, Cultured , GPI-Linked Proteins/genetics , GPI-Linked Proteins/physiology , Humans , Mice , Mice, Knockout , Microscopy, Atomic Force , RNA Interference , RNA, Small Interfering , Semaphorins/genetics
10.
J Mol Recognit ; 28(11): 687-98, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26011000

ABSTRACT

The fungus Candida albicans is the most common cause of mycotic infections in immunocompromised hosts. Little is known about the initial interactions between Candida and immune cell receptors, such as the C-type lectin dendritic cell-specific intracellular cell adhesion molecule-3 (ICAM-3)-grabbing non-integrin (DC-SIGN), because a detailed characterization at the structural level is lacking. DC-SIGN recognizes specific Candida-associated molecular patterns, that is, mannan structures present in the cell wall of Candida. The molecular recognition mechanism is however poorly understood. We postulated that small differences in mannan-branching may result in considerable differences in the binding affinity. Here, we exploit atomic force microscope-based dynamic force spectroscopy with single Candida cells to gain better insight in the carbohydrate recognition capacity of DC-SIGN. We demonstrate that slight differences in the N-mannan structure of Candida, that is, the absence or presence of a phosphomannan side chain, results in differences in the recognition by DC-SIGN as follows: (i) it contributes to the compliance of the outer cell wall of Candida, and (ii) its presence results in a higher binding energy of 1.6 kB T. The single-bond affinity of tetrameric DC-SIGN for wild-type C. albicans is ~10.7 kB T and a dissociation constant kD of 23 µM, which is relatively strong compared with other carbohydrate-protein interactions described in the literature. In conclusion, this study shows that DC-SIGN specifically recognizes mannan patterns on C. albicans with high affinity. Knowledge on the binding pocket of DC-SIGN and its pathogenic ligands will lead to a better understanding of how fungal-associated carbohydrate structures are recognized by receptors of the immune system and can ultimately contribute to the development of new anti-fungal drugs.


Subject(s)
Candida albicans/metabolism , Cell Adhesion Molecules/metabolism , Lectins, C-Type/metabolism , Receptors, Cell Surface/metabolism , Binding Sites/physiology , Carbohydrates , Cell Adhesion/physiology , Cell Wall/metabolism , Ligands , Mannans/metabolism , Microscopy, Atomic Force/methods , Protein Binding/physiology , Spectrum Analysis/methods
11.
J Cell Sci ; 127(Pt 5): 1052-1064, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24424029

ABSTRACT

Dendritic cells sample the environment for antigens and play an important role in establishing the link between innate and acquired immunity. Dendritic cells contain mechanosensitive adhesive structures called podosomes that consist of an actin-rich core surrounded by integrins, adaptor proteins and actin network filaments. They facilitate cell migration via localized degradation of extracellular matrix. Here, we show that podosomes of human dendritic cells locate to spots of low physical resistance in the substrate (soft spots) where they can evolve into protrusive structures. Pathogen recognition receptors locate to these protrusive structures where they can trigger localized antigen uptake, processing and presentation to activate T-cells. Our data demonstrate a novel role in antigen sampling for the podosomes of dendritic cells.


Subject(s)
Antigen Presentation , Cell Surface Extensions/metabolism , Dendritic Cells/metabolism , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/ultrastructure , Endocytosis , Humans , Matrix Metalloproteinase 14/metabolism , Tubulin/metabolism
12.
Sci Signal ; 5(250): ra82, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23150881

ABSTRACT

Signaling by the serine and threonine kinase Akt (also known as protein kinase B), a pathway that is common to all eukaryotic cells, is central to cell survival, proliferation, and gene induction. We sought to elucidate the mechanisms underlying regulation of the kinase activity of Akt in the immune system. We found that the four-transmembrane protein CD37 was essential for B cell survival and long-lived protective immunity. CD37-deficient (Cd37(-/-)) mice had reduced numbers of immunoglobulin G (IgG)-secreting plasma cells in lymphoid organs compared to those in wild-type mice, which we attributed to increased apoptosis of plasma cells in the germinal centers of the spleen, areas in which B cells proliferate and are selected. CD37 was required for the survival of IgG-secreting plasma cells in response to binding of vascular cell adhesion molecule 1 to the α(4)ß(1) integrin. Impaired α(4)ß(1) integrin-dependent Akt signaling in Cd37(-/-) IgG-secreting plasma cells was the underlying cause responsible for impaired cell survival. CD37 was required for the mobility and clustering of α(4)ß(1) integrins in the plasma membrane, thus regulating the membrane distribution of α(4)ß(1) integrin necessary for activation of the Akt survival pathway in the immune system.


Subject(s)
Antigens, CD/immunology , Antigens, Neoplasm/immunology , Cell Movement/immunology , Integrin alpha4beta1/immunology , Plasma Cells/immunology , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction/immunology , Tetraspanins/immunology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Movement/genetics , Cell Survival/genetics , Cell Survival/immunology , Germinal Center/immunology , Germinal Center/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Mice , Mice, Knockout , Plasma Cells/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Spleen/immunology , Spleen/metabolism , Tetraspanins/genetics , Tetraspanins/metabolism
13.
J Biol Chem ; 287(46): 38946-55, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23019323

ABSTRACT

The C-type lectin DC-SIGN expressed on dendritic cells (DCs) facilitates capture and internalization of a plethora of different pathogens. Although it is known that DC-SIGN organizes in nanoclusters at the surface of DCs, the molecular mechanisms responsible for this well defined nanopatterning and role in viral binding remain enigmatic. By combining biochemical and advanced biophysical techniques, including optical superresolution and single particle tracking, we demonstrate that DC-SIGN intrinsic nanoclustering strictly depends on its molecular structure. DC-SIGN nanoclusters exhibited free, Brownian diffusion on the cell membrane. Truncation of the extracellular neck region, known to abrogate tetramerization, significantly reduced nanoclustering and concomitantly increased lateral diffusion. Importantly, DC-SIGN nanocluster dissolution exclusively compromised binding to nanoscale size pathogens. Monte Carlo simulations revealed that heterogeneity on nanocluster density and spatial distribution confers broader binding capabilities to DC-SIGN. As such, our results underscore a direct relationship between spatial nanopatterning, driven by intermolecular interactions between the neck regions, and receptor diffusion to provide DC-SIGN with the exquisite ability to dock pathogens at the virus length scale. Insight into how virus receptors are organized prior to virus binding and how they assemble into functional platforms for virus docking is helpful to develop novel strategies to prevent virus entry and infection.


Subject(s)
Antigen-Presenting Cells/metabolism , Cell Adhesion Molecules/metabolism , Lectins, C-Type/metabolism , Lectins/chemistry , Receptors, Cell Surface/metabolism , Animals , Binding Sites , Biophysics/methods , CHO Cells , Cell Membrane/metabolism , Clathrin/metabolism , Cricetinae , Cricetulus , Cytoplasm/metabolism , Dendritic Cells/cytology , Diffusion , Humans , Mice , Models, Biological , NIH 3T3 Cells , Protein Binding , Surface Properties , Time Factors
14.
PLoS One ; 6(7): e22328, 2011.
Article in English | MEDLINE | ID: mdl-21799824

ABSTRACT

Phagocytosis is a complex process that involves membranelipid remodeling and the attraction and retention of key effector proteins. Phagosome phenotype depends on the type of receptor engaged and can be influenced by extracellular signals. Interleukin 4 (IL-4) is a cytokine that induces the alternative activation of macrophages (MΦs) upon prolonged exposure, triggering a different cell phenotype that has an altered phagocytic capacity. In contrast, the direct effects of IL-4 during phagocytosis remain unknown. Here, we investigate the impact of short-term IL-4 exposure (1 hour) during phagocytosis of IgG-opsonized yeast particles by MΦs. By time-lapse confocal microscopy of GFP-tagged lipid-sensing probes, we show that IL-4 increases the negative charge of the phagosomal membrane by prolonging the presence of the negatively charged second messenger PI(3,4,5)P3. Biochemical assays reveal an enhanced PI3K/Akt activity upon phagocytosis in the presence of IL-4. Blocking the specific class I PI3K after the onset of phagocytosis completely abrogates the IL-4-induced changes in lipid remodeling and concomitant membrane charge. Finally, we show that IL-4 direct signaling leads to a significantly prolonged retention profile of the signaling molecules Rac1 and Rab5 to the phagosomal membrane in a PI3K-dependent manner. This protracted early phagosome phenotype suggests an altered maturation, which is supported by the delayed phagosome acidification measured in the presence of IL-4. Our findings reveal that molecular differences in IL-4 levels, in the extracellular microenvironment, influence the coordination of lipid remodeling and protein recruitment, which determine phagosome phenotype and, eventually, fate. Endosomal and phagosomal membranes provide topological constraints to signaling molecules. Therefore, changes in the phagosome phenotype modulated by extracellular factors may represent an additional mechanism that regulates the outcome of phagocytosis and could have significant impact on the net biochemical output of a cell.


Subject(s)
Interleukin-4/pharmacology , Membrane Lipids/metabolism , Phagosomes/drug effects , Phagosomes/metabolism , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Line , Hydrogen-Ion Concentration , Immunoglobulin G/immunology , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Membrane Lipids/chemistry , Mice , Phagocytosis/drug effects , Phagosomes/enzymology , Phosphatidylinositols/metabolism , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Time Factors , Zymosan/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...