Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
PLoS Biol ; 22(3): e3002551, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38466773

RESUMEN

Mammals have 6 highly conserved actin isoforms with nonredundant biological functions. The molecular basis of isoform specificity, however, remains elusive due to a lack of tools. Here, we describe the development of IntAct, an internal tagging strategy to study actin isoforms in fixed and living cells. We identified a residue pair in ß-actin that permits tag integration and used knock-in cell lines to demonstrate that IntAct ß-actin expression and filament incorporation is indistinguishable from wild type. Furthermore, IntAct ß-actin remains associated with common actin-binding proteins (ABPs) and can be targeted in living cells. We demonstrate the usability of IntAct for actin isoform investigations by showing that actin isoform-specific distribution is maintained in human cells. Lastly, we observed a variant-dependent incorporation of tagged actin variants into yeast actin patches, cables, and cytokinetic rings demonstrating cross species applicability. Together, our data indicate that IntAct is a versatile tool to study actin isoform localization, dynamics, and molecular interactions.


Asunto(s)
Actinas , Proteínas de Microfilamentos , Animales , Humanos , Actinas/genética , Actinas/metabolismo , Proteínas de Microfilamentos/metabolismo , Citoesqueleto de Actina/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Citoesqueleto/metabolismo , Saccharomyces cerevisiae/metabolismo , Mamíferos/metabolismo
2.
Biomacromolecules ; 20(7): 2587-2597, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31150222

RESUMEN

Polymer brushes are extensively used for the preparation of bioactive surfaces. They form a platform to attach functional (bio)molecules and control the physicochemical properties of the surface. These brushes are nearly exclusively prepared from flexible polymers, even though much stiffer brushes from semiflexible polymers are frequently found in nature, which exert bioactive functions that are out of reach for flexible brushes. Synthetic semiflexible polymers, however, are very rare. Here, we use polyisocyanopeptides (PICs) to prepare high-density semiflexible brushes on different substrate geometries. For bioconjugation, we developed routes with two orthogonal click reactions, based on the strain-promoted azide-alkyne cycloaddition reaction and the (photoactivated) tetrazole-ene cycloaddition reaction. We found that for high brush densities, multiple bonds between the polymer and the substrate are necessary, which was achieved in a block copolymer strategy. Whether the desired biomolecules are conjugated to the PIC polymer before or after brush formation depends on the dimensions and required densities of the biomolecules and the curvature of the substrate. In either case, we provide mild, aqueous, and highly modular reaction strategies, which make PICs a versatile addition to the toolbox for generating semiflexible bioactive polymer brush surfaces.


Asunto(s)
Reacción de Cicloadición , Péptidos/química , Péptidos/síntesis química , Polimerizacion , Propiedades de Superficie
3.
J Cell Sci ; 129(2): 298-313, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26621033

RESUMEN

Podosomes are actin-rich adhesion structures that depend on Arp2/3-complex-based actin nucleation. We now report the identification of the formins FHOD1 and INF2 as novel components and additional actin-based regulators of podosomes in primary human macrophages. FHOD1 surrounds the podosome core and is also present at podosome-connecting cables, whereas INF2 localizes at the podosome cap structure. Using a variety of microscopy-based methods; including a semiautomated podosome reformation assay, measurement of podosome oscillations, FRAP analysis of single podosomes, and structured illumination microscopy, both formins were found to regulate different aspects of podosome-associated contractility, with FHOD1 mediating actomyosin contractility between podosomes, and INF2 regulating contractile events at individual podosomes. Moreover, INF2 was found to be a crucial regulator of podosome de novo formation and size. Collectively, we identify FHOD1 and INF2 as novel regulators of inter- and intra-structural contractility of podosomes. Podosomes thus present as one of the few currently identified structures which depend on the concerted activity of both Arp2/3 complex and specific formins and might serve as a model system for the analysis of complex actin architectures in cells.


Asunto(s)
Proteínas Fetales/fisiología , Proteínas de Microfilamentos/fisiología , Proteínas Nucleares/fisiología , Podosomas/fisiología , Actinas/metabolismo , Células Cultivadas , Matriz Extracelular/metabolismo , Forminas , Humanos , Macrófagos/metabolismo , Macrófagos/ultraestructura , Podosomas/ultraestructura
4.
J Immunol ; 197(7): 2715-25, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27566820

RESUMEN

Potent immunotherapies are urgently needed to boost antitumor immunity and control disease in cancer patients. As dendritic cells (DCs) are the most powerful APCs, they are an attractive means to reinvigorate T cell responses. An appealing strategy to use the effective Ag processing and presentation machinery, T cell stimulation and cross-talk capacity of natural DC subsets is in vivo tumor Ag delivery. In this context, endocytic C-type lectin receptors are attractive targeting molecules. In this study, we investigated whether CLEC12A efficiently delivers tumor Ags into human DC subsets, facilitating effective induction of CD4(+) and CD8(+) T cell responses. We confirmed that CLEC12A is selectively expressed by myeloid cells, including the myeloid DC subset (mDCs) and the plasmacytoid DC subset (pDCs). Moreover, we demonstrated that these DC subsets efficiently internalize CLEC12A, whereupon it quickly translocates to the early endosomes and subsequently routes to the lysosomes. Notably, CLEC12A Ab targeting did not negatively affect DC maturation or function. Furthermore, CLEC12A-mediated delivery of keyhole limpet hemocyanin resulted in enhanced proliferation and cytokine secretion by keyhole limpet hemocyanin-experienced CD4(+) T cells. Most importantly, CLEC12A-targeted delivery of HA-1 long peptide resulted in efficient Ag cross-presentation by mDCs and pDCs, leading to strong ex vivo activation of HA-1-specific CD8(+) T cells of patients after allogeneic stem cell transplantation. Collectively, these data indicate that CLEC12A is an effective new candidate with great potential for in vivo Ag delivery into mDCs and pDCs, thereby using the specialized functions and cross-talk capacity of these DC subsets to boost tumor-reactive T cell immunity in cancer patients.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Lectinas Tipo C/inmunología , Neoplasias/inmunología , Receptores Mitogénicos/inmunología , Células Cultivadas , Células Dendríticas/citología , Humanos
5.
J Biol Chem ; 287(46): 38946-55, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23019323

RESUMEN

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.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Moléculas de Adhesión Celular/metabolismo , Lectinas Tipo C/metabolismo , Lectinas/química , Receptores de Superficie Celular/metabolismo , Animales , Sitios de Unión , Biofisica/métodos , Células CHO , Membrana Celular/metabolismo , Clatrina/metabolismo , Cricetinae , Cricetulus , Citoplasma/metabolismo , Células Dendríticas/citología , Difusión , Humanos , Ratones , Modelos Biológicos , Células 3T3 NIH , Unión Proteica , Propiedades de Superficie , Factores de Tiempo
6.
Blood ; 118(15): 4111-9, 2011 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-21860028

RESUMEN

Targeting antigens to dendritic cell (DC)-specific receptors, such as DC-SIGN, induces potent T cell-mediated immune responses. DC-SIGN is a transmembrane C-type lectin receptor with a long extracellular neck region and a carbohydrate recognition domain (CRD). Thus far, only antibodies binding the CRD have been used to target antigens to DC-SIGN. We evaluated the endocytic pathway triggered by antineck antibodies as well as their intracellular routing and ability to induce CD8(+) T-cell activation. In contrast to anti-CRD antibodies, antineck antibodies induced a clathrin-independent mode of DC-SIGN internalization, as demonstrated by the lack of colocalization with clathrin and the observation that silencing clathrin did not affect antibody internalization in human DCs. Interestingly, we observed that anti-neck and anti-CRD antibodies were differentially routed within DCs. Whereas anti-CRD antibodies were mainly routed to late endosomal compartments, anti-neck antibodies remained associated with early endosomal compartments positive for EEA-1 and MHC class I for up to 2 hours after internalization. Finally, cross-presentation of protein antigen conjugated to antineck antibodies was approximately 1000-fold more effective than nonconjugated antigen. Our studies demonstrate that anti-neck antibodies trigger a distinct mode of DC-SIGN internalization that shows potential for targeted vaccination strategies.


Asunto(s)
Presentación de Antígeno/fisiología , Linfocitos T CD8-positivos/inmunología , Moléculas de Adhesión Celular/inmunología , Reactividad Cruzada/fisiología , Células Dendríticas/inmunología , Endosomas/inmunología , Lectinas Tipo C/inmunología , Lisosomas/inmunología , Receptores de Superficie Celular/inmunología , Animales , Anticuerpos/genética , Anticuerpos/inmunología , Antígenos/genética , Antígenos/inmunología , Células CHO , Moléculas de Adhesión Celular/genética , Cricetinae , Cricetulus , Endosomas/genética , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunidad Celular/fisiología , Lectinas Tipo C/genética , Lisosomas/genética , Ratones , Ratones Transgénicos , Estructura Terciaria de Proteína , Receptores de Superficie Celular/genética , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/inmunología
7.
Int J Nanomedicine ; 18: 1599-1612, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37013026

RESUMEN

Introduction: There has recently been a surge of interest in mesoporous bioactive glass nanoparticles (MBGNs) as multi-functional nanocarriers for application in bone-reconstructive and -regenerative surgery. Their excellent control over their structural and physicochemical properties renders these nanoparticles suitable for the intracellular delivery of therapeutic agents to combat degenerative bone diseases, such as bone infection, or bone cancer. Generally, the therapeutic efficacy of nanocarriers strongly depends on the efficacy of their cellular uptake, which is determined by numerous factors including cellular features and the physicochemical characteristics of nanocarriers, particularly surface charge. In this study, we have systematically investigated the effect of the surface charge of MBGNs doped with copper as a model therapeutic agent on cellular uptake by both macrophages and pre-osteoblast cells involved in bone healing and bone infections to guide the future design of MBGN-based nanocarriers. Methods: Cu-MBGNs with negative, neutral, and positive surface charges were synthesized and their cellular uptake efficiency was assessed. Additionally, the intracellular fate of internalized nanoparticles along with their ability to deliver therapeutic cargo was studied in detail. Results: The results showed that both cell types internalized Cu-MBGNs regardless of their surface charge, indicating that cellular uptake of nanoparticles is a complex process influenced by multiple factors. This similarity in cellular uptake was attributed to the formation of a protein corona surrounding the nanoparticles when exposed to protein-rich biological media, which masks the original nanoparticle surface. Once internalized, the nanoparticles were found to mainly colocalize with lysosomes, exposing them to a more compartmentalized and acidic environment. Furthermore, we verified that Cu-MBGNs released their ionic components (Si, Ca, and Cu ions) in both acidic and neutral environments, leading to the delivery of these therapeutic cargos intracellularly. Conclusion: The effective internalization of Cu-MBGNs and their ability to deliver cargos intracellularly highlight their potential as intracellular delivery nanocarriers for bone-regenerative and -healing applications.


Asunto(s)
Células Madre Mesenquimatosas , Nanopartículas , Nanopartículas/química , Regeneración Ósea , Cicatrización de Heridas , Vidrio/química
8.
Proc Natl Acad Sci U S A ; 106(44): 18557-62, 2009 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-19850864

RESUMEN

Recruitment of receptor proteins to lipid rafts has been proposed as an important mechanism to regulate their cellular function. In particular, rafts have been implicated in regulation of integrin-mediated cell adhesion, although the underlying mechanism remains elusive. We used single-molecule near-field optical microscopy (NSOM) with localization accuracy of approximately 3 nm, to capture the spatio-functional relationship between the integrin LFA-1 and raft components (GPI-APs) on immune cells. Dual color nanoscale imaging revealed the existence of a nanodomain GPI-AP subpopulation that further concentrated in regions smaller than 250 nm, suggesting a hierarchical prearrangement of GPI-APs on resting monocytes. We previously demonstrated that in quiescent monocytes, LFA-1 preorganizes in nanoclusters. We now show that integrin nanoclusters are spatially different but reside proximal to GPI-AP nanodomains, forming hotspots on the cell surface. Ligand-mediated integrin activation resulted in an interconversion from monomers to nanodomains of GPI-APs and the generation of nascent adhesion sites where integrin and GPI-APs colocalized at the nanoscale. Cholesterol depletion significantly affected the reciprocal distribution pattern of LFA-1 and GPI-APs in the resting state, and LFA-1 adhesion to its ligand. As such, our data demonstrate the existence of nanoplatforms as essential intermediates in nascent cell adhesion. Since raft association with a variety of membrane proteins other than LFA-1 has been documented, we propose that hotspots regions enriched with raft components and functional receptors may constitute a prototype of nanoscale inter-receptor assembly and correspond to a generic mechanism to offer cells with privileged areas for rapid cellular function and responses to the outside world.


Asunto(s)
Glicosilfosfatidilinositoles/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Monocitos/citología , Monocitos/metabolismo , Nanoestructuras/química , Adhesión Celular , Línea Celular , Membrana Celular/metabolismo , Colesterol/deficiencia , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Ligandos , Modelos Moleculares
9.
Front Immunol ; 13: 869031, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35603178

RESUMEN

Congenital disorders of glycosylation (CDG) are inherited metabolic diseases characterized by mutations in enzymes involved in different steps of protein glycosylation, leading to aberrant synthesis, attachment or processing of glycans. Recently, immunological dysfunctions in several CDG types have been increasingly documented. Despite these observations, detailed studies on immune cell dysfunction in PMM2-CDG and other CDG types are still scarce. Studying PMM2-CDG patient immune cells is challenging due to limited availability of patient material, which is a result of the low incidence of the disease and the often young age of the subjects. Dedicated immune cell models, mimicking PMM2-CDG, could circumvent many of these problems and facilitate research into the mechanisms of immune dysfunction. Here we provide initial observations about the immunophenotype and the phagocytic function of primary PMM2-CDG monocytes. Furthermore, we assessed the suitability of two different glycosylation-impaired human monocyte models: tunicamycin-treated THP-1 monocytes and PMM2 knockdown THP-1 monocytes induced by shRNAs. We found no significant differences in primary monocyte subpopulations of PMM2-CDG patients as compared to healthy individuals but we did observe anomalous surface glycosylation patterns in PMM2-CDG patient monocytes as determined using fluorescent lectin binding. We also looked at the capacity of monocytes to bind and internalize fungal particles and found a slightly increased uptake of C. albicans by PMM2-CDG monocytes as compared to healthy monocytes. Tunicamycin-treated THP-1 monocytes showed a highly decreased uptake of fungal particles, accompanied by a strong decrease in glycosylation levels and a high induction of ER stress. In contrast and despite a drastic reduction of the PMM2 enzyme activity, PMM2 knockdown THP-1 monocytes showed no changes in global surface glycosylation levels, levels of fungal particle uptake similar to control monocytes, and no ER stress induction. Collectively, these initial observations suggest that the absence of ER stress in PMM2 knockdown THP-1 cells make this model superior over tunicamycin-treated THP-1 cells and more comparable to primary PMM2-CDG monocytes. Further development and exploitation of CDG monocyte models will be essential for future in-depth studies to ultimately unravel the mechanisms of immune dysfunction in CDG.


Asunto(s)
Trastornos Congénitos de Glicosilación , Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/metabolismo , Glicosilación , Humanos , Monocitos/metabolismo , Fosfotransferasas (Fosfomutasas)/deficiencia , Tunicamicina/metabolismo , Tunicamicina/farmacología
10.
Eur J Immunol ; 39(7): 1923-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19585517

RESUMEN

Dendritic cells (DC), professional Ag-presenting cells located in mucosae and lymphoid organs, operate at the interface of innate and adaptive immunity and are likely the first cells to encounter invading HIV-1. Although the C-type lectin DC-Specific ICAM-3-grabbing non-integrin (DC-SIGN) binds to several viruses, including HIV-1, its direct involvement in viral entry remains controversial. Despite its central role in DC function, little is known about the underlying molecular mechanism(s) of DC-SIGN-mediated Ag uptake. Here, we analyzed the early stages of DC-SIGN-mediated endocytosis and demonstrate that both membrane cholesterol and dynamin are required. Confocal microscopy and clathrin RNAi showed that DC-SIGN-mediated internalization occurs via clathrin-coated pits. Electron microscopy of ultrathin sections showed the involvement of DC-SIGN in clathrin-dependent HIV-1 internalization by DC. Currently, DC-specific C-type lectins are considered potential target in anti-tumor clinical trials. Detailed information about how different Ag are internalized via these receptors will facilitate the rational design of targeted therapeutic strategies.


Asunto(s)
Antígenos/metabolismo , Moléculas de Adhesión Celular/metabolismo , Clatrina/metabolismo , VIH-1/metabolismo , Lectinas Tipo C/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Células CHO , Moléculas de Adhesión Celular/genética , Membrana Celular/metabolismo , Colesterol/metabolismo , Clatrina/genética , Cricetinae , Cricetulus , Células Dendríticas/citología , Células Dendríticas/metabolismo , Células Dendríticas/ultraestructura , Dinaminas/genética , Dinaminas/metabolismo , Endocitosis , Humanos , Lectinas Tipo C/genética , Microscopía Confocal , Microscopía Electrónica , ARN Interferente Pequeño/genética , Receptores de Superficie Celular/genética , Solubilidad , Transfección
11.
Front Immunol ; 11: 613286, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33643295

RESUMEN

Prostaglandin E2 (PGE2) is a lipid mediator that modulates the function of myeloid immune cells such as macrophages and dendritic cells (DCs) through the activation of the G protein-coupled receptors EP2 and EP4. While both EP2 and EP4 signaling leads to an elevation of intracellular cyclic adenosine monophosphate (cAMP) levels through the stimulating Gαs protein, EP4 also couples to the inhibitory Gαi protein to decrease the production of cAMP. The receptor-specific contributions to downstream immune modulatory functions are still poorly defined. Here, we employed quantitative imaging methods to characterize the early EP2 and EP4 signaling events in myeloid cells and their contribution to the dissolution of adhesion structures called podosomes, which is a first and essential step in DC maturation. We first show that podosome loss in DCs is primarily mediated by EP4. Next, we demonstrate that EP2 and EP4 signaling leads to distinct cAMP production profiles, with EP4 inducing a transient cAMP response and EP2 inducing a sustained cAMP response only at high PGE2 levels. We further find that simultaneous EP2 and EP4 stimulation attenuates cAMP production, suggesting a reciprocal control of EP2 and EP4 signaling. Finally, we demonstrate that efficient signaling of both EP2 and EP4 relies on an intact microtubule network. Together, these results enhance our understanding of early EP2 and EP4 signaling in myeloid cells. Considering that modulation of PGE2 signaling is regarded as an important therapeutic possibility in anti-tumor immunotherapy, our findings may facilitate the development of efficient and specific immune modulators of PGE2 receptors.


Asunto(s)
Microtúbulos/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal/fisiología , Animales , Línea Celular , AMP Cíclico/metabolismo , Células Dendríticas/metabolismo , Humanos , Ratones , Células Mieloides/metabolismo , Células RAW 264.7
12.
J Cell Biol ; 164(1): 145-55, 2004 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-14709546

RESUMEN

The C-type lectin dendritic cell (DC)-specific intercellular adhesion molecule grabbing non-integrin (DC-SIGN; CD209) facilitates binding and internalization of several viruses, including HIV-1, on DCs, but the underlying mechanism for being such an efficient phagocytic pathogen-recognition receptor is poorly understood. By high resolution electron microscopy, we demonstrate a direct relation between DC-SIGN function as viral receptor and its microlocalization on the plasma membrane. During development of human monocyte-derived DCs, DC-SIGN becomes organized in well-defined microdomains, with an average diameter of 200 nm. Biochemical experiments and confocal microscopy indicate that DC-SIGN microdomains reside within lipid rafts. Finally, we show that the organization of DC-SIGN in microdomains on the plasma membrane is important for binding and internalization of virus particles, suggesting that these multimolecular assemblies of DC-SIGN act as a docking site for pathogens like HIV-1 to invade the host.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Membrana Celular/metabolismo , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Infecciones por Virus ARN/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Virales/metabolismo , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/ultraestructura , Membrana Celular/ultraestructura , Membrana Celular/virología , Células Cultivadas , Células Dendríticas/ultraestructura , Células Dendríticas/virología , Endocitosis/fisiología , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , VIH-1/patogenicidad , VIH-1/fisiología , Humanos , Inmunohistoquímica , Lectinas Tipo C/inmunología , Lectinas Tipo C/ultraestructura , Microdominios de Membrana/metabolismo , Microdominios de Membrana/ultraestructura , Microscopía Electrónica , Monocitos/metabolismo , Monocitos/ultraestructura , Monocitos/virología , Estructura Terciaria de Proteína/fisiología , Infecciones por Virus ARN/inmunología , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/ultraestructura , Receptores Virales/inmunología , Receptores Virales/ultraestructura
13.
Mol Biol Cell ; 17(10): 4270-81, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16855029

RESUMEN

The beta2-integrin LFA-1 facilitates extravasation of monocytes (MOs) into the underlying tissues, where MOs can differentiate into dendritic cells (DCs). Although DCs express LFA-1, unlike MOs, they cannot bind to ICAM-1. We hypothesized that an altered integrin organization on the DC plasma membrane might cause this effect and investigated the relationship between membrane organization and function of LFA-1 on MOs and DCs. High-resolution mapping of LFA-1 surface distribution revealed that on MOs LFA-1 function is associated with a distribution in well-defined nanoclusters (100-150-nm diameter). Interestingly, a fraction of these nanoclusters contains primed LFA-1 molecules expressing the specific activation-dependent L16-epitope. Live imaging of MO-T-cell conjugates showed that only these primed nanoclusters are dynamically recruited to the cellular interface forming micrometer-sized assemblies engaged in ligand binding and linked to talin. We conclude that besides affinity regulation, LFA-1 function is controlled by at least three different avidity patterns: random distributed inactive molecules, well-defined ligand-independent proactive nanoclusters, and ligand-triggered micrometer-sized macroclusters.


Asunto(s)
Membrana Celular/metabolismo , Células Dendríticas/fisiología , Antígeno-1 Asociado a Función de Linfocito/fisiología , Monocitos/fisiología , Adhesión Celular , Agregación Celular , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Microdominios de Membrana/fisiología , Modelos Biológicos , Linfocitos T/fisiología
14.
Sci Rep ; 9(1): 3556, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30837487

RESUMEN

Local membrane phospholipid enrichment serves as docking platform for signaling proteins involved in many processes including cell adhesion and migration. Tissue-resident dendritic cells (DCs) assemble actomyosin-based structures called podosomes, which mediate adhesion and degradation of extracellular matrix for migration and antigen sampling. Recent evidence suggested the involvement of phospholipase D (PLD) and its product phosphatidic acid (PA) in podosome formation, but the spatiotemporal control of this process is poorly characterized. Here we determined the role of PLD1 and PLD2 isoforms in regulating podosome formation and dynamics in human primary DCs by combining PLD pharmacological inhibition with a fluorescent PA sensor and fluorescence microscopy. We found that ongoing PLD2 activity is required for the maintenance of podosomes, whereas both PLD1 and PLD2 control the early stages of podosome assembly. Furthermore, we captured the formation of PA microdomains accumulating at the membrane cytoplasmic leaflet of living DCs, in dynamic coordination with nascent podosome actin cores. Finally, we show that both PLD1 and PLD2 activity are important for podosome-mediated matrix degradation. Our results provide novel insight into the isoform-specific spatiotemporal regulation of PLD activity and further our understanding of the role of cell membrane phospholipids in controlling localized actin polymerization and cell protrusion.


Asunto(s)
Microdominios de Membrana/metabolismo , Ácidos Fosfatidicos/metabolismo , Fosfolipasa D/metabolismo , Podosomas/metabolismo , Transducción de Señal , Actinas/metabolismo , Células Dendríticas/citología , Humanos
15.
Nat Commun ; 10(1): 5171, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31729386

RESUMEN

Basement membrane transmigration during embryonal development, tissue homeostasis and tumor invasion relies on invadosomes, a collective term for invadopodia and podosomes. An adequate structural framework for this process is still missing. Here, we reveal the modular actin nano-architecture that enables podosome protrusion and mechanosensing. The podosome protrusive core contains a central branched actin module encased by a linear actin module, each harboring specific actin interactors and actin isoforms. From the core, two actin modules radiate: ventral filaments bound by vinculin and connected to the plasma membrane and dorsal interpodosomal filaments crosslinked by myosin IIA. On stiff substrates, the actin modules mediate long-range substrate exploration, associated with degradative behavior. On compliant substrates, the vinculin-bound ventral actin filaments shorten, resulting in short-range connectivity and a focally protrusive, non-degradative state. Our findings redefine podosome nanoscale architecture and reveal a paradigm for how actin modularity drives invadosome mechanosensing in cells that breach tissue boundaries.


Asunto(s)
Actinas/química , Actinas/metabolismo , Podosomas/metabolismo , Actinas/genética , Animales , Adhesión Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Movimiento Celular , Células Cultivadas , Células Dendríticas/química , Células Dendríticas/citología , Células Dendríticas/metabolismo , Humanos , Mecanotransducción Celular , Ratones , Podosomas/química , Podosomas/genética
16.
iScience ; 22: 240-255, 2019 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-31786520

RESUMEN

Endogenous extracellular Galectins constitute a novel mechanism of membrane protein organization at the cell surface. Although Galectins are also highly expressed intracellularly, their cytosolic functions are poorly understood. Here, we investigated the role of Galectin-9 in dendritic cell (DC) surface organization and function. By combining functional, super-resolution and atomic force microscopy experiments to analyze membrane stiffness, we identified intracellular Galectin-9 to be indispensable for plasma membrane integrity and structure in DCs. Galectin-9 knockdown studies revealed intracellular Galectin-9 to directly control cortical membrane structure by modulating Rac1 activity, providing the underlying mechanism of Galectin-9-dependent actin cytoskeleton organization. Consequent to its role in maintaining plasma membrane structure, phagocytosis studies revealed that Galectin-9 was essential for C-type-lectin receptor-mediated pathogen uptake by DCs. This was confirmed by the impaired phagocytic capacity of Galectin-9-null murine DCs. Together, this study demonstrates a novel role for intracellular Galectin-9 in modulating DC function, which may be evolutionarily conserved.

17.
J Leukoc Biol ; 81(3): 729-40, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17135574

RESUMEN

The role of dendritic cell-specific ICAM-3-grabbing nonintegrin (DC-SIGN) in DC-T cell communication was assessed by analyzing the effect of DC-SIGN-blocking mAb in MLR. The results show that the degree of inhibition by DC-SIGN and LFA-1 mAb depends on the magnitude of the MLR and the maturation status of the DC. Addition of DC-SIGN mAb at several time-points during MLR showed that DC-SIGN is involved early on in DC-T cell contacts. This initial role is masked by strong adhesive and costimulatory mechanisms, indicating a short-lived effect of DC-SIGN in DC-T cell interactions. To examine this concept in more detail, the percentage of PBL capable of binding DC-SIGN was determined. Analysis of several donors revealed that 1-20% PBL bind to beads coated with recombinant DC-SIGN, and the DC-SIGN-binding cells comprised all major cell subsets found in blood. PBL isolated from a donor with high DC-SIGN-binding capacity were more prone to blocking by DC-SIGN mAb in MLR than PBL from a donor with low DC-SIGN-binding capacity. This study indicates an initial and transient role for DC-SIGN in T cell proliferation, which becomes apparent when T cell proliferation is low and when the percentage of DC-SIGN binding PBL is high.


Asunto(s)
Moléculas de Adhesión Celular/biosíntesis , Células Dendríticas/inmunología , Lectinas Tipo C/biosíntesis , Receptores de Superficie Celular/biosíntesis , Linfocitos T/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Sitios de Unión , Moléculas de Adhesión Celular/antagonistas & inhibidores , Diferenciación Celular/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Humanos , Lectinas Tipo C/antagonistas & inhibidores , Prueba de Cultivo Mixto de Linfocitos , Antígeno-1 Asociado a Función de Linfocito/inmunología , Receptores de Superficie Celular/antagonistas & inhibidores , Relación Estructura-Actividad , Linfocitos T/citología , Linfocitos T/efectos de los fármacos
18.
Front Immunol ; 9: 1908, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30186284

RESUMEN

Podosomes are multimolecular cytoskeletal structures that coordinate the migration of tissue-resident dendritic cells (DCs). They consist of a protrusive actin-rich core and an adhesive integrin-rich ring that contains adaptor proteins such as vinculin and zyxin. Individual podosomes are typically interconnected by a dense network of actin filaments giving rise to large podosome clusters. The actin density in podosome clusters complicates the analysis of podosomes by light microscopy alone. Here, we present an optimized procedure for performing super-resolution correlative light and electron microscopy (SR-CLEM) to study the organization of multiple proteins with respect to actin in podosome clusters at the ventral plasma membrane of DCs. We demonstrate that our procedure is suited to correlate at least three colors in super-resolution Airyscan microscopy with scanning electron microscopy (SEM). Using this procedure, we first reveal an intriguing complexity in the organization of ventral and radiating actin filaments in clusters formed by DCs which was not properly detected before by light microscopy alone. Next, we demonstrate a differential organization of vinculin and zyxin with respect to the actin filaments at podosomes. While vinculin mostly resides at sites where the actin filaments connect to the cell membrane, zyxin is primarily associated with filaments close to and on top of the core. Finally, we reveal a novel actin-based structure with SEM that connects closely associated podosome cores and which may be important for podosome topography sensing. Interestingly, these interpodosomal connections, in contrast to the radiating and ventral actin filaments appear to be insensitive to inhibition of actin polymerization suggesting that these pools of actin are not dynamically coupled. Together, our work demonstrates the power of correlating different imaging modalities for studying multimolecular cellular structures and could potentially be further exploited to study processes at the ventral plasma membrane of immune cells such as clathrin-mediated endocytosis or immune synapse formation.


Asunto(s)
Células Dendríticas/metabolismo , Células Dendríticas/ultraestructura , Podosomas/metabolismo , Podosomas/ultraestructura , Actinas/química , Actinas/metabolismo , Biomarcadores , Humanos , Microscopía , Microscopía Electrónica , Unión Proteica , Multimerización de Proteína , Flujo de Trabajo
19.
Front Immunol ; 9: 2333, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30356797

RESUMEN

Activation of the T cell receptor (TCR) on the T cell through ligation with antigen-MHC complex of an antigen-presenting cell (APC) is an essential process in the activation of T cells and induction of the subsequent adaptive immune response. Upon activation, the TCR, together with its associated co-receptor CD3 complex, assembles in signaling microclusters that are transported to the center of the organizational structure at the T cell-APC interface termed the immunological synapse (IS). During IS formation, local cell surface receptors and associated intracellular molecules are reorganized, ultimately creating the typical bull's eye-shaped pattern of the IS. CD6 is a surface glycoprotein receptor, which has been previously shown to associate with CD3 and co-localize to the center of the IS in static conditions or stable T cell-APC contacts. In this study, we report the use of different experimental set-ups analyzed with microscopy techniques to study the dynamics and stability of CD6-TCR/CD3 interaction dynamics and stability during IS formation in more detail. We exploited antibody spots, created with microcontact printing, and antibody-coated beads, and could demonstrate that CD6 and the TCR/CD3 complex co-localize and are recruited into a stimulatory cluster on the cell surface of T cells. Furthermore, we demonstrate, for the first time, that CD6 forms microclusters co-localizing with TCR/CD3 microclusters during IS formation on supported lipid bilayers. These co-localizing CD6 and TCR/CD3 microclusters are both radially transported toward the center of the IS formed in T cells, in an actin polymerization-dependent manner. Overall, our findings further substantiate the role of CD6 during IS formation and provide novel insight into the dynamic properties of this CD6-TCR/CD3 complex interplay. From a methodological point of view, the biophysical approaches used to characterize these receptors are complementary and amenable for investigation of the dynamic interactions of other membrane receptors.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Fenómenos Biofísicos , Complejo CD3/metabolismo , Linfocitos T/fisiología , Actinas/química , Actinas/metabolismo , Antígenos CD/química , Antígenos de Diferenciación de Linfocitos T/química , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Sinapsis Inmunológicas/fisiología , Unión Proteica , Multimerización de Proteína , Transporte de Proteínas , Complejo Receptor-CD3 del Antígeno de Linfocito T/química , Complejo Receptor-CD3 del Antígeno de Linfocito T/metabolismo
20.
Sci Rep ; 7(1): 6713, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28751750

RESUMEN

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.


Asunto(s)
Candida albicans/química , Moléculas de Adhesión Celular/química , Células Dendríticas/microbiología , Glicocálix/química , Interacciones Huésped-Patógeno , Lectinas Tipo C/química , Polisacáridos/química , Receptores de Superficie Celular/química , Animales , Sitios de Unión , Fenómenos Biomecánicos , Células CHO , Candida albicans/metabolismo , Adhesión Celular , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Cricetulus , Células Dendríticas/metabolismo , Células Dendríticas/ultraestructura , Expresión Génica , Glicocálix/metabolismo , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Microscopía de Fuerza Atómica , Mutación , Polisacáridos/metabolismo , Cultivo Primario de Células , Dominios Proteicos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA