Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Traffic ; 24(10): 453-462, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37403269

RESUMEN

Each cell in a multicellular organism permanently adjusts the concentration of its cell surface proteins. In particular, epithelial cells tightly control the number of carriers, transporters and cell adhesion proteins at their plasma membrane. However, sensitively measuring the cell surface concentration of a particular protein of interest in live cells and in real time represents a considerable challenge. Here, we introduce a novel approach based on split luciferases, which uses one luciferase fragment as a tag on the protein of interest and the second fragment as a supplement to the extracellular medium. Once the protein of interest arrives at the cell surface, the luciferase fragments complement and generate luminescence. We compared the performance of split Gaussia luciferase and split Nanoluciferase by using a system to synchronize biosynthetic trafficking with conditional aggregation domains. The best results were achieved with split Nanoluciferase, for which luminescence increased more than 6000-fold upon recombination. Furthermore, we showed that our approach can separately detect and quantify the arrival of membrane proteins at the apical and basolateral plasma membrane in single polarized epithelial cells by detecting the luminescence signals with a microscope, thus opening novel avenues for characterizing the variations in trafficking in individual epithelial cells.


Asunto(s)
Células Epiteliales , Proteínas de la Membrana , Proteínas de la Membrana/metabolismo , Células Epiteliales/metabolismo , Membrana Celular/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Polaridad Celular
2.
EMBO Rep ; 24(4): e55971, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36856136

RESUMEN

Pseudomonas aeruginosa is a Gram-negative bacterium causing morbidity and mortality in immuno-compromised humans. It produces a lectin, LecB, that is considered a major virulence factor, however, its impact on the immune system remains incompletely understood. Here we show that LecB binds to endothelial cells in human skin and mice and disrupts the transendothelial passage of leukocytes in vitro. It impairs the migration of dendritic cells into the paracortex of lymph nodes leading to a reduced antigen-specific T cell response. Under the effect of the lectin, endothelial cells undergo profound cellular changes resulting in endocytosis and degradation of the junctional protein VE-cadherin, formation of an actin rim, and arrested cell motility. This likely negatively impacts the capacity of endothelial cells to respond to extracellular stimuli and to generate the intercellular gaps for allowing leukocyte diapedesis. A LecB inhibitor can restore dendritic cell migration and T cell activation, underlining the importance of LecB antagonism to reactivate the immune response against P. aeruginosa infection.


Asunto(s)
Pseudomonas aeruginosa , Migración Transendotelial y Transepitelial , Humanos , Animales , Ratones , Células Endoteliales/metabolismo , Lectinas/metabolismo , Lectinas/farmacología , Inmunidad
3.
Cell ; 140(4): 540-53, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20178746

RESUMEN

Nascent transport intermediates detach from donor membranes by scission. This process can take place in the absence of dynamin, notably in clathrin-independent endocytosis, by mechanisms that are yet poorly defined. We show here that in cells scission of Shiga toxin-induced tubular endocytic membrane invaginations is preceded by cholesterol-dependent membrane reorganization and correlates with the formation of membrane domains on model membranes, suggesting that domain boundary forces are driving tubule membrane constriction. Actin triggers scission by inducing such membrane reorganization process. Tubule occurrence is indeed increased upon cellular depletion of the actin nucleator component Arp2, and the formation of a cortical actin shell in liposomes is sufficient to trigger the scission of Shiga toxin-induced tubules in a cholesterol-dependent but dynamin-independent manner. Our study suggests that membranes in tubular Shiga toxin-induced invaginations are poised to undergo actin-triggered reorganization leading to scission by a physical mechanism that may function independently from or in synergy with pinchase activity.


Asunto(s)
Actinas/metabolismo , Membrana Celular/metabolismo , Endocitosis , Colesterol/metabolismo , Dinaminas/metabolismo , Células HeLa , Humanos , Toxinas Shiga/metabolismo
4.
Genes Dev ; 31(17): 1754-1769, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28982759

RESUMEN

The Bcl-2 family protein Bim triggers mitochondrial apoptosis. Bim is expressed in nonapoptotic cells at the mitochondrial outer membrane, where it is activated by largely unknown mechanisms. We found that Bim is regulated by formation of large protein complexes containing dynein light chain 1 (DLC1). Bim rapidly inserted into cardiolipin-containing membranes in vitro and recruited DLC1 to the membrane. Bim binding to DLC1 induced the formation of large Bim complexes on lipid vesicles, on isolated mitochondria, and in intact cells. Native gel electrophoresis and gel filtration showed Bim-containing mitochondrial complexes of several hundred kilodaltons in all cells tested. Bim unable to form complexes was consistently more active than complexed Bim, which correlated with its substantially reduced binding to anti-apoptotic Bcl-2 proteins. At endogenous levels, Bim surprisingly bound only anti-apoptotic Mcl-1 but not Bcl-2 or Bcl-XL, recruiting only Mcl-1 into large complexes. Targeting of DLC1 by RNAi in human cell lines induced disassembly of Bim-Mcl-1 complexes and the proteasomal degradation of Mcl-1 and sensitized the cells to the Bcl-2/Bcl-XL inhibitor ABT-737. Regulation of apoptosis at mitochondria thus extends beyond the interaction of monomers of proapoptotic and anti-apoptotic Bcl-2 family members but involves more complex structures of proteins at the mitochondrial outer membrane, and targeting complexes may be a novel therapeutic strategy.


Asunto(s)
Apoptosis/genética , Proteína 11 Similar a Bcl2/metabolismo , Dineínas/metabolismo , Mitocondrias/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Animales , Proteína 11 Similar a Bcl2/genética , Células CACO-2 , Línea Celular Tumoral , Regulación de la Expresión Génica , Células HeLa , Humanos , Células MCF-7 , Ratones , Unión Proteica , Multimerización de Proteína/genética , Estabilidad Proteica , Interferencia de ARN , Proteína X Asociada a bcl-2/genética
5.
Cell Mol Life Sci ; 79(10): 513, 2022 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-36097202

RESUMEN

The link between cancer and aberrant glycosylation has recently become evident. Glycans and their altered forms, known as tumour-associated carbohydrate antigens (TACAs), are diverse, complex and difficult to target therapeutically. Lectins are naturally occurring glycan-binding proteins  that offer a unique opportunity to recognise TACAs. T cells expressing chimeric antigen receptors (CARs) have proven to be a successful immunotherapy against leukaemias, but so far have shown limited success in solid tumours. We developed a panel of lectin-CARs that recognise the glycosphingolipid globotriaosylceramide (Gb3), which is overexpressed in various cancers, such as Burkitt's lymphoma, colorectal, breast and pancreatic. We have selected the following lectins: Shiga toxin's B-subunit from Shigella dysenteriae, LecA from Pseudomonas aeruginosa, and the engineered lectin Mitsuba from Mytilus galloprovincialis as antigen-binding domains and fused them to a well-known second-generation CAR. The Gb3-binding lectin-CARs have demonstrated target-specific cytotoxicity against Burkitt's lymphoma-derived cell lines as well as solid tumour cells from colorectal and triple-negative breast cancer. Our findings reveal the big potential of lectin-based CARs as therapeutical applications to target Gb3 and other TACAs expressed in haematological malignancies and solid tumours.


Asunto(s)
Linfoma de Burkitt , Neoplasias Colorrectales , Receptores Quiméricos de Antígenos , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/terapia , Humanos , Lectinas/metabolismo , Polisacáridos/metabolismo , Linfocitos T
6.
Angew Chem Int Ed Engl ; 62(7): e202215535, 2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36398566

RESUMEN

Bacterial adhesion, biofilm formation and host cell invasion of the ESKAPE pathogen Pseudomonas aeruginosa require the tetravalent lectins LecA and LecB, which are therefore drug targets to fight these infections. Recently, we have reported highly potent divalent galactosides as specific LecA inhibitors. However, they suffered from very low solubility and an intrinsic chemical instability due to two acylhydrazone motifs, which precluded further biological evaluation. Here, we isosterically substituted the acylhydrazones and systematically varied linker identity and length between the two galactosides necessary for LecA binding. The optimized divalent LecA ligands showed improved stability and were up to 1000-fold more soluble. Importantly, these properties now enabled their biological characterization. The lead compound L2 potently inhibited LecA binding to lung epithelial cells, restored wound closure in a scratch assay and reduced the invasiveness of P. aeruginosa into host cells.


Asunto(s)
Adhesinas Bacterianas , Pseudomonas aeruginosa , Humanos , Adhesinas Bacterianas/química , Pseudomonas aeruginosa/metabolismo , Factores de Virulencia/metabolismo , Galactósidos/química , Galactósidos/metabolismo , Galactósidos/farmacología , Adhesión Bacteriana
7.
J Transl Med ; 20(1): 578, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36494671

RESUMEN

BACKGROUND: Aberrant glycosylation patterns play a crucial role in the development of cancer cells as they promote tumor growth and aggressiveness. Lectins recognize carbohydrate antigens attached to proteins and lipids on cell surfaces and represent potential tools for application in cancer diagnostics and therapy. Among the emerging cancer therapies, immunotherapy has become a promising treatment modality for various hematological and solid malignancies. Here we present an approach to redirect the immune system into fighting cancer by targeting altered glycans at the surface of malignant cells. We developed a so-called "lectibody", a bispecific construct composed of a lectin linked to an antibody fragment. This lectibody is inspired by bispecific T cell engager (BiTEs) antibodies that recruit cytotoxic T lymphocytes (CTLs) while simultaneously binding to tumor-associated antigens (TAAs) on cancer cells. The tumor-related glycosphingolipid globotriaosylceramide (Gb3) represents the target of this proof-of-concept study. It is recognized with high selectivity by the B-subunit of the pathogen-derived Shiga toxin, presenting opportunities for clinical development. METHODS: The lectibody was realized by conjugating an anti-CD3 single-chain antibody fragment to the B-subunit of Shiga toxin to target Gb3+ cancer cells. The reactive non-canonical amino acid azidolysine (AzK) was inserted at predefined single positions in both proteins. The azido groups were functionalized by bioorthogonal conjugation with individual linkers that facilitated selective coupling via an alternative bioorthogonal click chemistry reaction. In vitro cell-based assays were conducted to evaluate the antitumoral activity of the lectibody. CTLs, Burkitt´s lymphoma-derived cells and colorectal adenocarcinoma cell lines were screened in flow cytometry and cytotoxicity assays for activation and lysis, respectively. RESULTS: This proof-of-concept study demonstrates that the lectibody activates T cells for their cytotoxic signaling, redirecting CTLs´ cytotoxicity in a highly selective manner and resulting in nearly complete tumor cell lysis-up to 93%-of Gb3+ tumor cells in vitro. CONCLUSIONS: This research highlights the potential of lectins in targeting certain tumors, with an opportunity for new cancer treatments. When considering a combinatorial strategy, lectin-based platforms of this type offer the possibility to target glycan epitopes on tumor cells and boost the efficacy of current therapies, providing an additional strategy for tumor eradication and improving patient outcomes.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Humanos , Linfocitos T Citotóxicos , Complejo CD3/metabolismo , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Biespecíficos/química , Activación de Linfocitos , Toxina Shiga , Fragmentos de Inmunoglobulinas , Muerte Celular , Lectinas
8.
Cell Microbiol ; 23(8): e13326, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33720490

RESUMEN

Photorhabdus luminescens Tc toxins are large tripartite ABC-type toxin complexes, composed of TcA, TcB and TcC proteins. Tc toxins are widespread and have shown a tropism for a variety of targets including insect, mammalian and human cells. However, their receptors and the specific mechanisms of uptake into target cells remain unknown. Here, we show that the TcA protein TcdA1 interacts with N-glycans, particularly Lewis X/Y antigens. This is confirmed using N-acetylglucosamine transferase I (Mgat1 gene product)-deficient Chinese hamster ovary (CHO) Lec1 cells, which are highly resistant to intoxication by the Tc toxin complex most likely due to the absence of complex N-glycans. Restoring Mgat1 gene activity, and hence complex N-glycan biosynthesis, recapitulated the sensitivity of these cells to the toxin. Exogenous addition of Lewis X trisaccharide partially inhibits intoxication in wild-type cells. Additionally, sialic acid also largely reduced binding of the Tc toxin. Moreover, proteolytic activation of TcdA1 alters glycan-binding and uptake into target cells. The data suggest that TcdA1-binding is most likely multivalent, and carbohydrates probably work cooperatively to facilitate binding and intoxication.


Asunto(s)
Toxinas Bacterianas , Photorhabdus , Animales , Células CHO , Cricetinae , Cricetulus , Humanos , Polisacáridos
9.
Cell Mol Life Sci ; 78(24): 8165-8186, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34731252

RESUMEN

B cell superantigens crosslink conserved domains of B cell receptors (BCRs) and cause dysregulated, polyclonal B cell activation irrespective of normal BCR-antigen complementarity. The cells typically succumb to activation-induced cell death, which can impede the adaptive immune response and favor infection. In the present study, we demonstrate that the fucose-binding lectin of Burkholderia ambifaria, BambL, bears functional resemblance to B cell superantigens. By engaging surface glycans, the bacterial lectin activated human peripheral blood B cells, which manifested in the surface expression of CD69, CD54 and CD86 but became increasingly cytotoxic at higher concentrations. The effects were sensitive to BCR pathway inhibitors and excess fucose, which corroborates a glycan-driven mode of action. Interactome analyses in a model cell line suggest BambL binds directly to glycans of the BCR and regulatory coreceptors. In vitro, BambL triggered BCR signaling and induced CD19 internalization and degradation. Owing to the lectin's six binding sites, we propose a BCR activation model in which BambL functions as a clustering hub for receptor glycans, modulates normal BCR regulation, and induces cell death through exhaustive activation.


Asunto(s)
Linfocitos B/metabolismo , Proteínas Bacterianas/metabolismo , Burkholderia/metabolismo , Lectinas/metabolismo , Polisacáridos/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Superantígenos/metabolismo , Linfocitos B/inmunología , Proteínas Bacterianas/inmunología , Sitios de Unión , Humanos , Lectinas/inmunología , Polisacáridos/inmunología , Unión Proteica , Receptores de Antígenos de Linfocitos B/inmunología , Transducción de Señal , Superantígenos/inmunología
10.
Cell Mol Life Sci ; 78(7): 3637-3656, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33555391

RESUMEN

The opportunistic pathogen Pseudomonas aeruginosa has gained precedence over the years due to its ability to develop resistance to existing antibiotics, thereby necessitating alternative strategies to understand and combat the bacterium. Our previous work identified the interaction between the bacterial lectin LecA and its host cell glycosphingolipid receptor globotriaosylceramide (Gb3) as a crucial step for the engulfment of P. aeruginosa via the lipid zipper mechanism. In this study, we define the LecA-associated host cell membrane domain by pull-down and mass spectrometry analysis. We unraveled a predilection of LecA for binding to saturated, long fatty acyl chain-containing Gb3 species in the extracellular membrane leaflet and an induction of dynamic phosphatidylinositol (3,4,5)-trisphosphate (PIP3) clusters at the intracellular leaflet co-localizing with sites of LecA binding. We found flotillins and the GPI-anchored protein CD59 not only to be an integral part of the LecA-interacting membrane domain, but also majorly influencing bacterial invasion as depletion of either of these host cell proteins resulted in about 50% reduced invasiveness of the P. aeruginosa strain PAO1. In summary, we report that the LecA-Gb3 interaction at the extracellular leaflet induces the formation of a plasma membrane domain enriched in saturated Gb3 species, CD59, PIP3 and flotillin thereby facilitating efficient uptake of PAO1.


Asunto(s)
Antígenos CD59/metabolismo , Membrana Celular/metabolismo , Interacciones Huésped-Patógeno , Pulmón/microbiología , Proteínas de la Membrana/metabolismo , Pseudomonas aeruginosa/aislamiento & purificación , Trihexosilceramidas/metabolismo , Transporte Biológico , Antígenos CD59/genética , Endocitosis , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/patología , Humanos , Pulmón/metabolismo , Pulmón/patología , Proteínas de la Membrana/genética , Pseudomonas aeruginosa/fisiología , Transducción de Señal
11.
Glycoconj J ; 38(5): 625-647, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34390447

RESUMEN

Glycans have been shown to function as versatile molecular signals in cells. This prompted us to look at their roles in endocytosis, endolysosomal system and autophagy. We start by introducing the cell biological aspects of these pathways, the concept of the sugar code, and provide an overview on the role of glycans in the targeting of lysosomal proteins and in lysosomal functions. Moreover, we review evidence on the regulation of endocytosis and autophagy by glycans. Finally, we discuss the emerging concept that cytosolic exposure of luminal glycans, and their detection by endogenous lectins, provides a mechanism for the surveillance of the integrity of the endolysosomal compartments, and serves their eventual repair or disposal.


Asunto(s)
Autofagia/fisiología , Endocitosis/fisiología , Lisosomas/fisiología , Polisacáridos/metabolismo , Regulación de la Expresión Génica , Proteínas/genética , Proteínas/metabolismo
12.
Bioinformatics ; 35(13): 2340-2342, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30475993

RESUMEN

MOTIVATION: Giant Unilamellar Vesicles (GUVs) are widely used synthetic membrane systems that mimic native membranes and cellular processes. Various fluorescence imaging techniques can be employed for their characterization. In order to guarantee a fast and unbiased analysis of imaging data, the development of automated recognition and processing steps is required. RESULTS: We developed a fast and versatile Fiji-based macro for the analysis of digital microscopy images of GUVs. This macro was designed to investigate membrane dye incorporation and protein binding to membranes. Moreover, we propose a fluorescence intensity-based method to quantitatively assess protein binding. AVAILABILITY AND IMPLEMENTATION: The ImageJ distribution package FIJI is freely available online: https://imagej.net/Fiji. The macro file GUV-AP.ijm is available at https://github.com/AG-Roemer/GUV-AP. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Liposomas Unilamelares
13.
J Am Soc Nephrol ; 30(12): 2338-2353, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31732614

RESUMEN

BACKGROUND: Mutations in about 50 genes have been identified as monogenic causes of nephrotic syndrome, a frequent cause of CKD. These genes delineated the pathogenetic pathways and rendered significant insight into podocyte biology. METHODS: We used whole-exome sequencing to identify novel monogenic causes of steroid-resistant nephrotic syndrome (SRNS). We analyzed the functional significance of an SRNS-associated gene in vitro and in podocyte-like Drosophila nephrocytes. RESULTS: We identified hemizygous missense mutations in the gene TBC1D8B in five families with nephrotic syndrome. Coimmunoprecipitation assays indicated interactions between TBC1D8B and active forms of RAB11. Silencing TBC1D8B in HEK293T cells increased basal autophagy and exocytosis, two cellular functions that are independently regulated by RAB11. This suggests that TBC1D8B plays a regulatory role by inhibiting endogenous RAB11. Coimmunoprecipitation assays showed TBC1D8B also interacts with the slit diaphragm protein nephrin, and colocalizes with it in immortalized cell lines. Overexpressed murine Tbc1d8b with patient-derived mutations had lower affinity for endogenous RAB11 and nephrin compared with wild-type Tbc1d8b protein. Knockdown of Tbc1d8b in Drosophila impaired function of the podocyte-like nephrocytes, and caused mistrafficking of Sns, the Drosophila ortholog of nephrin. Expression of Rab11 RNAi in nephrocytes entailed defective delivery of slit diaphragm protein to the membrane, whereas RAB11 overexpression revealed a partial phenotypic overlap to Tbc1d8b loss of function. CONCLUSIONS: Novel mutations in TBC1D8B are monogenic causes of SRNS. This gene inhibits RAB11. Our findings suggest that RAB11-dependent vesicular nephrin trafficking plays a role in the pathogenesis of nephrotic syndrome.


Asunto(s)
Proteínas de Unión al Calcio/genética , Mutación Missense , Síndrome Nefrótico/genética , Podocitos/metabolismo , Vesículas Transportadoras/fisiología , Proteínas de Transporte Vesicular/genética , Proteínas de Unión al GTP rab/metabolismo , Animales , Autofagia , Línea Celular Transformada , Perros , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Exocitosis , Silenciador del Gen , Células HEK293 , Humanos , Inmunoglobulinas/metabolismo , Células de Riñón Canino Madin Darby , Proteínas de la Membrana/metabolismo , Síndrome Nefrótico/metabolismo , Fenotipo , Mapeo de Interacción de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Secuenciación del Exoma
14.
Langmuir ; 35(9): 3534-3544, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30802059

RESUMEN

The binding of the pentameric capsid protein VP1 of simian virus 40 to its glycosphingolipid receptor GM1 is a key step for the entry of the virus into the host cell. Recent experimental studies have shown that the interaction of variants of soluble VP1 pentamers with giant unilamellar vesicles composed of GM1, DOPC, and cholesterol leads to the formation of tubular membrane invaginations to the inside of the vesicles, mimicking the initial steps of endocytosis. We have used coarse-grained and atomistic molecular dynamics (MD) simulations to study the interaction of VP1 with GM1/DOPC/cholesterol bilayers. In the presence of one VP1 protein, we monitor the formation of small local negative curvature and membrane thinning at the protein binding site as well as reduction of area per lipid. These membrane deformations are also observed under cholesterol-free conditions. However, here, the number of GM1 molecules attached to the VP1 binding pockets increases. The membrane curvature is slightly increased for asymmetric GM1 distribution that mimics conditions in vivo, compared to symmetric GM1 distributions which are often applied in experiments. Slightly smaller inward curvature was observed in atomistic control simulations. Binding of four VP1 proteins leads to an increase of the average intrinsic area per lipid in the protein binding leaflet. Membrane fluctuations appear to be the driving force of VP1 aggregation, as was previously shown for membrane-adhering particles because no VP1 aggregation is observed in the absence of a lipid membrane.


Asunto(s)
Proteínas de la Cápside/metabolismo , Membrana Dobles de Lípidos/metabolismo , Receptores de Superficie Celular/metabolismo , Virus 40 de los Simios/química , Colesterol/química , Gangliósido G(M1)/química , Membrana Dobles de Lípidos/química , Simulación de Dinámica Molecular , Fosfatidilcolinas/química
15.
Biochim Biophys Acta Mol Cell Res ; 1864(7): 1236-1245, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28428058

RESUMEN

The human pathogen Pseudomonas aeruginosa induces phosphorylation of the adaptor protein CrkII by activating the non-receptor tyrosine kinase Abl to promote its uptake into host cells. So far, specific factors of P. aeruginosa, which induce Abl/CrkII signalling, are entirely unknown. In this research, we employed human lung epithelial cells H1299, Chinese hamster ovary cells and P. aeruginosa wild type strain PAO1 to study the invasion process of P. aeruginosa into host cells by using microbiological, biochemical and cell biological approaches such as Western Blot, immunofluorescence microscopy and flow cytometry. Here, we demonstrate that the host glycosphingolipid globotriaosylceramide, also termed Gb3, represents a signalling receptor for the P. aeruginosa lectin LecA to induce CrkII phosphorylation at tyrosine 221. Alterations in Gb3 expression and LecA function correlate with CrkII phosphorylation. Interestingly, phosphorylation of CrkIIY221 occurs independently of Abl kinase. We further show that Src family kinases transduce the signal induced by LecA binding to Gb3, leading to CrkY221 phosphorylation. In summary, we identified LecA as a bacterial factor, which utilizes a so far unrecognized mechanism for phospho-CrkIIY221 induction by binding to the host glycosphingolipid receptor Gb3. The LecA/Gb3 interaction highlights the potential of glycolipids to mediate signalling processes across the plasma membrane and should be further elucidated to gain deeper insights into this non-canonical mechanism of activating host cell processes.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Globósidos/metabolismo , Proteínas Proto-Oncogénicas c-crk/metabolismo , Pseudomonas aeruginosa/patogenicidad , Transducción de Señal , Trihexosilceramidas/metabolismo , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Interacciones Huésped-Patógeno , Humanos , Fosforilación , Procesamiento Proteico-Postraduccional , Pseudomonas aeruginosa/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , Familia-src Quinasas/metabolismo
16.
Biochim Biophys Acta ; 1860(2): 392-401, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26546712

RESUMEN

BACKGROUND: Fruiting body lectins have been proposed to act as effector proteins in the defense of fungi against parasites and predators. The Marasmius oreades agglutinin (MOA) is a lectin from the fairy ring mushroom with specificity for Galα1-3Gal containing carbohydrates. This lectin is composed of an N-terminal carbohydrate-binding domain and a C-terminal dimerization domain. The dimerization domain of MOA shows in addition calcium-dependent cysteine protease activity, similar to the calpain family. METHODS: Cell detachment assay, cell viability assay, immunofluorescence, live cell imaging and Western blot using MDCKII cell line. RESULTS: In this study, we demonstrate in MDCKII cells that after internalization, MOA protease activity induces profound physiological cellular responses, like cytoskeleton rearrangement, cell detachment and cell death. These changes are preceded by a decrease in FAK phosphorylation and an internalization and degradation of ß1-integrin, consistent with a disruption of integrin-dependent cell adhesion signaling. Once internalized, MOA accumulates in late endosomal compartments. CONCLUSION: Our results suggest a possible toxic mechanism of MOA, which consists of disturbing the cell adhesion and the cell viability. GENERAL SIGNIFICANCE: After being ingested by a predator, MOA might exert a protective role by diminishing host cell integrity.


Asunto(s)
Aglutininas/fisiología , Integrina beta1/fisiología , Marasmius/química , Animales , Adhesión Celular , Células Cultivadas , Clatrina/fisiología , Perros , Dinaminas/fisiología , Endocitosis , Endosomas/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/fisiología
17.
Biochim Biophys Acta ; 1863(6 Pt A): 1106-18, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26862060

RESUMEN

Pseudomonas aeruginosa is an opportunistic pathogen that induces severe lung infections such as ventilator-associated pneumonia and acute lung injury. Under these conditions, the bacterium diminishes epithelial integrity and inhibits tissue repair mechanisms, leading to persistent infections. Understanding the involved bacterial virulence factors and their mode of action is essential for the development of new therapeutic approaches. In our study we discovered a so far unknown effect of the P. aeruginosa lectin LecB on host cell physiology. LecB alone was sufficient to attenuate migration and proliferation of human lung epithelial cells and to induce transcriptional activity of NF-κB. These effects are characteristic of impaired tissue repair. Moreover, we found a strong degradation of ß-catenin, which was partially recovered by the proteasome inhibitor lactacystin. In addition, LecB induced loss of cell-cell contacts and reduced expression of the ß-catenin targets c-myc and cyclin D1. Blocking of LecB binding to host cell plasma membrane receptors by soluble l-fucose prevented these changes in host cell behavior and signaling, and thereby provides a powerful strategy to suppress LecB function. Our findings suggest that P. aeruginosa employs LecB as a virulence factor to induce ß-catenin degradation, which then represses processes that are directly linked to tissue recovery.


Asunto(s)
Proteínas Bacterianas/farmacología , Células Epiteliales/efectos de los fármacos , Lectinas/farmacología , beta Catenina/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Proteínas Bacterianas/genética , Western Blotting , Comunicación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/farmacología , Relación Dosis-Respuesta a Droga , Células Epiteliales/citología , Células Epiteliales/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Integrina beta1/metabolismo , Lectinas/genética , Microscopía Confocal , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Proteínas Recombinantes/farmacología , Factor de Transcripción ReIA/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
18.
Blood ; 125(21): 3287-96, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25784678

RESUMEN

B-cell antigen receptor (BCR) expression is a key feature of most B-cell lymphomas, but the mechanisms of BCR signal induction and the involvement of autoantigen recognition remain unclear. In follicular lymphoma (FL) B cells, BCR expression is retained despite a chromosomal translocation that links the antiapoptotic gene BCL2 to the regulatory elements of immunoglobulin genes, thereby disrupting 1 heavy-chain allele. A remarkable feature of FL-BCRs is the acquisition of potential N-glycosylation sites during somatic hypermutation. The introduced glycans carry mannose termini, which create potential novel binding sites for mannose-specific lectins. Here, we investigated the effect of N-linked variable-region glycosylation for BCR interaction with cognate antigen and with lectins of different origins. N-glycans were found to severely impair BCR specificity and affinity to the initial cognate antigen. In addition, we found that lectins from Pseudomonas aeruginosa and Burkholderia cenocepacia bind and stimulate FL cells. Human exposure to these bacteria can occur by contact with soil and water. In addition, they represent opportunistic pathogens in susceptible hosts. Understanding the role of bacterial lectins might elucidate the pathogenesis of FL and establish novel therapeutic approaches.


Asunto(s)
Infecciones Bacterianas/complicaciones , Región Variable de Inmunoglobulina/inmunología , Lectinas/inmunología , Linfoma Folicular/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Infecciones Bacterianas/inmunología , Citometría de Flujo , Glicosilación , Humanos , Región Variable de Inmunoglobulina/química , Linfoma Folicular/complicaciones , Infecciones Oportunistas/complicaciones , Infecciones Oportunistas/inmunología , Polisacáridos/metabolismo , Receptores de Antígenos de Linfocitos B/química , Receptores de Antígenos de Linfocitos B/metabolismo
19.
Proc Natl Acad Sci U S A ; 111(11): 4127-32, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24591614

RESUMEN

Emerging data suggest that in polarized epithelial cells newly synthesized apical and basolateral plasma membrane proteins traffic through different endosomal compartments en route to the respective cell surface. However, direct evidence for trans-endosomal pathways of plasma membrane proteins is still missing and the mechanisms involved are poorly understood. Here, we imaged the entire biosynthetic route of rhodopsin-GFP, an apical marker in epithelial cells, synchronized through recombinant conditional aggregation domains, in live Madin-Darby canine kidney cells using spinning disk confocal microscopy. Our experiments directly demonstrate that rhodopsin-GFP traffics through apical recycling endosomes (AREs) that bear the small GTPase Rab11a before arriving at the apical membrane. Expression of dominant-negative Rab11a drastically reduced apical delivery of rhodopsin-GFP and caused its missorting to the basolateral membrane. Surprisingly, functional inhibition of dynamin-2 trapped rhodopsin-GFP at AREs and caused aberrant accumulation of coated vesicles on AREs, suggesting a previously unrecognized role for dynamin-2 in the scission of apical carrier vesicles from AREs. A second set of experiments, using a unique method to carry out total internal reflection fluorescence microscopy (TIRFM) from the apical side, allowed us to visualize the fusion of rhodopsin-GFP carrier vesicles, which occurred randomly all over the apical plasma membrane. Furthermore, two-color TIRFM showed that Rab11a-mCherry was present in rhodopsin-GFP carrier vesicles and was rapidly released upon fusion onset. Our results provide direct evidence for a role of AREs as a post-Golgi sorting hub in the biosynthetic route of polarized epithelia, with Rab11a regulating cargo sorting at AREs and carrier vesicle docking at the apical membrane.


Asunto(s)
Vías Biosintéticas/fisiología , Polaridad Celular/fisiología , Células Epiteliales/citología , Rodopsina/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Cartilla de ADN/genética , Perros , Aparato de Golgi/metabolismo , Immunoblotting , Inmunohistoquímica , Células de Riñón Canino Madin Darby , Microscopía Confocal , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Plásmidos/genética , Transporte de Proteínas/fisiología , Rodopsina/biosíntesis , Vesículas Transportadoras/metabolismo
20.
Proc Natl Acad Sci U S A ; 111(35): 12895-900, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25136128

RESUMEN

Glycosphingolipids are important structural constituents of cellular membranes. They are involved in the formation of nanodomains ("lipid rafts"), which serve as important signaling platforms. Invasive bacterial pathogens exploit these signaling domains to trigger actin polymerization for the bending of the plasma membrane and the engulfment of the bacterium--a key process in bacterial uptake. However, it is unknown whether glycosphingolipids directly take part in the membrane invagination process. Here, we demonstrate that a "lipid zipper," which is formed by the interaction between the bacterial surface lectin LecA and its cellular receptor, the glycosphingolipid Gb3, triggers plasma membrane bending during host cell invasion of the bacterium Pseudomonas aeruginosa. In vitro experiments with Gb3-containing giant unilamellar vesicles revealed that LecA/Gb3-mediated lipid zippering was sufficient to achieve complete membrane engulfment of the bacterium. In addition, theoretical modeling elucidated that the adhesion energy of the LecA-Gb3 interaction is adequate to drive the engulfment process. In cellulo experiments demonstrated that inhibition of the LecA/Gb3 lipid zipper by either lecA knockout, Gb3 depletion, or application of soluble sugars that interfere with LecA binding to Gb3 significantly lowered P. aeruginosa uptake by host cells. Of note, membrane engulfment of P. aeruginosa occurred independently of actin polymerization, thus corroborating that lipid zippering alone is sufficient for this crucial first step of bacterial host-cell entry. Our study sheds new light on the impact of glycosphingolipids in the cellular invasion of bacterial pathogens and provides a mechanistic explication of the initial uptake processes.


Asunto(s)
Actinas/metabolismo , Glicoesfingolípidos/metabolismo , Microdominios de Membrana/microbiología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/metabolismo , Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana/fisiología , Membrana Celular/metabolismo , Membrana Celular/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Glucolípidos/metabolismo , Membrana Dobles de Lípidos/metabolismo , Microdominios de Membrana/metabolismo , Modelos Biológicos , Transducción de Señal/fisiología , Esfingolípidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA