Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(38): e2310914120, 2023 09 19.
Article in English | MEDLINE | ID: mdl-37695903

ABSTRACT

Extracellular vesicles (EVs) are membrane-limited organelles mediating cell-to-cell communication in health and disease. EVs are of high medical interest, but their rational use for diagnostics or therapies is restricted by our limited understanding of the molecular mechanisms governing EV biology. Here, we tested whether PDZ proteins, molecular scaffolds that support the formation, transport, and function of signal transduction complexes and that coevolved with multicellularity, may represent important EV regulators. We reveal that the PDZ proteome (ca. 150 proteins in human) establishes a discrete number of direct interactions with the tetraspanins CD9, CD63, and CD81, well-known EV constituents. Strikingly, PDZ proteins interact more extensively with syndecans (SDCs), ubiquitous membrane proteins for which we previously demonstrated an important role in EV biogenesis, loading, and turnover. Nine PDZ proteins were tested in loss-of-function studies. We document that these PDZ proteins regulate both tetraspanins and SDCs, differentially affecting their steady-state levels, subcellular localizations, metabolism, endosomal budding, and accumulations in EVs. Importantly, we also show that PDZ proteins control the levels of heparan sulfate at the cell surface that functions in EV capture. In conclusion, our study establishes that the extensive networking of SDCs, tetraspanins, and PDZ proteins contributes to EV heterogeneity and turnover, highlighting an important piece of the molecular framework governing intracellular trafficking and intercellular communication.


Subject(s)
Extracellular Vesicles , Signal Transduction , Humans , Biological Transport , Cell Communication , Cell Division , Syndecans , Transcription Factors
2.
Proc Natl Acad Sci U S A ; 117(11): 5913-5922, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32108028

ABSTRACT

Exosomes, extracellular vesicles (EVs) of endosomal origin, emerge as master regulators of cell-to-cell signaling in physiology and disease. Exosomes are highly enriched in tetraspanins (TSPNs) and syndecans (SDCs), the latter occurring mainly in proteolytically cleaved form, as membrane-spanning C-terminal fragments of the proteins. While both protein families are membrane scaffolds appreciated for their role in exosome formation, composition, and activity, we currently ignore whether these work together to control exosome biology. Here we show that TSPN6, a poorly characterized tetraspanin, acts as a negative regulator of exosome release, supporting the lysosomal degradation of SDC4 and syntenin. We demonstrate that TSPN6 tightly associates with SDC4, the SDC4-TSPN6 association dictating the association of TSPN6 with syntenin and the TSPN6-dependent lysosomal degradation of SDC4-syntenin. TSPN6 also inhibits the shedding of the SDC4 ectodomain, mimicking the effects of matrix metalloproteinase inhibitors. Taken together, our data identify TSPN6 as a regulator of the trafficking and processing of SDC4 and highlight an important physical and functional interconnection between these membrane scaffolds for the production of exosomes. These findings clarify our understanding of the molecular determinants governing EV formation and have potentially broad impact for EV-related biomedicine.


Subject(s)
Exosomes/metabolism , Syntenins/metabolism , Tetraspanins/metabolism , Cell Communication , Exosomes/genetics , Extracellular Vesicles/metabolism , Humans , Lysosomes/metabolism , MCF-7 Cells , Matrix Metalloproteinases/metabolism , Protein Transport , Syndecan-4/metabolism , Syndecans/metabolism
3.
Molecules ; 26(24)2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34946571

ABSTRACT

HIV-1 transactivating factor Tat is released by infected cells. Extracellular Tat homodimerizes and engages several receptors, including integrins, vascular endothelial growth factor receptor 2 (VEGFR2) and heparan sulfate proteoglycan (HSPG) syndecan-1 expressed on various cells. By means of experimental cell models recapitulating the processes of lymphocyte trans-endothelial migration, here, we demonstrate that upon association with syndecan-1 expressed on lymphocytes, Tat triggers simultaneously the in cis activation of lymphocytes themselves and the in trans activation of endothelial cells (ECs). This "two-way" activation eventually induces lymphocyte adhesion and spreading onto the substrate and vascular endothelial (VE)-cadherin reorganization at the EC junctions, with consequent endothelial permeabilization, leading to an increased extravasation of Tat-presenting lymphocytes. By means of a panel of biochemical activation assays and specific synthetic inhibitors, we demonstrate that during the above-mentioned processes, syndecan-1, integrins, FAK, src and ERK1/2 engagement and activation are needed in the lymphocytes, while VEGFR2, integrin, src and ERK1/2 are needed in the endothelium. In conclusion, the Tat/syndecan-1 complex plays a central role in orchestrating the setup of the various in cis and in trans multimeric complexes at the EC/lymphocyte interface. Thus, by means of computational molecular modelling, docking and dynamics, we also provide a characterization at an atomic level of the binding modes of the Tat/heparin interaction, with heparin herein used as a structural analogue of the heparan sulfate chains of syndecan-1.


Subject(s)
Endothelium/metabolism , Heparan Sulfate Proteoglycans/metabolism , Lymphocytes/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Cell Adhesion , Cell Movement , Endothelium/chemistry , Heparan Sulfate Proteoglycans/chemistry , Humans , Lymphocytes/chemistry , Models, Molecular , Molecular Structure , Stereoisomerism , Tumor Cells, Cultured , tat Gene Products, Human Immunodeficiency Virus/chemistry
4.
Adv Exp Med Biol ; 1221: 285-307, 2020.
Article in English | MEDLINE | ID: mdl-32274714

ABSTRACT

Exosomes are secreted vesicles involved in signaling processes. The biogenesis of a class of these extracellular vesicles depends on syntenin, and on the interaction of this cytosolic protein with syndecans. Heparanase, largely an endosomal enzyme, acts as a regulator of the syndecan-syntenin-exosome biogenesis pathway. The upregulation of syntenin and heparanase in cancers may support the suspected roles of exosomes in tumor biology.


Subject(s)
Exosomes/metabolism , Glucuronidase/metabolism , Humans , Syndecans , Syntenins
5.
Proc Natl Acad Sci U S A ; 114(47): 12495-12500, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29109268

ABSTRACT

The cytoplasmic tyrosine kinase SRC controls cell growth, proliferation, adhesion, and motility. The current view is that SRC acts primarily downstream of cell-surface receptors to control intracellular signaling cascades. Here we reveal that SRC functions in cell-to-cell communication by controlling the biogenesis and the activity of exosomes. Exosomes are viral-like particles from endosomal origin that can reprogram recipient cells. By gain- and loss-of-function studies, we establish that SRC stimulates the secretion of exosomes having promigratory activity on endothelial cells and that syntenin is mandatory for SRC exosomal function. Mechanistically, SRC impacts on syndecan endocytosis and on syntenin-syndecan endosomal budding, upstream of ARF6 small GTPase and its effector phospholipase D2, directly phosphorylating the conserved juxtamembrane DEGSY motif of the syndecan cytosolic domain and syntenin tyrosine 46. Our study uncovers a function of SRC in cell-cell communication, supported by syntenin exosomes, which is likely to contribute to tumor-host interactions.


Subject(s)
Cell Communication/genetics , Exosomes/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Oncogene Protein pp60(v-src)/genetics , Syntenins/genetics , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Amino Acid Motifs , Cell Movement , Cell Proliferation , Culture Media, Conditioned/pharmacology , Endocytosis , Endosomes/metabolism , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , MCF-7 Cells , Oncogene Protein pp60(v-src)/metabolism , Phospholipase D/genetics , Phospholipase D/metabolism , Phosphorylation , Signal Transduction , Syndecans/genetics , Syndecans/metabolism , Syntenins/metabolism
6.
Biol Cell ; 107(10): 331-41, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26032692

ABSTRACT

Cells communicate with their environment in various ways, including by secreting vesicles. Secreted vesicles are loaded with proteins, lipids and RNAs that compose 'a signature' of the cell of origin and potentially can reprogram recipient cells. Secreted vesicles recently gained in interest for medicine. They represent potential sources of biomarkers that can be collected from body fluids and, by disseminating pathogenic proteins, might also participate in systemic diseases like cancer, atherosclerosis and neurodegeneration. The mechanisms controlling the biogenesis and the uptake of secreted vesicles are poorly understood. Some of these vesicles originate from endosomes and are called 'exosomes'. In this review, we recapitulate recent insight on the role of the syndecan (SDC) heparan sulphate proteoglycans, the small intracellular adaptor syntenin and associated regulators in the biogenesis and loading of exosomes with cargo. SDC-syntenin-associated regulators include the endosomal sorting complex required for transport accessory component ALG-2-interacting protein X, the small GTPase adenosine 5'-diphosphate-ribosylation factor 6, the lipid-modifying enzyme phospholipase D2 and the endoglycosidase heparanase. All these molecules appear to support the budding of SDC-syntenin and associated cargo into the lumen of endosomes. This highlights a major mechanism for the formation of intraluminal vesicles that will be released as exosomes.


Subject(s)
Exosomes/metabolism , Syndecans/metabolism , Syntenins/metabolism , Animals , Humans
7.
J Cell Sci ; 126(Pt 21): 4856-61, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23986479

ABSTRACT

Proteolytic processing of amyloid-ß precursor protein (APP) generates the amyloid-ß peptide, which plays a central role in Alzheimer disease. The physiological function of APP and its proteolytic fragments, however, remains barely understood. Here we show that, on the basis of its binding characteristics, the secreted ectodomain of APP (sAPP) is a new member of the heparin-binding growth factor superfamily. Like other of its members, sAPP binds in a bivalent manner to the plasma membrane with two different subdomains. The N-terminal growth-factor-like domain (GFLD) is necessary and sufficient for protein-receptor binding, whereas the E2-domain mediates interaction with membrane-anchored heparan sulfate proteoglycans (HSPGs). The membrane-anchored HSPGs function as low-affinity co-receptors for sAPP and enhance the affinity to the sAPP receptor. Our findings provide a solid basis for the further identification of this receptor.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Glypicans/metabolism , Receptors, Cell Surface/metabolism , Syndecan-2/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Animals , CHO Cells , Cricetulus , Glypicans/genetics , Humans , Mice , Neurons/metabolism , Protein Binding , Protein Structure, Tertiary , Receptors, Cell Surface/genetics , Syndecan-2/genetics
8.
J Cell Sci ; 125(Pt 5): 1129-40, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22399807

ABSTRACT

Epiboly, the spreading and the thinning of the blastoderm to cover the yolk cell and close the blastopore in fish embryos, is central to the process of gastrulation. Despite its fundamental importance, little is known about the molecular mechanisms that control this coordinated cell movement. By a combination of knockdown studies and rescue experiments in zebrafish (Danio rerio), we show that epiboly relies on the molecular networking of syntenin with syndecan heparan sulphate proteoglycans, which act as co-receptors for adhesion molecules and growth factors. Furthermore, we show that the interaction of syntenin with phosphatidylinositol 4,5-bisphosphate (PIP2) and with the small GTPase ADP-ribosylation factor 6 (Arf6), which regulate the endocytic recycling of syndecan, is necessary for epiboly progression. Analysis of the earliest cellular defects suggests a role for syntenin in the autonomous vegetal expansion of the yolk syncytial layer and the rearrangement of the actin cytoskeleton in extra-embryonic tissues, but not in embryonic cell fate determination. This study identifies the importance of the syntenin-syndecan-PIP2-Arf6 complex for the progression of fish epiboly and establishes its key role in directional cell movements during early development.


Subject(s)
Gastrulation/physiology , Syndecans/metabolism , Syntenins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Animals , Cell Movement/physiology , Cytoskeleton/genetics , Gene Knockdown Techniques , Mice , Molecular Sequence Data , Phosphatidylinositol 4,5-Diphosphate/metabolism , Syntenins/genetics , Zebrafish/metabolism , Zebrafish/physiology , Zebrafish Proteins/genetics
9.
EMBO Rep ; 12(10): 1039-46, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21836636

ABSTRACT

Here we identify a new role for Syndecan (Sdc), the only transmembrane heparan sulphate proteoglycan in Drosophila, in tracheal development. Sdc is required cell autonomously for efficient directed migration and fusion of dorsal branch cells, but not for dorsal branch formation per se. The cytoplasmic domain of Sdc is dispensable, indicating that Sdc does not transduce a signal by itself. Although the branch-specific phenotype of sdc mutants resembles those seen in the absence of Slit/Robo2 signalling, genetic interaction experiments indicate that Sdc also helps to suppress Slit/Robo2 signalling. We conclude that Sdc cell autonomously regulates Slit/Robo2 signalling in tracheal cells to guarantee ordered directional migration and branch fusion.


Subject(s)
Cell Movement , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Syndecans/metabolism , Animals , Base Sequence , Cell Movement/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Gene Expression Regulation , Gene Order , Molecular Sequence Data , Phenotype , Protein Stability , Sequence Alignment , Signal Transduction , Syndecans/genetics , Trachea/metabolism , Roundabout Proteins
10.
Biochem J ; 448(1): 73-82, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22920187

ABSTRACT

The PC (proprotein convertase) furin cleaves a large variety of proproteins and hence plays a major role in many pathologies. Therefore furin inhibition might be a good strategy for therapeutic intervention, and several furin inhibitors have been generated, although none are entirely furin-specific. To reduce potential side effects caused by cross-reactivity with other proteases, dromedary heavy-chain-derived nanobodies against catalytically active furin were developed as specific furin inhibitors. The nanobodies bound only to furin but not to other PCs. Upon overexpression in cell lines, they inhibited the cleavage of two different furin substrates, TGFß (transforming growth factor ß) and GPC3 (glypican 3). Purified nanobodies could inhibit the cleavage of diphtheria toxin into its enzymatically active A fragment, but did not inhibit cleavage of a small synthetic peptide-based substrate, suggesting a mode-of-action based on steric hindrance. The dissociation constant of purified nanobody 14 is in the nanomolar range. The nanobodies were non-competitive inhibitors with an inhibitory constant in the micromolar range as demonstrated by Dixon plot. Furthermore, anti-furin nanobodies could protect HEK (human embryonic kidney)-293T cells from diphtheria-toxin-induced cytotoxicity as efficiently as the PC inhibitor nona-D-arginine. In conclusion, these antibody-based single-domain nanobodies represent the first generation of highly specific non-competitive furin inhibitors.


Subject(s)
Furin/antagonists & inhibitors , Single-Domain Antibodies/pharmacology , Animals , Antibody Specificity , Camelus , Catalysis/drug effects , Coumarins/metabolism , Diphtheria Toxin/metabolism , Endocytosis , Furin/chemistry , Furin/immunology , Furin/metabolism , Glypicans/metabolism , HEK293 Cells/metabolism , Humans , Kinetics , Mice , Oligopeptides/metabolism , Peptide Fragments/metabolism , Proprotein Convertases/metabolism , Protein Binding/drug effects , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Substrate Specificity , Transforming Growth Factor beta/metabolism
11.
EMBO Mol Med ; 15(11): e17570, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37819151

ABSTRACT

The crosstalk between cancer and stromal cells plays a critical role in tumor progression. Syntenin is a small scaffold protein involved in the regulation of intercellular communication that is emerging as a target for cancer therapy. Here, we show that certain aggressive forms of acute myeloid leukemia (AML) reduce the expression of syntenin in bone marrow stromal cells (BMSC). Stromal syntenin deficiency, in turn, generates a pro-tumoral microenvironment. From serial transplantations in mice and co-culture experiments, we conclude that syntenin-deficient BMSC stimulate AML aggressiveness by promoting AML cell survival and protein synthesis. This pro-tumoral activity is supported by increased expression of endoglin, a classical marker of BMSC, which in trans stimulates AML translational activity. In short, our study reveals a vicious signaling loop potentially at the heart of AML-stroma crosstalk and unsuspected tumor-suppressive effects of syntenin that need to be considered during systemic targeting of syntenin in cancer therapy.


Subject(s)
Leukemia, Myeloid, Acute , Syntenins , Animals , Mice , Syntenins/genetics , Syntenins/metabolism , Down-Regulation , Leukemia, Myeloid, Acute/metabolism , Signal Transduction , Stromal Cells/metabolism , Tumor Microenvironment
12.
J Biol Chem ; 286(9): 7577-86, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21193412

ABSTRACT

Pregnancy-specific ß1 glycoproteins (PSGs) are the most abundant fetal proteins in the maternal bloodstream in late pregnancy. They are secreted by the syncytiotrophoblast and are detected around day 14 postfertilization. There are 11 human PSG genes, which encode a family of proteins exhibiting significant conservation at the amino acid level. We and others have proposed that PSGs have an immune modulatory function. In addition, we recently postulated that they are proangiogenic due to their ability to induce the secretion of VEGF-A and the formation of tubes by endothelial cells. The cellular receptor(s) for human PSGs remain unknown. Therefore, we conducted these studies to identify the receptor for PSG1, the highest expressed member of the family. We show that removal of cell surface glycosaminoglycans (GAGs) by enzymatic or chemical treatment of cells or competition with heparin completely inhibited binding of PSG1. In addition, PSG1 did not bind to cells lacking heparan or chondroitin sulfate on their surface, and binding was restored upon transfection with all four syndecans and glypican-1. Importantly, the presence of GAGs on the surface of endothelial cells was required for the ability of PSG1 to induce tube formation. This finding indicates that the PSG1-GAG interaction mediates at least some of the PSG1 proposed functions.


Subject(s)
Chondroitin Sulfates/metabolism , Heparitin Sulfate/metabolism , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Trophoblasts/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Endothelial Cells/metabolism , Female , HeLa Cells , Heparin/metabolism , Humans , Jurkat Cells , Mice , NIH 3T3 Cells , Neovascularization, Physiologic/physiology , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/genetics , Syndecans/metabolism , Transfection , Trophoblasts/cytology
13.
Front Cell Dev Biol ; 10: 886381, 2022.
Article in English | MEDLINE | ID: mdl-35669514

ABSTRACT

Matrix metalloproteinases (MMPs) are key players in matrix remodeling and their function has been particularly investigated in cancer biology. Indeed, through extracellular matrix (ECM) degradation and shedding of diverse cell surface macromolecules, they are implicated in different steps of tumor development, from local expansion by growth to tissue invasion and metastasis. Interestingly, MMPs are also components of extracellular vesicles (EVs). EVs are membrane-limited organelles that cells release in their extracellular environment. These "secreted" vesicles are now well accepted players in cell-to-cell communication. EVs have received a lot of interest in recent years as they are also envisioned as sources of biomarkers and as potentially outperforming vehicles for the delivery of therapeutics. Molecular machineries governing EV biogenesis, cargo loading and delivery to recipient cells are complex and still under intense investigation. In this review, we will summarize the state of the art of our knowledge about the molecular mechanisms implicated in MMP trafficking and secretion. We focus on MT1-MMP, a major effector of invasive cell behavior. We will also discuss how this knowledge is of interest for a better understanding of EV-loading of MMPs. Such knowledge might be of use to engineer novel strategies for cancer treatment. A better understanding of these mechanisms could also be used to design more efficient EV-based therapies.

14.
Blood ; 114(15): 3335-42, 2009 Oct 08.
Article in English | MEDLINE | ID: mdl-19661268

ABSTRACT

The HIV-1 transactivating factor Tat accumulates on the surface of endothelium by interacting with heparan sulfate proteoglycans (HSPGs). Tat also interacts with B-lymphoid Namalwa cells but only when these overexpress HSPGs after syndecan-1 cDNA transfection (SYN-NCs). Accordingly, SYN-NCs, but not mock-transfected cells, adhere to endothelial cells (ECs) when Tat is bound to the surface of either one of the 2 cell types or when SYN-NCs are transfected with a Tat cDNA. Moreover, endogenously produced Tat bound to cell-surface HSPGs mediates cell adhesion of HIV(+) ACH-2 lymphocytes to the endothelium. This heterotypic lymphocyte-EC interaction is prevented by HSPG antagonist or heparinase treatment, but not by integrin antagonists and requires the homodimerization of Tat protein. Tat tethered to the surface of SYN-NCs or of peripheral blood monocytes from healthy donors promotes their transendothelial migration in vitro in response to CXCL12 or CCL5, respectively, and SYN-NC extravasation in vivo in a zebrafish embryo model of inflammation. In conclusion, Tat homodimers bind simultaneously to HSPGs expressed on lymphoid and EC surfaces, leading to HSPG/Tat-Tat/HSPG quaternary complexes that physically link HSPG-bearing lymphoid cells to the endothelium, promoting their extravasation. These data provide new insights about how lymphoid cells extravasate during HIV infection.


Subject(s)
Cell Movement , Endothelium, Vascular/metabolism , HIV Infections/metabolism , HIV-1/metabolism , Lymphocytes/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Cattle , Cell Adhesion , Cell Line, Tumor , Chemokine CCL5/pharmacology , Chemokine CXCL12/pharmacology , Embryo, Nonmammalian/metabolism , HIV Infections/genetics , Heparan Sulfate Proteoglycans , Humans , Protein Multimerization , Syndecan-1 , Transfection , Zebrafish/genetics , Zebrafish/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics
15.
Nucleic Acids Res ; 37(13): e93, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19483100

ABSTRACT

Targeted gene silencing by RNA interference allows the study of gene function in plants and animals. In cell culture and small animal models, genetic screens can be performed--even tissue-specifically in Drosophila--with genome-wide RNAi libraries. However, a major problem with the use of RNAi approaches is the unavoidable false-positive error caused by off-target effects. Until now, this is minimized by computational RNAi design, comparing RNAi to the mutant phenotype if known, and rescue with a presumed ortholog. The ultimate proof of specificity would be to restore expression of the same gene product in vivo. Here, we present a simple and efficient method to rescue the RNAi-mediated knockdown of two independent genes in Drosophila. By exploiting the degenerate genetic code, we generated Drosophila RNAi Escape Strategy Construct (RESC) rescue proteins containing frequent silent mismatches in the complete RNAi target sequence. RESC products were no longer efficiently silenced by RNAi in cell culture and in vivo. As a proof of principle, we rescue the RNAi-induced loss of function phenotype of the eye color gene white and tracheal defects caused by the knockdown of the heparan sulfate proteoglycan syndecan. Our data suggest that RESC is widely applicable to rescue and validate ubiquitous or tissue-specific RNAi and to perform protein structure-function analysis.


Subject(s)
Drosophila/genetics , Gene Knockdown Techniques , RNA Interference , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Animals , Cell Line , Drosophila/anatomy & histology , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Eye/anatomy & histology , Eye Proteins/antagonists & inhibitors , Eye Proteins/genetics , Genes, Insect , Phenotype , Syndecans/antagonists & inhibitors , Syndecans/genetics
16.
Med Sci (Paris) ; 37(12): 1101-1107, 2021 Dec.
Article in French | MEDLINE | ID: mdl-34928212

ABSTRACT

Exosomes are small extracellular vesicles derived from endosomal compartments. The molecular mechanisms supporting the biology of exosomes, from their biogenesis to their internalization by target cells, rely on 'dedicated' membrane proteins. These mechanisms of action need to be further clarified. This will help to better understand how exosome composition and heterogeneity are established. This would also help to rationalize their use as source of biomarkers and therapeutic tools. Here we discuss how syndecans and tetraspanins, two families of membrane scaffold proteins, cooperate to regulate different steps of exosome biology.


TITLE: Tétraspanines et syndécanes - Complices dans le « trafic ¼ des exosomes ? ABSTRACT: Les exosomes sont de petites vésicules extracellulaires qui sont produites dans des compartiments endosomaux. Les mécanismes moléculaires sur lesquels reposent la biologie des exosomes, de leur biogenèse à leur internalisation par les cellules cibles, font notamment appel à des protéines membranaires particulières. Ces mécanismes méritent d'être clarifiés, afin de mieux comprendre la complexité de la composition des exosomes et de rationaliser leur utilisation comme biomarqueurs ou comme outils thérapeutiques. Nous discutons ici comment les syndécanes et les tétraspanines, deux familles de protéines d'échafaudage, coopèrent pour réguler les différentes étapes de la biologie des exosomes.


Subject(s)
Exosomes , Crime , Syndecans , Tetraspanins
17.
Sci Rep ; 11(1): 4083, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33602969

ABSTRACT

Exosomal transfers represent an important mode of intercellular communication. Syntenin is a small scaffold protein that, when binding ALIX, can direct endocytosed syndecans and syndecan cargo to budding endosomal membranes, supporting the formation of intraluminal vesicles that compose the source of a major class of exosomes. Syntenin, however, can also support the recycling of these same components to the cell surface. Here, by studying mice and cells with syntenin-knock out, we identify syntenin as part of dedicated machinery that integrates both the production and the uptake of secreted vesicles, supporting viral/exosomal exchanges. This study significantly extends the emerging role of heparan sulfate proteoglycans and syntenin as key components for macromolecular cargo internalization into cells.


Subject(s)
Exosomes/metabolism , Syntenins/physiology , Animals , Exosomes/virology , Gene Expression Regulation , Gene Knockout Techniques/methods , Humans , MCF-7 Cells , Mice , Syntenins/metabolism , Transduction, Genetic
18.
Dev Cell ; 9(3): 377-88, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16139226

ABSTRACT

Syndecans are heparan sulfate proteoglycans that modulate the activity of several growth factors and cell adhesion molecules. PDZ domains in the adaptor protein syntenin interact with syndecans and with the phosphoinositide PIP(2), which is involved in the regulation of the actin cytoskeleton and membrane trafficking. Here, we show that the syntenin PDZ domain-PIP(2) interaction controls Arf6-mediated syndecan recycling through endosomal compartments. FGF receptor accompanies syndecan along the syntenin-mediated recycling pathway, in a heparan sulfate- and FGF-dependent manner. Syndecans that cannot recycle via this pathway become trapped intracellularly and inhibit cell spreading. This syntenin-mediated syndecan recycling pathway may regulate the surface availability of a number of cell adhesion and signaling molecules.


Subject(s)
ADP-Ribosylation Factors/metabolism , Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Proteoglycans/metabolism , ADP-Ribosylation Factor 6 , Cell Adhesion/physiology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Endosomes/metabolism , Endosomes/ultrastructure , Humans , Models, Biological , Phosphatidylinositol 4,5-Diphosphate/chemistry , Syndecan-2 , Syndecans , Syntenins
19.
Biochem Biophys Res Commun ; 394(3): 780-5, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20233579

ABSTRACT

In the mouse, development of the lateral semicircular canal of the inner ear is sensitive to Bmp4 heterozygosity. In the C57BL6 background 30% of the heterozygotes display circling behavior, 66% have a specific defect in the vestibular part of the inner ear, namely the constriction, interruption or absence of the lateral semicircular canal. Only mice having both ears affected display circling behavior. In the (C57BL6xCBA)N1 background, the penetrance of the canal phenotype is greatly reduced, and bilateral lateral canal defect is not sufficient to induce circling. We found association of the canal phenotype with the genotype of markers on chromosome 14 and 4, co-localizing with Ecs and Eclb identified in the Ecl mouse with similar lateral canal defects. Candidate genes to contain the causal mutation are Bmp4 on chromosome 14, and Rere on chromosome 4.


Subject(s)
Behavior, Animal , Bone Morphogenetic Protein 4/genetics , Semicircular Canals/abnormalities , Animals , Ear, Inner/abnormalities , Haplotypes , Heterozygote , Mice , Mice, Inbred C57BL , Phenotype
20.
Acta Neuropathol ; 119(2): 211-20, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19636575

ABSTRACT

Alzheimer's disease (AD) is characterized by pathological lesions such as amyloid-beta (Abeta) plaques and cerebral amyloid angiopathy. Both these lesions consist mainly of aggregated Abeta protein and this aggregation is affected by macromolecules such as heparan sulfate (HS) proteoglycans. Previous studies demonstrated that HS enhances fibrillogenesis of Abeta and that this enhancement is dependent on the degree of sulfation of HS. In addition, it has been reported that these sulfation epitopes do not occur randomly but have a defined tissue distribution. Until now, the distribution of sulfation epitopes of HS has not yet been studied in human brain. We investigated whether a specific HS epitope is associated with Abeta plaques by performing immunohistochemistry on occipital neocortical and hippocampal tissue sections from AD patients using five HS epitope-specific phage display antibodies. Antibodies recognizing highly N-sulfated HS demonstrated the highest level of staining in both fibrillar Abeta plaques and non-fibrillar Abeta plaques, whereas antibodies recognizing HS regions with a lower degree of N-sulfate modifications were only immunoreactive with fibrillar Abeta plaques. Thus, our results suggest that a larger variety of HS epitopes is associated with fibrillar Abeta plaques, but the HS epitopes associated with non-fibrillar Abeta plaques seem to be more restricted, selectively consisting of highly N-sulfated epitopes.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/pathology , Heparitin Sulfate/metabolism , Plaque, Amyloid/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Brain/metabolism , Female , Heparitin Sulfate/chemistry , Humans , Immunohistochemistry , Male , Middle Aged , Plaque, Amyloid/chemistry , Plaque, Amyloid/pathology
SELECTION OF CITATIONS
SEARCH DETAIL