Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Nat Methods ; 18(9): 1013-1026, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34446922

RESUMO

Extracellular vesicles (EVs) are nano-sized lipid bilayer vesicles released by virtually every cell type. EVs have diverse biological activities, ranging from roles in development and homeostasis to cancer progression, which has spurred the development of EVs as disease biomarkers and drug nanovehicles. Owing to the small size of EVs, however, most studies have relied on isolation and biochemical analysis of bulk EVs separated from biofluids. Although informative, these approaches do not capture the dynamics of EV release, biodistribution, and other contributions to pathophysiology. Recent advances in live and high-resolution microscopy techniques, combined with innovative EV labeling strategies and reporter systems, provide new tools to study EVs in vivo in their physiological environment and at the single-vesicle level. Here we critically review the latest advances and challenges in EV imaging, and identify urgent, outstanding questions in our quest to unravel EV biology and therapeutic applications.


Assuntos
Vesículas Extracelulares , Microscopia/métodos , Animais , Corantes/química , Epitopos , Vesículas Extracelulares/química , Vesículas Extracelulares/patologia , Vesículas Extracelulares/fisiologia , Corantes Fluorescentes/química , Humanos
2.
EMBO J ; 30(11): 2115-29, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21527913

RESUMO

The ubiquitous Epstein Barr virus (EBV) exploits human B-cell development to establish a persistent infection in ∼90% of the world population. Constitutive activation of NF-κB by the viral oncogene latent membrane protein 1 (LMP1) has an important role in persistence, but is a risk factor for EBV-associated lymphomas. Here, we demonstrate that endogenous LMP1 escapes degradation upon accumulation within intraluminal vesicles of multivesicular endosomes and secretion via exosomes. LMP1 associates and traffics with the intracellular tetraspanin CD63 into vesicles that lack MHC II and sustain low cholesterol levels, even in 'cholesterol-trapping' conditions. The lipid-raft anchoring sequence FWLY, nor ubiquitylation of the N-terminus, controls LMP1 sorting into exosomes. Rather, C-terminal modifications that retain LMP1 in Golgi compartments preclude assembly within CD63-enriched domains and/or exosomal discharge leading to NF-κB overstimulation. Interference through shRNAs further proved the antagonizing role of CD63 in LMP1-mediated signalling. Thus, LMP1 exploits CD63-enriched microdomains to restrain downstream NF-κB activation by promoting trafficking in the endosomal-exosomal pathway. CD63 is thus a critical mediator of LMP1 function in- and outside-infected (tumour) cells.


Assuntos
Antígenos CD/metabolismo , Endossomos/metabolismo , Exossomos/metabolismo , Herpesvirus Humano 4/imunologia , NF-kappa B/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Proteínas da Matriz Viral/metabolismo , Linhagem Celular , Herpesvirus Humano 4/patogenicidade , Humanos , Ligação Proteica , Transporte Proteico , Tetraspanina 30
3.
Nat Cell Biol ; 2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38886558

RESUMO

Extracellular vesicles such as exosomes are now recognized as key players in intercellular communication. Their role is influenced by the specific repertoires of proteins and lipids, which are enriched when they are generated as intraluminal vesicles (ILVs) in multivesicular endosomes. Here we report that a key component of small extracellular vesicles, the tetraspanin CD63, sorts cholesterol to ILVs, generating a pool that can be mobilized by the NPC1/2 complex, and exported via exosomes to recipient cells. In the absence of CD63, cholesterol is retrieved from the endosomes by actin-dependent vesicular transport, placing CD63 and cholesterol at the centre of a balance between inward and outward budding of endomembranes. These results establish CD63 as a lipid-sorting mechanism within endosomes, and show that ILVs and exosomes are alternative providers of cholesterol.

4.
iScience ; 26(8): 107412, 2023 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-37575190

RESUMO

The human cytomegalovirus (HCMV)-encoded chemokine receptor US28 contributes to various aspects of the viral life cycle and promotes immune evasion by scavenging chemokines from the microenvironment of HCMV-infected cells. In contrast to the plasma membrane localization of most human chemokine receptors, US28 has a predominant intracellular localization. In this study, we used immunofluorescence and electron microscopy to determine the localization of US28 upon exogenous expression, as well as in HCMV-infected cells. We observed that US28 localizes to late endosomal compartments called multivesicular bodies (MVBs), where it is sorted in intraluminal vesicles. Live-cell total internal reflection fluorescence (TIRF) microscopy revealed that US28-containing MVBs can fuse with the plasma membrane, resulting in the secretion of US28 on exosomes. Exosomal US28 binds the chemokines CX3CL1 and CCL5, and US28-containing exosomes inhibited the CX3CL1-CX3CR1 signaling axis. These findings suggest that exosomal release of US28 contributes to chemokine scavenging and immune evasion by HCMV.

5.
bioRxiv ; 2023 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-37645762

RESUMO

The extracellular matrix (ECM) supports blood vessel architecture and functionality and undergoes active remodelling during vascular repair and atherogenesis. Vascular smooth muscle cells (VSMCs) are essential for vessel repair and, via their secretome, are able to invade from the vessel media into the intima to mediate ECM remodelling. Accumulation of fibronectin (FN) is a hallmark of early vascular repair and atherosclerosis and here we show that FN stimulates VSMCs to secrete small extracellular vesicles (sEVs) by activating the ß1 integrin/FAK/Src pathway as well as Arp2/3-dependent branching of the actin cytoskeleton. Spatially, sEV were secreted via filopodia-like cellular protrusions at the leading edge of migrating cells. We found that sEVs are trapped by the ECM in vitro and colocalise with FN in symptomatic atherosclerotic plaques in vivo. Functionally, ECM-trapped sEVs induced the formation of focal adhesions (FA) with enhanced pulling forces at the cellular periphery. Proteomic and GO pathway analysis revealed that VSMC-derived sEVs display a cell adhesion signature and are specifically enriched with collagen VI. In vitro assays identified collagen VI as playing the key role in cell adhesion and invasion. Taken together our data suggests that the accumulation of FN is a key early event in vessel repair acting to promote secretion of collage VI enriched sEVs by VSMCs. These sEVs stimulate migration and invasion by triggering peripheral focal adhesion formation and actomyosin contraction to exert sufficient traction forces to enable VSMC movement within the complex vascular ECM network.

6.
J Cell Biol ; 221(12)2022 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-36136097

RESUMO

Exosomes are endosome-derived extracellular vesicles involved in intercellular communication. They are generated as intraluminal vesicles within endosomal compartments that fuse with the plasma membrane (PM). The molecular events that generate secretory endosomes and lead to the release of exosomes are not well understood. We identified a subclass of non-proteolytic endosomes at prelysosomal stage as the compartment of origin of CD63 positive exosomes. These compartments undergo a Rab7a/Arl8b/Rab27a GTPase cascade to fuse with the PM. Dynamic endoplasmic reticulum (ER)-late endosome (LE) membrane contact sites (MCS) through ORP1L have the distinct capacity to modulate this process by affecting LE motility, maturation state, and small GTPase association. Thus, exosome secretion is a multi-step process regulated by GTPase switching and MCS, highlighting the ER as a new player in exosome-mediated intercellular communication.


Assuntos
Retículo Endoplasmático , Endossomos , Exossomos , Proteínas rab de Ligação ao GTP , Transporte Biológico , Comunicação Celular , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Endossomos/enzimologia , Exossomos/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
7.
Adv Drug Deliv Rev ; 176: 113815, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34058284

RESUMO

Extracellular Vesicles (EVs) are released during various pathophysiological processes and reflect the state of their cell of origin. Once released, they can propagate through biological fluids, target cells, deliver their content and elicit functional responses. These specific features would allow their harnessing as biomarkers, drug nano-vehicles and therapeutic intrinsic modulators. However, the further development of their potential therapeutic application is hampered by the lack of knowledge about how EVs behave in vivo. Recent advances in the field of imaging EVs in vivo now allow live-tracking of endogenous and exogenous EV in various model organisms at high spatiotemporal resolution to define their distribution, half-life and fate. This review highlights current imaging tools available to image EVs in vivo and how live imaging especially in the zebrafish embryo can bring further insights into the characterization of EVs dynamics, biodistribution and functions to potentiate their development for therapeutic applications.


Assuntos
Vesículas Extracelulares/metabolismo , Modelos Animais , Peixe-Zebra/metabolismo , Animais , Biomarcadores/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Embrião não Mamífero/metabolismo , Humanos , Distribuição Tecidual
8.
FASEB Bioadv ; 3(11): 918-929, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34761174

RESUMO

To harmoniously coordinate the activities of all its different cell types, a multicellular organism critically depends on intercellular communication. One recently discovered mode of intercellular cross-talk is based on the exchange of "extracellular vesicles" (EVs). EVs are nano-sized heterogeneous lipid bilayer vesicles enriched in a variety of biomolecules that mediate short- and long-distance communication between different cells, and between cells and their environment. Numerous studies have demonstrated important aspects pertaining to the dynamics of their release, their uptake, and sub-cellular fate and roles in vitro. However, to demonstrate these and other aspects of EV biology in a relevant, fully physiological context in vivo remains challenging. In this review we analyze the state of the art of EV imaging in vivo, focusing in particular on zebrafish as a promising model to visualize, study, and characterize endogenous EVs in real-time and expand our understanding of EV biology at cellular and systems level.

9.
Nat Commun ; 12(1): 4389, 2021 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-34282141

RESUMO

Despite their roles in intercellular communications, the different populations of extracellular vesicles (EVs) and their secretion mechanisms are not fully characterized: how and to what extent EVs form as intraluminal vesicles of endocytic compartments (exosomes), or at the plasma membrane (PM) (ectosomes) remains unclear. Here we follow intracellular trafficking of the EV markers CD9 and CD63 from the endoplasmic reticulum to their residency compartment, respectively PM and late endosomes. We observe transient co-localization at both places, before they finally segregate. CD9 and a mutant CD63 stabilized at the PM are more abundantly released in EVs than CD63. Thus, in HeLa cells, ectosomes are more prominent than exosomes. By comparative proteomic analysis and differential response to neutralization of endosomal pH, we identify a few surface proteins likely specific of either exosomes (LAMP1) or ectosomes (BSG, SLC3A2). Our work sets the path for molecular and functional discrimination of exosomes and small ectosomes in any cell type.


Assuntos
Exossomos/metabolismo , Tetraspanina 29/metabolismo , Tetraspanina 30/metabolismo , Comunicação Celular , Membrana Celular/metabolismo , Endossomos/metabolismo , Vesículas Extracelulares/metabolismo , Cadeia Pesada da Proteína-1 Reguladora de Fusão , Técnicas de Inativação de Genes , Células HeLa , Humanos , Proteínas de Membrana/metabolismo , Transporte Proteico , Proteômica
10.
Nat Protoc ; 15(1): 102-121, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31836866

RESUMO

Exosomes are small extracellular vesicles with a diameter of 40-150 nm, and are implicated in cellular homeostasis and cell-cell communication. They can be secreted in bulk in response to cell-extrinsic and cell-intrinsic signals that cause multivesicular body (MVB) fusion with the plasma membrane (PM). However, research on the regulation of exosome release is hampered by the failure of current methods to capture the dynamics of exosome release. Here we describe how live imaging with tetraspanin-based pH-sensitive fluorescent reporters can quantify the MVB-PM fusion rate of single cells. Our approach enables identification of exogenous stimuli, signaling pathways, and fusion complexes, and can map subcellular sites of fusion events. In addition, dual-color imaging can be used to assess simultaneous release of different cargo by MVB exocytosis. This protocol describes the complete imaging experiment, consisting of transient expression of tetraspanin reporters (2 d), live-cell (dual-color) total internal reflection fluorescence microscopy (30-60 min per condition), and semiautomatic image analysis by using a newly developed ImageJ macro (±30 min per condition).


Assuntos
Exossomos/metabolismo , Fusão de Membrana , Imagem Molecular/métodos , Análise de Célula Única , Células HeLa , Humanos , Fatores de Tempo
11.
Trends Cell Biol ; 29(10): 770-776, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31400828

RESUMO

Extracellular vesicles (EVs) circulate in the body fluids of all organisms where they participate in intercellular cross-organ communication. Tracking and understanding these nanosized objects has been hampered by the low resolution of imaging techniques and by the lack of appropriate animal models. The use of zebrafish embryos permits visualization of EVs at unprecedented spatiotemporal resolution using light and electron microscopy. This enables the study of endogenous physiological EVs and pathological EVs side by side, and further unravels their mechanisms of biogenesis, biodistribution, and target cells throughout the organism. These developments will contribute to a better understanding of the in vivo (patho)physiology of EVs.


Assuntos
Células Endoteliais/citologia , Vesículas Extracelulares/metabolismo , Microscopia/métodos , Animais , Comunicação Celular , Modelos Animais de Doenças , Exossomos/metabolismo , Humanos , Neoplasias/patologia , Análise Espaço-Temporal , Peixe-Zebra
12.
Dev Cell ; 48(4): 573-589.e4, 2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30745143

RESUMO

Extracellular vesicles (EVs) are released by most cell types but providing evidence for their physiological relevance remains challenging due to a lack of appropriate model organisms. Here, we developed an in vivo model to study EV function by expressing CD63-pHluorin in zebrafish embryos. A combination of imaging methods and proteomic analysis allowed us to study biogenesis, composition, transfer, uptake, and fate of individual endogenous EVs. We identified a subpopulation of EVs with exosome features, released in a syntenin-dependent manner from the yolk syncytial layer into the blood circulation. These exosomes are captured, endocytosed, and degraded by patrolling macrophages and endothelial cells in the caudal vein plexus (CVP) in a scavenger receptor- and dynamin-dependent manner. Interference with exosome biogenesis affected CVP growth, suggesting a role in trophic support. Altogether, our work represents a system for studying endogenous EV function in vivo with high spatiotemporal accuracy, demonstrating functional inter-organ communication by exosomes.


Assuntos
Transporte Biológico/fisiologia , Células Endoteliais/metabolismo , Exossomos/metabolismo , Vesículas Extracelulares/metabolismo , Animais , Células Cultivadas , Proteômica/métodos , Peixe-Zebra
13.
Dev Cell ; 48(4): 554-572.e7, 2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30745140

RESUMO

Tumor extracellular vesicles (EVs) mediate the communication between tumor and stromal cells mostly to the benefit of tumor progression. Notably, tumor EVs travel in the bloodstream, reach distant organs, and locally modify the microenvironment. However, visualizing these events in vivo still faces major hurdles. Here, we describe an approach for tracking circulating tumor EVs in a living organism: we combine chemical and genetically encoded probes with the zebrafish embryo as an animal model. We provide a first description of tumor EVs' hemodynamic behavior and document their intravascular arrest. We show that circulating tumor EVs are rapidly taken up by endothelial cells and blood patrolling macrophages and subsequently stored in degradative compartments. Finally, we demonstrate that tumor EVs activate macrophages and promote metastatic outgrowth. Overall, our study proves the usefulness and prospects of zebrafish embryo to track tumor EVs and dissect their role in metastatic niches formation in vivo.


Assuntos
Células Endoteliais/citologia , Vesículas Extracelulares/metabolismo , Neoplasias/patologia , Microambiente Tumoral/fisiologia , Animais , Comunicação Celular/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Exossomos/metabolismo , Células Estromais/metabolismo , Peixe-Zebra
14.
J Extracell Vesicles ; 8(1): 1684862, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31762963

RESUMO

Paracrine and endocrine roles have increasingly been ascribed to extracellular vesicles (EVs) generated by multicellular organisms. Central to the biogenesis, content, and function of EVs are their delimiting lipid bilayer membranes. To evaluate research progress on membranes and EVs, the International Society for Extracellular Vesicles (ISEV) conducted a workshop in March 2018 in Baltimore, Maryland, USA, bringing together key opinion leaders and hands-on researchers who were selected on the basis of submitted applications. The workshop was accompanied by two scientific surveys and covered four broad topics: EV biogenesis and release; EV uptake and fusion; technologies and strategies used to study EV membranes; and EV transfer and functional assays. In this ISEV position paper, we synthesize the results of the workshop and the related surveys to outline important outstanding questions about EV membranes and describe areas of consensus. The workshop discussions and survey responses reveal that while much progress has been made in the field, there are still several concepts that divide opinion. Good consensus exists in some areas, including particular aspects of EV biogenesis, uptake and downstream signalling. Areas with little to no consensus include EV storage and stability, as well as whether and how EVs fuse with target cells. Further research is needed in these key areas, as a better understanding of membrane biology will contribute substantially towards advancing the field of extracellular vesicles.

15.
J Extracell Vesicles ; 4: 26334, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25865256

RESUMO

The Epstein-Barr virus (EBV)-encoded oncoprotein latent membrane protein 1 (LMP1) constitutively activates nuclear factor κB (NFκB) from intracellular membranes to promote cell growth and survival. LMP1 associates with CD63 in intracellular membranes and is released via exosomes. Whether tumour necrosis factor (TNF) receptor-associated factors (TRAFs) mediate LMP1 NFκB signalling from endosomes and modulate exosomal sorting is unknown. In this article, we show that LMP1-TRAF2 signalling complexes accumulate at endosomes in a palmitoylation-dependent manner, thereby driving LMP1-dependent oncogenicity. Palmitoylation is a reversible post-translational modification and is considered to function as a membrane anchor for proteins. Mutagenesis studies showed that LMP1-TRAF2 trafficking to endosomes is dependent on one single cysteine residue (C78), a known palmitoylation site of LMP1. Notably, growth assays in soft agar revealed that oncogenic properties of the palmitoylation-deficient LMP1 mutant C78A were diminished compared to wild-type LMP1. Since LMP1 recruitment of TRAF2 and downstream NFκB signalling were not affected by a disturbance in palmitoylation, the specific localization of LMP1 at endosomal membranes appears crucial for its transforming potential. The importance of palmitoylation for trafficking to and signalling from endosomal membranes was not restricted to LMP1, as similar observations were made for the cellular oncoproteins Src and Fyn. Despite abundant LMP1-TRAF2 association at endosomal membranes TRAF2 could not be detected in exosomes by Western blotting or proteomics. Interestingly, point mutations that prevented TRAF binding strongly promoted the sorting and release of LMP1 via exosomes. These observations reveal that LMP1-TRAF2 complexes at endosomes support oncogenic NFκB activation and suggest that LMP1 dissociates from the activated signalling complexes upon sorting into intraluminal vesicles. We propose that "signalling endosomes" in EBV-infected tumour cells can fuse with the plasma membrane, explaining LMP1 release via exosomes.

16.
Stem Cell Res Ther ; 6: 127, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26129847

RESUMO

INTRODUCTION: Administration of mesenchymal stem cells (MSCs) represents a promising treatment option for patients suffering from immunological and degenerative disorders. Accumulating evidence indicates that the healing effects of MSCs are mainly related to unique paracrine properties, opening opportunities for secretome-based therapies. Apart from soluble factors, MSCs release functional small RNAs via extracellular vesicles (EVs) that seem to convey essential features of MSCs. Here we set out to characterize the full small RNAome of MSC-produced exosomes. METHODS: We set up a protocol for isolating exosomes released by early passage adipose- (ASC) and bone marrow-MSCs (BMSC) and characterized them via electron microscopy, protein analysis and small RNA-sequencing. We developed a bioinformatics pipeline to define the exosome-enclosed RNA species and performed the first complete small RNA characterization of BMSCs and ASCs and their corresponding exosomes in biological replicates. RESULTS: Our analysis revealed that primary ASCs and BMSCs have highly similar small RNA expression profiles dominated by miRNAs and snoRNAs (together 64-71 %), of which 150-200 miRNAs are present at physiological levels. In contrast, the miRNA pool in MSC exosomes is only 2-5 % of the total small RNAome and is dominated by a minor subset of miRNAs. Nevertheless, the miRNAs in exosomes do not merely reflect the cellular content and a defined set of miRNAs are overrepresented in exosomes compared to the cell of origin. Moreover, multiple highly expressed miRNAs are precluded from exosomal sorting, consistent with the notion that these miRNAs are involved in functional repression of RNA targets. While ASC and BMSC exosomes are similar in RNA class distribution and composition, we observed striking differences in the sorting of evolutionary conserved tRNA species that seems associated with the differentiation status of MSCs, as defined by Sox2, POU5F1A/B and Nanog expression. CONCLUSIONS: We demonstrate that primary MSCs release small RNAs via exosomes, which are increasingly implicated in intercellular communications. tRNAs species, and in particular tRNA halves, are preferentially released and their specific sorting into exosomes is related to MSC tissue origin and stemness. These findings may help to understand how MSCs impact neighboring or distant cells with possible consequences for their therapeutic usage.


Assuntos
Tecido Adiposo/citologia , Células da Medula Óssea/citologia , Exossomos/metabolismo , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , RNA de Transferência/metabolismo , Regiões 3' não Traduzidas , Sequência de Bases , Diferenciação Celular , Células Cultivadas , Exossomos/genética , Humanos , Células-Tronco Mesenquimais/citologia , Microscopia Eletrônica , RNA/química , RNA/isolamento & purificação , Análise de Sequência de RNA , Transcriptoma
17.
Methods Mol Biol ; 1024: 53-68, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23719942

RESUMO

The isolation and analysis of microRNAs (miRNAs) contained in microvesicles and in particular nano-sized exosomes has become an increasingly important tool to understand their widespread impact in various fundamental and interactive cellular processes. Fundamental studies regarding exosome biogenesis and miRNA sorting may ultimately unravel their potency as a promising class of highly specific disease biomarkers. Here we describe the methods and protocols used in our laboratory to isolate and purify exosomes, how we extract the (small) RNA content, how to analyze copy numbers, and finally how to measure exosome-mediated transfer of these molecules into recipient cells. Our techniques have been optimized for the detection of Epstein-Barr virus (EBV)-encoded miRNAs that are loaded into exosomes. We discuss how a focus on EBV-miRNA detection may yield important new clues into exosome-mediated cross talk by B cells in humans.


Assuntos
Linfócitos B/química , Exossomos/química , Herpesvirus Humano 4/isolamento & purificação , MicroRNAs/isolamento & purificação , RNA Viral/isolamento & purificação , Linfócitos B/citologia , Linfócitos B/virologia , Comunicação Celular , Técnicas de Cocultura , Cultura em Câmaras de Difusão , Exossomos/genética , Corantes Fluorescentes , Células HEK293 , Células HeLa , Herpesvirus Humano 4/genética , Humanos , MicroRNAs/genética , Transporte de RNA , RNA Viral/genética , Ultracentrifugação
18.
Commun Integr Biol ; 5(1): 88-93, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22482020

RESUMO

Tight control of intracellular signaling is essential for developmental processes such as cell differentiation, migration but also for maintaining tissue homeostasis. Disruption in the control of these signaling pathways can result in cell death (apoptosis), anergy or uncontrolled cell proliferation and growth leading to cancer. In multicellular organisms, timely termination of signaling is thus equally important as initiation. Known pathways for downregulating membrane receptor-mediated signaling are mediated via specialized endosomal organelles known as lysosomes and proteosomes that degrade such proteins in the cytoplasm. An alternative pathway for attenuating receptor-mediated signaling was recently discovered independently by the group of M. Caplan and our own group.1,2 It appears that apart from the classical protein degradation machineries, the release of signaling proteins also effectively restricts signaling of at least two major signal transduction routes; the canonical Wnt/ß-catenin and NFκB pathways. Expelling proteins from the cell, rather than coordinated degradation in lysosomes may involve defined protein modifications, such as ubiquitination, myristyolation, and/or palmitoylation, but little experimental data are currently available. Although the secretion of proteins via exosomes starts by accumulation within multivesicular bodies (MVBs), a key distinction with degredatory MVBs is that exosome-producing MVBs seem to preferentially fuse with the plasmamembrane (Fig. 1). Here we discuss the latest developments in the biology of exosomes and their unexpected effect on intracellular signal transduction.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa