Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Nat Cell Biol ; 26(1): 22-23, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38228825
2.
Article in English | MEDLINE | ID: mdl-38288044

ABSTRACT

Extracellular vesicles (EVs) play a key role both in physiological balance and homeostasis and in disease processes through their ability to participate in intercellular signaling and communication. An ever-expanding knowledge pool and a myriad of functional properties ascribed to EVs point to a new language of communication in biological systems that has opened a path for the discovery and implementation of novel diagnostic applications. EVs originate in the endosomal network and via non-random shedding from the plasma membrane by mechanisms that allow the packaging of functional cargoes, including proteins, lipids, and genetic materials. Deciphering the molecular mechanisms that govern packaging, secretion and targeted delivery of extracellular vesicle-borne cargo will be required to establish EVs as important signaling entities, especially when ascribing functional properties to a heterogeneous population of vesicles. Several molecular cascades operate within the endosomal network and at the plasma membrane that recognize and segregate cargos as a prelude to vesicle budding and release. EVs are transferred between cells and operate as vehicles in biological fluids within tissues and within the microenvironment where they are responsible for short- and long-range targeted information. In this review, we focus on the remarkable capacity of EVs to establish a dialogue between cells and within tissues, often operating in parallel to the endocrine system, we highlight selected examples of past and recent studies on the functions of EVs in health and disease.

3.
FASEB Bioadv ; 4(11): 691-693, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36349297
4.
FASEB Bioadv ; 4(5): 284-297, 2022 May.
Article in English | MEDLINE | ID: mdl-35520390

ABSTRACT

Extracellular vesicles (EVs), exosomes and microvesicles, is a burgeoning field of biological and biomedical research that may change our understanding of cell communication in plants and animals while holding great promise for the diagnosis of disease and the development of therapeutics. However, the challenge remains to develop a general hypothesis about the role of EVs in physiological homeostasis and pathobiology across kingdoms. While they can act systemically, EVs are often seen to operate locally within a microenvironment. This microenvironment is built as a collection of microunits comprised of cells that interact with each other via EV exchange, EV signaling, EV seeding, and EV disposal. We propose that microunits are part of a larger matrix at the tissue level that collectively communicates with the surrounding environment, including other end-organ systems. Herein, we offer a working model that encompasses the various facets of EV function in the context of the cell biology and physiology of multicellular organisms.

5.
J Extracell Vesicles ; 10(14): e12144, 2021 12.
Article in English | MEDLINE | ID: mdl-34919343

ABSTRACT

Extracellular vesicles (EVs) are small cargo-bearing vesicles released by cells into the extracellular space. The field of EVs has grown exponentially over the past two decades; this growth follows the realisation that EVs are not simply a waste disposal system as had originally been suggested by some, but also a complex cell-to-cell communication mechanism. Indeed, EVs have been shown to transfer functional cargo between cells and can influence several biological processes. These small biological particles are also deregulated in disease. As we approach the 75th anniversary of the first experiments in which EVs were unknowingly isolated, it seems right to take stock and look back on how the field started, and has since exploded into its current state. Here we review the early experiments, summarise key findings that have propelled the field, describe the growth of an organised EV community, discuss the current state of the field, and identify key challenges that need to be addressed.


Subject(s)
Cell-Derived Microparticles/metabolism , Exosomes/metabolism , Extracellular Vesicles/metabolism , Humans
6.
FASEB Bioadv ; 3(6): 456-458, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34124600

ABSTRACT

To keep abreast of developments in the biological sciences and in parallel fields such as medical education, FASEB BioAdvances (FBA) has created a special collections category, FBA special collections (FBA SC), that target, among other topics, emerging disciplines in the biomedical sciences. This FBA SC is focused on the emerging field of extracellular vesicles (EVs) and homeostasis. Leading investigators in the biology of EVs around the globe have contributed to this collection of articles that cover the gamut of research activities from biogenesis and secretion to physiological function.

7.
FASEB Bioadv ; 3(4): 203-204, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33842846
9.
FASEB Bioadv ; 2(9): 525, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32923986
10.
FASEB Bioadv ; 2(6): 329-330, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32617518
12.
Physiology (Bethesda) ; 34(3): 169-177, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30968753

ABSTRACT

Extracellular vesicles (EVs), cell-derived membrane structures, are secreted after fusion of endosomes with the plasma membrane (exosomes) or shed from the plasma membrane (microvesicles). EVs play a key role both in physiological balance and homeostasis and in disease processes by their ability to participate in intercellular signaling and communication.


Subject(s)
Cell Communication , Cell-Derived Microparticles/metabolism , Exosomes/metabolism , Homeostasis , Animals , Humans , Macrophages/metabolism , Melanocytes/metabolism , Mesenchymal Stem Cells/metabolism , Signal Transduction
13.
FASEB J ; 33(5): 6129-6139, 2019 05.
Article in English | MEDLINE | ID: mdl-30715917

ABSTRACT

Healthy repair of cutaneous injury is a coordinated response of inflammatory cells, secreted factors, and biologically active extracellular vesicles (EVs). Although constitutive release of EVs into biologic fluids is a hallmark of cultured cells and tumors, their payload and biologic activity appears to be tightly regulated. We show that Tre-2/Bub2/Cdc16 (TBC1) domain family member 3 (TBC1D3) drives the release of an EV population that causes a decrease in phosphorylation of the transcription factor signal transducer and activator of transcription 3 in naive recipient cells. To explore the biologic activity of EVs in vivo, we used a mouse model of sterile subcutaneous inflammation to determine the payload and biologic activity of EVs released into the microenvironment by committed myeloid lineages and stroma. Expression of TBC1D3 in macrophages altered the payload of their released EVs, including RNA-binding proteins, molecular motors, and proteins regulating secretory pathways. A wound-healing model demonstrated that closure was delayed by EVs released under the control of TBC1D3. We show that modulating the secretory repertoire of a cell regulates EV payload and biologic activity that affects outcomes in tissue repair and establishes a strategy for modifying EVs mediating specific biologic responses.-Qin, S., Dorschner, R. A., Masini, I., Lavoie-Gagne, O., Stahl, P. D., Costantini, T. W., Baird, A., Eliceiri, B. P. TBC1D3 regulates the payload and biological activity of extracellular vesicles that mediate tissue repair.


Subject(s)
Extracellular Vesicles/metabolism , GTPase-Activating Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Wound Healing , Adoptive Transfer , Animals , Extracellular Vesicles/transplantation , GTPase-Activating Proteins/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Proto-Oncogene Proteins/genetics , RAW 264.7 Cells , STAT Transcription Factors/metabolism , Signal Transduction , THP-1 Cells
14.
Essays Biochem ; 62(2): 119-124, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29765006

ABSTRACT

Over the course of the past several decades, the concept that extracellular vesicles, exosomes and microvesicles, operate as cellular "housekeepers" and as agents for communication between and among cells and tissues, has emerged into one of the most promising yet vexing problems facing the biomedical community. Already, extracellular vesicles from biological fluids are being used for diagnostic purposes and hopes abound for their use as therapeutic agents. However, the most basic mechanistic questions surrounding their biogenesis and function in cellular and tissue homeostasis remain largely unexplored. In this issue of Essays in Biochemistry, the rise of a new intercellular communications pathway is considered from many perspectives-cell biology, physiology, and pathophysiology.


Subject(s)
Exosomes/physiology , Extracellular Vesicles/physiology , Cell Communication , Humans
15.
Curr Opin Cell Biol ; 35: 69-77, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26001269

ABSTRACT

The release of extracellular vesicles (EVs) is a highly conserved process exploited by diverse organisms as a mode of intercellular communication. Vesicles of sizes ranging from 30 to 1000nm, or even larger, are generated by blebbing of the plasma membrane (microvesicles) or formed in multivesicular endosomes (MVEs) to be secreted by exocytosis as exosomes. Exosomes, microvesicles and other EVs contain membrane and cytosolic components that include proteins, lipids and RNAs, a composition that differs related to their site of biogenesis. Several mechanisms are involved in vesicle formation at the plasma membrane or in endosomes, which is reflected in their heterogeneity, size and composition. EVs have significant promise for therapeutics and diagnostics and for understanding physiological and pathological processes all of which have boosted research to find modulators of their composition, secretion and targeting.


Subject(s)
Exosomes/metabolism , Animals , Biological Transport , Cell Membrane/metabolism , Exocytosis , Extracellular Space/metabolism , Humans
16.
Diabetes ; 64(2): 353-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25157091

ABSTRACT

Increases in muscle energy needs activate AMPK and induce sarcolemmal recruitment of the fatty acid (FA) translocase CD36. The resulting rises in FA uptake and FA oxidation are tightly correlated, suggesting coordinated regulation. We explored the possibility that membrane CD36 signaling might influence AMPK activation. We show, using several cell types, including myocytes, that CD36 expression suppresses AMPK, keeping it quiescent, while it mediates AMPK activation by FA. These dual effects reflect the presence of CD36 in a protein complex with the AMPK kinase LKB1 (liver kinase B1) and the src kinase Fyn. This complex promotes Fyn phosphorylation of LKB1 and its nuclear sequestration, hindering LKB1 activation of AMPK. FA interaction with CD36 dissociates Fyn from the protein complex, allowing LKB1 to remain cytosolic and activate AMPK. Consistent with this, CD36(-/-) mice have constitutively active muscle and heart AMPK and enhanced FA oxidation of endogenous triglyceride stores. The molecular mechanism described, whereby CD36 suppresses AMPK, with FA binding to CD36 releasing this suppression, couples AMPK activation to FA availability and would be important for the maintenance of cellular FA homeostasis. Its dysfunction might contribute to the reported association of CD36 variants with metabolic complications of obesity in humans.


Subject(s)
AMP-Activated Protein Kinases/metabolism , CD36 Antigens/metabolism , Fatty Acids/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cell Line , Enzyme Activation , Gene Expression Regulation , Lipoproteins, LDL , Male , Mice , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism , Oxidation-Reduction , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Proto-Oncogene Proteins c-fyn , Triglycerides
17.
PLoS One ; 9(2): e90384, 2014.
Article in English | MEDLINE | ID: mdl-24587345

ABSTRACT

Rab5, the prototypical Rab GTPase and master regulator of the endocytic pathway, is encoded as three differentially expressed isoforms, Rab5A, Rab5B and Rab5C. Here, we examined the differential effects of Rab5 isoform silencing on cell motility and report that Rab5C, but neither Rab5A nor Rab5B, is selectively associated with the growth factor-activation of Rac1 and with enhanced cell motility. Initial observations revealed that silencing of Rab5C expression, but neither Rab5A nor Rab5C, led to spindle-shaped cells that displayed reduced formation of membrane ruffles. When subjected to a scratch wound assay, cells depleted of Rab5C, but not Rab5A or Rab5B, demonstrated reduced cell migration. U937 cells depleted of Rab5C also displayed reduced cell motility in a Transwell plate migration assay. To examine activation of Rac, HeLa cells stably expressing GFP-Rac1 were independently depleted of Rab5A, Rab5B or Rab5C and seeded onto coverslips imprinted with a crossbow pattern. 3-D GFP-Rac1 images of micro-patterned cells show that GFP-Rac1 was less localized to the cell periphery in the absence of Rab5C. To confirm the connection between Rab5C and Rac activation, HeLa cells depleted of Rab5 isoforms were starved and then stimulated with EGF. Rac1 pull-down assays revealed that EGF-stimulated Rac1 activity was significantly suppressed in Rab5C-suppressed cells. To determine whether events upstream of Rac activation were affected by Rab5C, we observed that EGF-stimulated Akt phosphorylation was suppressed in cells depleted of Rab5C. Finally, since spatio-temporal assembly/disassembly of adhesion complexes are essential components of cell migration, we examined the effect of Rab5 isoform depletion on the formation of focal adhesion complexes. Rab5C-depleted HeLa cells have significantly fewer focal adhesion foci, in accordance with the lack of persistent lamellipodial protrusions and reduced directional migration. We conclude that Rab5 isoforms selectively oversee the multiple signaling and trafficking events associated with the endocytic network.


Subject(s)
Endocytosis/genetics , Endosomes/metabolism , rab5 GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Adhesion , Cell Movement , Endosomes/ultrastructure , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , rab5 GTP-Binding Proteins/antagonists & inhibitors , rab5 GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein/genetics
18.
Infect Immun ; 81(7): 2426-36, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23630954

ABSTRACT

Pseudomonas aeruginosa, a Gram-negative opportunistic human pathogen, is a frequent cause of severe hospital-acquired infections. Effectors produced by the type III secretion system disrupt mammalian cell membrane trafficking and signaling and are integral to the establishment of P. aeruginosa infection. One of these effectors, ExoS, ADP-ribosylates several host cell proteins, including Ras and Rab GTPases. In this study, we demonstrated that Rab5 plays a critical role during early stages of P. aeruginosa invasion of J774-Eclone macrophages. We showed that live, but not heat-inactivated, P. aeruginosa inhibited phagocytosis and that this occurred in conjunction with downregulation of Rab5 activity. Inactivation of Rab5 was dependent on ExoS ADP-ribosyltransferase activity, and in J744-Eclone cells, ExoS ADP-ribosyltransferase activity caused a more severe inhibition of phagocytosis than ExoS Rho GTPase activity. Furthermore, we found that expression of Rin1, a Rab5 guanine exchange factor, but not Rabex5 and Rap6, partially reversed the inactivation of Rab5 during invasion of live P. aeruginosa. These studies provide evidence that live P. aeruginosa cells are able to influence their rate of phagocytosis in macrophages by directly regulating activation of Rab5.


Subject(s)
ADP Ribose Transferases/metabolism , Bacterial Toxins/metabolism , Macrophages/microbiology , Phagocytosis , Pseudomonas aeruginosa/pathogenicity , rab5 GTP-Binding Proteins/metabolism , ADP Ribose Transferases/genetics , Animals , Bacterial Toxins/genetics , Cell Line , Enzyme Activation , Exotoxins/genetics , Exotoxins/metabolism , Gene Expression Regulation, Enzymologic , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Intracellular Signaling Peptides and Proteins , Macrophages/enzymology , Mice , Proteolysis , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , RNA Interference , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/genetics
19.
Biochem Biophys Res Commun ; 434(2): 388-93, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23578663

ABSTRACT

Expression of the hominoid-specific oncoprotein TBC1D3 promotes enhanced cell growth and proliferation by increased activation of signal transduction through several growth factors. Recently we documented the role of CUL7 E3 ligase in growth factors-induced ubiquitination and degradation of TBC1D3. Here we expanded our study to discover additional molecular mechanisms that control TBC1D3 protein turnover. We report that TBC1D3 is palmitoylated on two cysteine residues: 318 and 325. The expression of double palmitoylation mutant TBC1D3:C318/325S resulted in protein mislocalization and enhanced growth factors-induced TBC1D3 degradation. Moreover, ubiquitination of TBC1D3 via CUL7 E3 ligase complex was increased by mutating the palmitoylation sites, suggesting that depalmitoylation of TBC1D3 makes the protein more available for ubiquitination and degradation. The results reported here provide novel insights into the molecular mechanisms that govern TBC1D3 protein degradation. Dysregulation of these mechanisms in vivo could potentially result in aberrant TBC1D3 expression and promote oncogenesis.


Subject(s)
GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Lipoylation , Proteolysis , Proto-Oncogene Proteins/metabolism , Ubiquitination , Cell Membrane/metabolism , Cullin Proteins/genetics , Cullin Proteins/metabolism , Cysteine/metabolism , Cytosol/metabolism , Electrophoresis, Polyacrylamide Gel , GTPase-Activating Proteins/genetics , HeLa Cells , Humans , Microscopy, Fluorescence , Multiprotein Complexes/metabolism , Protein Binding , Protein Transport , Proto-Oncogene Proteins/genetics , Transfection
20.
J Cell Biol ; 200(4): 367-71, 2013 Feb 18.
Article in English | MEDLINE | ID: mdl-23420870

ABSTRACT

Exosomes are extracellular membrane vesicles whose biogenesis by exocytosis of multivesicular endosomes was discovered in 1983. Since their discovery 30 years ago, it has become clear that exosomes contribute to many aspects of physiology and disease, including intercellular communication. We discuss the initial experiments that led to the discovery of exosomes and highlight some of the exciting current directions in the field.


Subject(s)
Cell Polarity/physiology , Endocytosis/physiology , Models, Biological , Saccharomyces/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...