Extracellular vesicle

Last updated

Extracellular vesicles (EVs) are lipid bilayer-delimited particles that are naturally released from almost all types of cells but, unlike a cell, cannot replicate. EVs range in diameter from near the size of the smallest physically possible unilamellar liposome (around 20-30 nanometers) to as large as 10 microns or more, although the vast majority of EVs are smaller than 200 nm. EVs can be divided according to size and synthesis route into exosomes, microvesicles and apoptotic bodies. [1] [2] The composition of EVs varies depending on their parent cells, encompassing proteins (e.g., adhesion molecules, cytoskeletons, cytokines, ribosomal proteins, growth factors, and metabolic enzymes), lipids (including cholesterol, lipid rafts, and ceramides), nucleic acids (such as DNA, mRNA, and miRNA), metabolites, and even organelles. [3] [4] Most cells that have been studied to date are thought to release EVs, including some archaeal, bacterial, fungal, and plant cells that are surrounded by cell walls. A wide variety of EV subtypes have been proposed, defined variously by size, biogenesis pathway, cargo, cellular source, and function, leading to a historically heterogenous nomenclature including terms like exosomes and ectosomes.

Contents

Numerous functions of EVs have been established or postulated. The first evidence for the existence of EVs was enabled by the ultracentrifuge, the electron microscope, and functional studies of coagulation in the mid-20th century. A sharp increase in interest in EVs occurred in the first decade of the 21st century following the discovery that EVs could transfer nucleic acids such as RNA from cell to cell. Associated with EVs from certain cells or tissues, nucleic acids could be easily amplified as markers of disease and also potentially traced back to a cell of origin, such as a tumor cell. When EVs are taken up by other cells, they may alter the behaviour of the recipient cell, for instance EVs released by colorectal cancer cells increase migration of fibroblasts and thus EVs are of importance in forming tumour landscapes. [5] This discovery also implied that EVs could be used for therapeutic purposes, such as delivering nucleic acids or other cargo to diseased tissue. This growing interest was paralleled by formation of companies and funding programs focused on development of EVs as biomarkers or therapies of disease, the founding of an International Society for Extracellular Vesicles (ISEV), and establishment of a scientific journal devoted to the field, the Journal of Extracellular Vesicles .

History

Evidence for the existence of EVs and their functions was first gathered by combined applications of ultracentrifugation, electron microscopy, and functional studies during the mid-20th century. [6] Ultracentrifuged pellets from blood plasma were reported to have procoagulant properties by Erwin Chargaff and Randolph West in 1946. [7] The platelet derivation and lipid-containing nature of these particles was further articulated by Peter Wolf. [8] Around the same time, H. Clarke Anderson and Ermanno Bonucci separately described the calcifying properties of EVs in bone matrix. [9] [10]

Although the extracellular and vesicular properties of EVs had been recognized by numerous groups by the 1970s, the term "extracellular vesicle" was first used in a manuscript title in 1971. [10] This electron microscopy study of the flagellate freshwater alga 'Ochromonas danica' reported release of EVs from membranes including those of flagella. Soon thereafter, EVs were seen to be released from follicular thyroid cells of the bat during arousal from hibernation, suggesting the possible involvement of EVs in endocrine processes. [11] Reports of EVs in intestinal villi samples and, for the first time, in material from human cancer (adenoma) [12] [13] [14] [15] referred back to even earlier publications that furnished similar evidence, although conclusions about EV release had not then been drawn. EVs were also described in bovine serum and cell culture conditioned medium [15] [14] with distinctions made between "vesicles of the multivesicular body" and "microvesicles." [15] [6] These studies further noted the similarities of EVs and enveloped viruses. [16]

In the early- to mid-1980s, the Stahl and Johnstone labs forged a deeper understanding of the release of EVs from reticulocytes, [17] [18] [19] while progress was also made on EVs shed from tumor cells. [20] [6] The reticulocyte research, in particular, showed that EVs could be released not only from the plasma membrane or surface of the cell, but also by fusion of the multivesicular body with the plasma membrane. During this time, EVs were described by many names, sometimes in the same manuscript, such as "shedding vesicles," "membrane fragments," "plasma membrane vesicles," "micro-vesicles/microvesicles," "exosomes," (previously used for mobile, transforming DNA elements in model organisms Drosophila and Neurospora [21] [22] ), "inclusion vesicles," and more, or referred to by organ of origin, such as "prostasomes" that were found to enhance sperm motility in semen. [23] [6]

The involvement of EVs in immune responses became increasingly clear in the 1990s with findings of the group of Graça Raposo and others. [24] [6] A clinical trial of dendritic cell-derived EVs was performed in France just before the turn of the century.[ citation needed ] Cells of the immune system were found capable of transferring transmembrane proteins via EVs. For example, the HIV co-receptors CCR5 and CXCR4 could be transferred from an HIV-susceptible cell to a refractory cell by "microparticles," rendering the recipient cell susceptible to infection. [25] [26]

Beginning in 2006, several laboratories reported that EVs contain nucleic acids and have the ability to transfer them from cell to cell. [27] [28] [29] [30] [31] [32] [6] [33] Nucleic acids including DNAs and RNAs were even found to be functional in the recipient cell. Whether carrying DNA, RNA, surface molecules, or other factors, the involvement of EVs in cancer progression aroused considerable interest, [34] leading to hypotheses that specific EVs could target specific cells due to "codes" displayed on their surface; [35] create or enhance a metastatic niche; [36] betray the presence of specific cancers; [37] or be used as a therapy to target cancer cells. [38] Meanwhile, strides were made in the understanding of vesicle biogenesis and subtypes. [39] [40] [41] [42]

Rapid growth of the EV research community in the early 2000s led to the creation of the International Society for Extracellular Vesicles (ISEV), which has led efforts for rigor and standardization in the field including establishment of the Journal of Extracellular Vesicles. A plethora of national and regional EV societies have also been formed. In 2012, the Director's Office of the US National Institutes of Health (NIH) announced a program for funding of EV and extracellular RNA studies, the Extracellular RNA Communication Consortium (ERCC), [43] which subsequently invested >USD 100 million in EV research. A second round of funding was announced in 2018. Commercial investment in EV diagnostics and therapeutics also grew during this time.[ citation needed ]

Biogenesis

Extracellular vesicles and particles (EVPs) are released by cells in different shapes and sizes. [1] Diverse EV subtypes have been proposed, with names such as ectosomes, microvesicles, microparticles, exosomes, oncosomes, apoptotic bodies, and more. [6] [44] [45] [46] These EV subtypes have been defined by various, often overlapping, definitions, based mostly on biogenesis (cell pathway, cell or tissue identity, condition of origin). [47] However, EV subtypes may also be defined by size, constituent molecules, function, or method of separation. Because of the bewildering and sometimes contradictory definitions of different EV subtypes, the current scientific consensus is that "extracellular vesicle" and variations thereon are the preferred nomenclature unless specific biogenetic origin can be demonstrated. [47] Subtypes of EVs may be defined by:

"a) physical characteristics of EVs, such as size ("small EVs" (sEVs) and "medium/large EVs" (m/lEVs), with ranges defined, for instance, respectively, <100nm or <200nm [small], or >200nm [large and/or medium]) or density (low, middle, high, with each range defined); b) biochemical composition (CD63+/CD81+- EVs, Annexin A5-stained EVs, etc.); or c) descriptions of conditions or cell of origin (podocyte EVs, hypoxic EVs, large oncosomes, apoptotic bodies)." [47]

Plasma membrane origin

The terms "ectosome," "microvesicle" (MV), and "microparticle" (MP) refer to particles released from the surface of cells. Technically, the platelets of certain vertebrates (which bud from megakaryocytes), as well as red blood cells (e.g., of adult humans) also fulfill the consensus definition of EVs. [47] Especially in the field of platelet research, MP has been the standard nomenclature. Formation of ectosomes may in some cases result from directed processes, and in others from shear forces or adherence of the PM to a surface.[ citation needed ]

Endosomal origin

Exosome biogenesis begins with pinching off of endosomal invaginations into the multivesicular body (MVB), forming intraluminal vesicles (ILVs). If the MVB fuses with the plasma membrane, the ILVs are released as "exosomes." The first publication to use the term "exosome" for EVs presented it as a synonym for "micro-vesicle." [48] The term has also been used for EVs within specific size ranges, EVs separated using specific methods, or even all EVs.

Apoptotic bodies

Apoptotic bodies are EVs that are released by dying cells undergoing apoptosis. Since apoptotic cells tend to display phosphatidylserine (PS) in the outer bilayer of the cell membrane, apoptotic bodies tend to externalize PS, although other EVs may also do so. Apoptotic bodies may be quite large (microns in diameter) but may also measure in the submicron range.

Large oncosomes

In addition to the very large EVs released during apoptosis, micron-sized EVs may be produced by cancer cells, neurons, and other cells. When produced by cancer cells, these particles are termed "large oncosomes" [49] [50] and may reach 20 microns or more in diameter. Large oncosomes can attain sizes comparable to individual cells, but they do not contain full nuclei. They have been shown to contribute to metastasis in a mouse model and a human fibroblast cell culture model of prostate cancer. [51] Cellular internalization of large oncosomes can reprogram non-neoplastic brain cells to divide and migrate in primary tissue culture, and higher numbers of large oncosomes isolated from blood samples from glioblastoma patients were correlated with more advanced disease progression. [52]

Exophers

Exophers are a class of large EV, approximately four microns in diameter, observed in model organisms ranging from Caenorhabditis elegans [53] to mice. [54] When genetically modified to express aggregating proteins, neurons were observed to sequester the aggregates into a portion of the cell and release them within a large EV called an exopher. They are hypothesized to be a mechanism for disposal of unwanted cellular material including protein aggregates and damaged organelles. [53] Exophers can remain connected to the cell body by a thin, membranous filament resembling a tunneling nanotube. [53]

Migrasomes

Migrasomes are large membrane-bound EVs, ranging from 0.5 to 3 microns in diameter, that form at the ends of retraction fibers left behind when cells migrate in a process termed "migracytosis." Migrasomes can continue to fill with cytosol and expand even as the originating cell moves away. Migrasomes were first observed in rat kidney cell culture, but they are produced by mouse and human cells as well. [55] Damaged mitochondria can be expelled from migrating cells inside of migrasomes, suggesting a functional role for this EV in mitochondrial homeostasis. [56]

Enveloped viruses

Enveloped viruses are a type of EV produced under the influence of viral infection. That is, the virion is composed of cellular membranes but contains proteins and nucleic acids produced from the viral genome. Some enveloped viruses can infect other cells even without a functional virion, when genomic material is transferred via EVs. Certain non-enveloped viruses may also reproduce with assistance from EVs. [57]

Isolation

Studying EVs and their cargo typically requires separation from a biological matrix (such as a complex fluid or tissue) so that the uniquely EV components can be analyzed. Many approaches have been used, including differential ultracentrifugation, density gradient ultracentrifugation, size exclusion chromatography, ultrafiltration, capillary electrophoresis, asymmetric-flow field-flow fractionation, and affinity/immunoaffinity capture methods. [47] [58] [6] [59] [60] [61] [33] Each method has its own recovery and purity outcomes: that is, what percentage of input EVs are obtained, and the ratio of "true" EV components to co-isolates. EV separation can also be influenced by pre-analytical variables. [62] [63] [64] [65]

Characterization

Population-level EV analysis

Separated or concentrated populations of EVs may be characterized by several means. Total concentration of molecules in categories such as protein, lipid or nucleic acid. Total particle counts in a preparation can also be estimated, for example by light-scattering techniques. Each measurement technology may have a specific size range for accurate quantitation, and very small EVs (<100 nm diameter) are not detected by many technologies. Molecular "fingerprints" of populations can be obtained by "omics" technologies like proteomics, lipidomics, and RNomics, or by techniques like Raman spectroscopy. Overall levels of unique molecules can also be measured in the population, such as tetraspanins, phosphatidylserine, or species of RNA. It has been proposed that purity of an EV preparation can be estimated by examining the ratio of one population-level measurement to another, e.g., the ratio of total protein or total lipid to total particles.[ citation needed ]

Single-particle analysis

Specialized methods are needed to study EVs at the single particle level. The challenge for any putative single-particle method is to identify the individual EV as a single, lipid-bilayer particle, and to provide additional information such as size, surface proteins, or nucleic acid content. Methods that have been used successfully for single-EV analysis include optical microscopy and flow cytometry (for large EVs, usually >200 nm), tunable resistive pulse sensing for evaluating EV size, concentration and zeta potential, as well as electron microscopy (no lower bound) and immuno electron microscopy, single-particle interferometric reflectance imaging (down to about 40 nm), and nano-flow cytometry (also to 40 nm). Some technologies allow the study of individual EVs without extensive prior separation from a biological matrix: to give a few examples, electron microscopy and flow cytometry.

Enriched and depleted markers

To demonstrate the presence of EVs in a preparation, as well as the relative depletion of non-EV particles or molecules, EV-enriched 'and' -depleted markers are necessary: [66] For example, the MISEV2018 guidelines recommend:

At least one membrane-associated marker as evidence of the lipid bilayer (e.g., a tetraspanin protein)
At least one cytoplasmic but ideally membrane-associated marker to show that the particle is not merely a membrane fragment
At least one "negative" or "depleted" marker: a "deep cellular" marker, a marker of a non-EV particle, or a soluble molecule not thought to be enriched in EVs. [47]

Usually, but not necessarily, the EV-enriched or -depleted markers are proteins that can be detected by Western blot, flow cytometry, ELISA, mass spectrometry, or other widely-available methods. Assaying for depleted markers is thought to be particularly important, as otherwise the purity of an EV preparation cannot be claimed. However, most studies of EVs prior to 2016 did not support claims of the presence of EVs by showing enriched markers, and <5% measured the presence of possible co-isolates/contaminants. [67] Despite the high need, a list of EV contaminants is not yet available to the EV research community. A recent study suggested density-gradient-based EV separation from biofluids as an experimental set-up to compile a list of contaminants for EV, based upon differential analysis of EV-enriched fractions versus soluble protein-enriched fractions. [68] Soluble proteins in blood, the Tamm-Horsfall protein (uromodulin) in urine, or proteins of the nucleus, Golgi apparatus, endoplasmic reticulum, or mitochondria in eukaryotic cells. The latter proteins may be found in large EVs or indeed any EVs, but are expected to be less concentrated in the EV than in the cell. [47]

Function

A wide variety of biological functions have been ascribed to EVs.[ citation needed ]

"Trash disposal": eliminating unwanted materials
Transfer of functional proteins
Transfer of functional DNA and RNA
Molecular recycling or "nutrition"
Signaling to the recipient cell via cell-surface or endosomal receptors
Creation of a metastatic niche for cancer
Pathfinding through the environment
Quorum sensing
Mediating host-commensal or parasite/pathogen interaction

Clinical significance

Aging

EVs have been implicated in senescence. Extracellular vesicle secretion is generally believed to increase with age due to DNA or mitochondrial damage and lipid peroxidation. [69] It has been demonstrated that exosomes released by senescent cells have a miRNA content that contributes to aging. [70] miRNAs play an essential role in senescence by negatively regulating the suppressors of p53, for example. [71]  

Furthermore, EVs play a role in overall chronic inflammation. The interorgan shuttling of EVs can mean that one disease is likely to promote the advancement of another, as is the case with NAFLD and the development of atherosclerosis. EVs released from steatosis-affected hepatocytes induce the release of inflammatory molecules from endothelial cells co-cultured with them. The co-cultured cells also show increased NF-κB activity. It has thus been demonstrated that EVs released by hepatocytes under NAFLD conditions cause vascular endothelial inflammation and promote atherosclerosis. [72]

EVs also have senolytic potential. EVs harvested from cardio-sphere-derived cells in young rats have been shown to reverse senescent processes in aged rats. The older rats’ endurance and cardiovascular function improved when they received a transfusion of EVs from younger animals. It is therefore believed that EVs hold promise as an anti-aging treatment in humans. [73]

Coagulation

Studies indicate that EVs may have a procoagulant effect in various diseases. [74] EVs can express phosphatidylserine (PS) on their surface. PS is an anionic phospholipid and PS+ EVs therefore provide a negatively charged surface which may facilitate formation of coagulation complexes. Under pathological conditions, EVs can sometimes express tissue factor (TF). TF is the most potent initiator of the coagulation cascade and is under normal conditions mainly contained to subvascular tissue.

Disease

EVs are believed to play a role in the spreading of different diseases. [75] [44] [76] Studies have shown that tumor cells send EVs to send signal to target resident cells, which can lead to tumor invasion and metastasis. [77] In vitro studies of Alzheimer's disease have shown that astrocytes that accumulate amyloid beta release EVs that cause neuronal apoptosis. [78] The content of the EVs was also affected by the exposure to amyloid beta and higher ApoE was found in EVs secreted by astrocyte exposed to amyloid beta. [79] An oncogenic mechanism illustrates how extracellular vesicles are produced by proliferative acute lymphoblastic leukemia cells and can target and compromise a healthy hematopoiesis system during leukemia development. [80]

T cell longevity

The fate of T cells can be determined by the transfer of telomeres via EVs from APCs. T cells that acquire telomeres in such a manner regain stem-like characteristics, avoiding senescence. The creation of long-lived memory T cells via an EV injection of telomeres enhances long-term immunological memory. [81]

As biomarkers

It has been suggested that EVs carrying nucleic acid cargo could serve as biomarkers for disease, especially in neurological disorders where it is difficult to assess the underlying pathology directly.

EVs facilitate communication between different parts of the CNS, [82] and therefore, EVs found in the blood of neurological patients contain molecules implicated in neurodegenerative diseases. [83] EVs carrying myeloid cargo, for example, have long been recognized as a biomarker of brain inflammation. [84] Furthermore, nucleic acids corresponding to APP, Aβ42, BACE1, and tau protein biomarkers were found to be associated with different neurodegenerative diseases. [85]

Using EVs to profile RNA expression patterns could therefore help diagnose certain diseases before a patient become symptomatic. Exosome Diagnostic (Cambridge, MA, USA), for example, has a patent for detecting neurodegenerative diseases and brain injury based on the measure of RNA-s (mRNA, miRNA, siRNA, or shRNA) associated with CSF-derived EVs. [86]

Related Research Articles

<span class="mw-page-title-main">Vesicle (biology and chemistry)</span> Any small, fluid-filled, spherical organelle enclosed by a membrane

In cell biology, a vesicle is a structure within or outside a cell, consisting of liquid or cytoplasm enclosed by a lipid bilayer. Vesicles form naturally during the processes of secretion (exocytosis), uptake (endocytosis), and the transport of materials within the plasma membrane. Alternatively, they may be prepared artificially, in which case they are called liposomes. If there is only one phospholipid bilayer, the vesicles are called unilamellar liposomes; otherwise they are called multilamellar liposomes. The membrane enclosing the vesicle is also a lamellar phase, similar to that of the plasma membrane, and intracellular vesicles can fuse with the plasma membrane to release their contents outside the cell. Vesicles can also fuse with other organelles within the cell. A vesicle released from the cell is known as an extracellular vesicle.

<span class="mw-page-title-main">Exosome (vesicle)</span> Membrane-bound extracellular vesicles

Exosomes, ranging in size from 30 to 150 nanometers, are membrane-bound extracellular vesicles (EVs) that are produced in the endosomal compartment of most eukaryotic cells. In multicellular organisms, exosomes and other EVs are found in biological fluids including saliva, blood, urine and cerebrospinal fluid. EVs have specialized functions in physiological processes, from coagulation and waste management to intercellular communication.

<span class="mw-page-title-main">Exosome complex</span> Protein complex that degrades RNA

The exosome complex is a multi-protein intracellular complex capable of degrading various types of RNA molecules. Exosome complexes are found in both eukaryotic cells and archaea, while in bacteria a simpler complex called the degradosome carries out similar functions.

<span class="mw-page-title-main">Adrenal tumor</span> Medical condition

An adrenal tumor or adrenal mass is any benign or malignant neoplasms of the adrenal gland, several of which are notable for their tendency to overproduce endocrine hormones. Adrenal cancer is the presence of malignant adrenal tumors, and includes neuroblastoma, adrenocortical carcinoma and some adrenal pheochromocytomas. Most adrenal pheochromocytomas and all adrenocortical adenomas are benign tumors, which do not metastasize or invade nearby tissues, but may cause significant health problems by unbalancing hormones.

<span class="mw-page-title-main">Microvesicle</span> Type of extracellular vesicle

Microvesicles are a type of extracellular vesicle (EV) that are released from the cell membrane. In multicellular organisms, microvesicles and other EVs are found both in tissues and in many types of body fluids. Delimited by a phospholipid bilayer, microvesicles can be as small as the smallest EVs or as large as 1000 nm. They are considered to be larger, on average, than intracellularly-generated EVs known as exosomes. Microvesicles play a role in intercellular communication and can transport molecules such as mRNA, miRNA, and proteins between cells.

<span class="mw-page-title-main">CD9</span> Human protein-encoding gene

CD9 is a gene encoding a protein that is a member of the transmembrane 4 superfamily also known as the tetraspanin family. It is a cell surface glycoprotein that consists of four transmembrane regions and has two extracellular loops that contain disulfide bonds which are conserved throughout the tetraspanin family. Also containing distinct palmitoylation sites that allows CD9 to interact with lipids and other proteins.

Prostasomes are extracellular vesicles secreted by the prostate gland epithelial cells into seminal fluid. They possess an unusual lipid composition and a tight and highly ordered structure of their lipid bilayer membrane, resembling that of lipid raft domains. Prostasomes appear to improve sperm motility and protect against attacks from the female immune defense during the passage to the egg.

ExoCarta is a manually curated database of exosomal proteins, RNA and lipids.

Jan Lötvall is a Swedish clinical allergist and scientist working on translational research primarily in the field of asthma. He is the former director of the Krefting Research Centre at the University of Gothenburg and is the Chief Scientific Officer of ExoCoBio.

Izon Science Limited is a nanotechnology company that develops and sells nano-scale particle analysis and isolation tools. Their main instruments are based on principles of size exclusion chromatography and tunable resistive pulse sensing. Izon’s size-exclusion chromatography columns and related solutions are also used by diagnostics companies focused on developing extracellular vesicle biomarkers.

Extracellular RNA (exRNA) describes RNA species present outside of the cells in which they were transcribed. Carried within extracellular vesicles, lipoproteins, and protein complexes, exRNAs are protected from ubiquitous RNA-degrading enzymes. exRNAs may be found in the environment or, in multicellular organisms, within the tissues or biological fluids such as venous blood, saliva, breast milk, urine, semen, menstrual blood, and vaginal fluid. Although their biological function is not fully understood, exRNAs have been proposed to play a role in a variety of biological processes including syntrophy, intercellular communication, and cell regulation. The United States National Institutes of Health (NIH) published in 2012 a set of Requests for Applications (RFAs) for investigating extracellular RNA biology. Funded by the NIH Common Fund, the resulting program was collectively known as the Extracellular RNA Communication Consortium (ERCC). The ERCC was renewed for a second phase in 2019.

<span class="mw-page-title-main">Tumor microenvironment</span> Surroundings of tumors including nearby cells and blood vessels

The tumor microenvironment is a complex ecosystem surrounding a tumor, composed of cancer cells, stromal tissue and the extracellular matrix. Mutual interaction between cancer cells and the different components of the tumor microenvironment support its growth and invasion in healthy tissues which correlates with tumor resistance to current treatments and poor prognosis. The tumor microenvironment is in constant change because of the tumor's ability to influence the microenvironment by releasing extracellular signals, promoting tumor angiogenesis and inducing peripheral immune tolerance, while the immune cells in the microenvironment can affect the growth and evolution of cancerous cells.

The secretome is the set of proteins expressed by an organism and secreted into the extracellular space. In humans, this subset of the proteome encompasses 13-20% of all proteins, including cytokines, growth factors, extracellular matrix proteins and regulators, and shed receptors. The secretome of a specific tissue can be measured by mass spectrometry and its analysis constitutes a type of proteomics known as secretomics.

The International Society for Extracellular Vesicles (ISEV) is an international scientific organization that focuses on the study of extracellular vesicles (EVs). These membrane-bound particles are released from all known cells and include exosomes, ectosomes, exophers, oncosomes, and more. Established in 2011, the society is a nonprofit organization. It is governed by an executive committee. The current president is Buzás Edit. Previous presidents were Clotilde Théry (2018-2022), Andrew Hill (2016-2018) and founding president Jan Lötvall (2011-2016). The society's journals are the Journal of Extracellular Vesicles and the Journal of Extracellular Biology. ISEV also publishes the international consensus guidelines for EV studies, the "Minimal information for studies of EVs" (MISEV).

The stem cell secretome is a collective term for the paracrine soluble factors produced by stem cells and utilized for their inter-cell communication. In addition to inter-cell communication, the paracrine factors are also responsible for tissue development, homeostasis and (re-)generation. The stem cell secretome consists of extracellular vesicles, specifically exosomes, microvesicles, membrane particles, peptides and small proteins (cytokines). The paracrine activity of stem cells, i.e. the stem cell secretome, has been found to be the predominant mechanism by which stem cell-based therapies mediate their effects in degenerative, auto-immune and/or inflammatory diseases. Though not only stem cells possess a secretome which influences their cellular environment, their secretome currently appears to be the most relevant for therapeutic use.

<i>Journal of Extracellular Vesicles</i> Academic journal

The Journal of Extracellular Vesicles, JEV, is a peer-reviewed open-access scientific journal of the International Society for Extracellular Vesicles (ISEV). As one of two official journals of ISEV, the other being the Journal of Extracellular Biology, JEV covers research on lipid bilayer-delimited particles known as extracellular vesicles (EVs). EVs are released from cells and include endosome-origin exosomes and plasma membrane-derived ectosomes/microvesicles. The journal was established in 2012 and is currently published by Wiley. The founding editors-in-chief were Clotilde Théry, Yong Song Gho, and Peter Quesenberry. The current editor-in-chief is Jan Lötvall.

<span class="mw-page-title-main">Kenneth Witwer</span> American biologist

Kenneth W. Witwer is an associate professor of molecular and comparative pathobiology and neurology at the Johns Hopkins University School of Medicine in Baltimore, Maryland, United States. As nominated President-Elect of the International Society for Extracellular Vesicles (ISEV), Witwer previously served as Secretary General and Executive Chair of Science and Meetings of the society. His laboratory studies extracellular vesicles (EVs), noncoding and extracellular RNA (exRNA), and enveloped viruses, including HIV and SARS-CoV-2. Witwer is the managing editor of the journal Cytotherapy and a member of the Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease. He has advised the US Environmental Protection Agency and the US National Institutes of Health and is an associate editor of the Journal of Extracellular Vesicles.

<span class="mw-page-title-main">Exopher</span>

Exophers are a type of membrane-bound extracellular vesicle (EV) that are released by budding out of cells into the extracellular space. Exophers can be released by neurons and muscle in the nematode Caenorhabditis elegans and also from murine cardiomyocytes. Exophers are notable for their large size, averaging approximately four microns in diameter, and they are able to expel whole organelles, such as mitochondria and lysosomes as cargo. An exopher can initially remain attached to the cell that produced it by a membranous filament that resembles a tunneling nanotube. Exophers share similarities with large oncosomes, but they differ in that they are produced by physiologically normal cells instead of aberrant cells associated with tumors.

<span class="mw-page-title-main">Intracellular delivery</span> Scientific research area

Intracellular delivery is the process of introducing external materials into living cells. Materials that are delivered into cells include nucleic acids, proteins, peptides, impermeable small molecules, synthetic nanomaterials, organelles, and micron-scale tracers, devices and objects. Such molecules and materials can be used to investigate cellular behavior, engineer cell operations or correct a pathological function.

Exosomes are small vesicles secreted by cells that play a crucial role in intercellular communication. They contain a variety of biomolecules, including proteins, nucleic acids and lipids, which can be transferred between cells to modulate cellular processes. Exosomes have been increasingly acknowledged as promising therapeutic tool and delivery platforms due to unique biological properties.

  1. Biocompatibility: Exosomes are naturally occurring particles in body, which makes them highly biocompatible and less likely to activate immune response.
  2. Targeting ability: Exosomes are assembled to express specific proteins or peptides, allowing them to target specific cells or tissues.
  3. Natural cargo carries: Exosomes can naturally transport a variety of biomolecules, including proteins, RNA and DNA, which can be used for therapeutic purposes.

References

  1. 1 2 Soleymani T, Chen TY, Gonzalez-Kozlova E, Dogra N (2023). "The human neurosecretome: extracellular vesicles and particles (EVPs) of the brain for intercellular communication, therapy, and liquid-biopsy applications". Frontiers in Molecular Biosciences. 10: 1156821. doi: 10.3389/fmolb.2023.1156821 . PMC   10229797 . PMID   37266331.
  2. Veziroglu EM, Mias GI (2020-07-17). "Characterizing Extracellular Vesicles and Their Diverse RNA Contents". Frontiers in Genetics. 11: 700. doi: 10.3389/fgene.2020.00700 . PMC   7379748 . PMID   32765582.
  3. Moghassemi, Saeid; Dadashzadeh, Arezoo; Sousa, Maria João; Vlieghe, Hanne; Yang, Jie; León-Félix, Cecibel María; Amorim, Christiani A. (June 2024). "Extracellular vesicles in nanomedicine and regenerative medicine: A review over the last decade". Bioactive Materials. 36: 126–156. doi:10.1016/j.bioactmat.2024.02.021. PMC   10915394 .
  4. Subedi P, Schneider M, Philipp J, Azimzadeh O, Metzger F, Moertl S, et al. (November 2019). "Comparison of methods to isolate proteins from extracellular vesicles for mass spectrometry-based proteomic analyses". Analytical Biochemistry. 584: 113390. doi: 10.1016/j.ab.2019.113390 . PMID   31401005.
  5. Clerici SP, Peppelenbosch M, Fuhler G, Consonni SR, Ferreira-Halder CV (2021-07-15). "Colorectal Cancer Cell-Derived Small Extracellular Vesicles Educate Human Fibroblasts to Stimulate Migratory Capacity". Frontiers in Cell and Developmental Biology. 9: 696373700. doi: 10.3389/fcell.2021.696373 . PMC   8320664 . PMID   34336845.
  6. 1 2 3 4 5 6 7 8 Yáñez-Mó M, Siljander PR, Andreu Z, Zavec AB, Borràs FE, Buzas EI, et al. (2015). "Biological properties of extracellular vesicles and their physiological functions". Journal of Extracellular Vesicles. 4: 27066. doi:10.3402/jev.v4.27066. PMC   4433489 . PMID   25979354.
  7. Chargaff E, West R (November 1946). "The biological significance of the thromboplastic protein of blood". The Journal of Biological Chemistry. 166 (1): 189–97. doi: 10.1016/S0021-9258(17)34997-9 . PMID   20273687.
  8. Wolf P (May 1967). "The nature and significance of platelet products in human plasma". British Journal of Haematology. 13 (3): 269–88. doi:10.1111/j.1365-2141.1967.tb08741.x. PMID   6025241. S2CID   19215210.
  9. Anderson HC (April 1969). "Vesicles associated with calcification in the matrix of epiphyseal cartilage". The Journal of Cell Biology. 41 (1): 59–72. doi:10.1083/jcb.41.1.59. PMC   2107736 . PMID   5775794.
  10. 1 2 Bonucci E (1970). "Fine structure and histochemistry of "calcifying globules" in epiphyseal cartilage". Zeitschrift für Zellforschung und Mikroskopische Anatomie. 103 (2): 192–217. doi:10.1007/BF00337312. PMID   5412827. S2CID   8633696.
  11. Nunez EA, Wallis J, Gershon MD (October 1974). "Secretory processes in follicular cells of the bat thyroid. 3. The occurrence of extracellular vesicles and colloid droplets during arousal from hibernation". The American Journal of Anatomy. 141 (2): 179–201. doi:10.1002/aja.1001410203. PMID   4415703.
  12. Chandler RL, Bird RG, Bland AP (November 1975). "Letter: Particles associated with microvillous border of intestinal mucosa". Lancet. 2 (7941): 931–2. doi:10.1016/s0140-6736(75)92175-3. PMID   53415. S2CID   40320534.
  13. De Broe M, Wieme R, Roels F (December 1975). "Letter: Membrane fragments with koinozymic properties released from villous adenoma of the rectum". Lancet. 2 (7946): 1214–5. doi:10.1016/s0140-6736(75)92709-9. PMID   53703. S2CID   32026872.
  14. 1 2 Benz EW, Moses HL (June 1974). "Small, virus-like particles detected in bovine sera by electron microscopy". Journal of the National Cancer Institute. 52 (6): 1931–4. doi:10.1093/jnci/52.6.1931. PMID   4834422.
  15. 1 2 3 Dalton AJ (May 1975). "Microvesicles and vesicles of multivesicular bodies versus "virus-like" particles". Journal of the National Cancer Institute. 54 (5): 1137–48. doi:10.1093/jnci/54.5.1137. PMID   165305.
  16. Yim K, Borgoni S, Chahwan R (April 2022). "Letter: Serum extracellular vesicles profiling is associated with COVID-19 progression and immune responses". J Extracell Biol. 1 (4): e37. doi:10.1002/jex2.37. PMC   9088353 . PMID   35574251.
  17. Pan BT, Johnstone RM (July 1983). "Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor". Cell. 33 (3): 967–78. doi:10.1016/0092-8674(83)90040-5. PMID   6307529. S2CID   33216388.
  18. Harding C, Heuser J, Stahl P (November 1984). "Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: demonstration of a pathway for receptor shedding". European Journal of Cell Biology. 35 (2): 256–63. PMID   6151502.
  19. Johnstone RM, Adam M, Hammond JR, Orr L, Turbide C (July 1987). "Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes)". The Journal of Biological Chemistry. 262 (19): 9412–20. doi: 10.1016/S0021-9258(18)48095-7 . PMID   3597417.
  20. Dvorak HF, Quay SC, Orenstein NS, Dvorak AM, Hahn P, Bitzer AM, Carvalho AC (May 1981). "Tumor shedding and coagulation". Science. 212 (4497): 923–4. Bibcode:1981Sci...212..923D. doi:10.1126/science.7195067. PMID   7195067.
  21. Fox AS, Yoon SB (November 1970). "DNA-induced transformation in Drosophila: locus-specificity and the establishment of transformed stocks". Proceedings of the National Academy of Sciences of the United States of America. 67 (3): 1608–15. Bibcode:1970PNAS...67.1608F. doi: 10.1073/pnas.67.3.1608 . PMC   283397 . PMID   5274483.
  22. Mishra NC, Tatum EL (December 1973). "Non-Mendelian inheritance of DNA-induced inositol independence in Neurospora". Proceedings of the National Academy of Sciences of the United States of America. 70 (12): 3875–9. Bibcode:1973PNAS...70.3875M. doi: 10.1073/pnas.70.12.3875 . PMC   427348 . PMID   4521213.
  23. Stegmayr B, Ronquist G (1982). "Promotive effect on human sperm progressive motility by prostasomes". Urological Research. 10 (5): 253–7. doi:10.1007/bf00255932. PMID   6219486. S2CID   26574697.
  24. Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV, Melief CJ, Geuze HJ (March 1996). "B lymphocytes secrete antigen-presenting vesicles". The Journal of Experimental Medicine. 183 (3): 1161–72. doi:10.1084/jem.183.3.1161. PMC   2192324 . PMID   8642258.
  25. Mack M, Kleinschmidt A, Brühl H, Klier C, Nelson PJ, Cihak J, et al. (July 2000). "Transfer of the chemokine receptor CCR5 between cells by membrane-derived microparticles: a mechanism for cellular human immunodeficiency virus 1 infection". Nature Medicine. 6 (7): 769–75. doi:10.1038/77498. PMID   10888925. S2CID   23027144.
  26. Rozmyslowicz T, Majka M, Kijowski J, Murphy SL, Conover DO, Poncz M, et al. (January 2003). "Platelet- and megakaryocyte-derived microparticles transfer CXCR4 receptor to CXCR4-null cells and make them susceptible to infection by X4-HIV". AIDS. 17 (1): 33–42. doi: 10.1097/00002030-200301030-00006 . PMID   12478067. S2CID   6619801.
  27. Baj-Krzyworzeka M, Szatanek R, Weglarczyk K, Baran J, Urbanowicz B, Brański P, et al. (July 2006). "Tumour-derived microvesicles carry several surface determinants and mRNA of tumour cells and transfer some of these determinants to monocytes". Cancer Immunology, Immunotherapy. 55 (7): 808–18. doi:10.1007/s00262-005-0075-9. PMID   16283305. S2CID   25723677.
  28. Ratajczak J, Wysoczynski M, Hayek F, Janowska-Wieczorek A, Ratajczak MZ (September 2006). "Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication". Leukemia. 20 (9): 1487–95. doi:10.1038/sj.leu.2404296. PMID   16791265. S2CID   6874345.
  29. Aliotta JM, Sanchez-Guijo FM, Dooner GJ, Johnson KW, Dooner MS, Greer KA, et al. (September 2007). "Alteration of marrow cell gene expression, protein production, and engraftment into lung by lung-derived microvesicles: a novel mechanism for phenotype modulation". Stem Cells. 25 (9): 2245–56. doi:10.1634/stemcells.2007-0128. PMC   3376082 . PMID   17556595.
  30. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO (June 2007). "Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells". Nature Cell Biology. 9 (6): 654–9. doi:10.1038/ncb1596. PMID   17486113. S2CID   8599814.
  31. Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. (December 2008). "Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers". Nature Cell Biology. 10 (12): 1470–6. doi:10.1038/ncb1800. PMC   3423894 . PMID   19011622.
  32. Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van Eijndhoven MA, Hopmans ES, Lindenberg JL, et al. (April 2010). "Functional delivery of viral miRNAs via exosomes". Proceedings of the National Academy of Sciences of the United States of America. 107 (14): 6328–33. Bibcode:2010PNAS..107.6328P. doi: 10.1073/pnas.0914843107 . PMC   2851954 . PMID   20304794.
  33. 1 2 Chetty VK, Ghanam J, Anchan S, Reinhardt K, Brenzel A, Gelléri M, Cremer C, Grueso-Navarro E, Schneider M, von Neuhoff N, Reinhardt D, Jablonska J, Nazarenko I, Thakur BK (April 2022). "Efficient Small Extracellular Vesicles (EV) Isolation Method and Evaluation of EV-Associated DNA Role in Cell-Cell Communication in Cancer". Cancers (Basel). 14 (9): 2068. doi: 10.3390/cancers14092068 . PMC   9099953 . PMID   35565197.
  34. Al-Nedawi K, Meehan B, Rak J (July 2009). "Microvesicles: messengers and mediators of tumor progression". Cell Cycle. 8 (13): 2014–8. doi: 10.4161/cc.8.13.8988 . PMID   19535896.
  35. Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, et al. (November 2015). "Tumour exosome integrins determine organotropic metastasis". Nature. 527 (7578): 329–35. Bibcode:2015Natur.527..329H. doi:10.1038/nature15756. PMC   4788391 . PMID   26524530.
  36. Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. (June 2012). "Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET". Nature Medicine. 18 (6): 883–91. doi:10.1038/nm.2753. PMC   3645291 . PMID   22635005.
  37. Melo SA, Sugimoto H, O'Connell JT, Kato N, Villanueva A, Vidal A, et al. (November 2014). "Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis". Cancer Cell. 26 (5): 707–21. doi:10.1016/j.ccell.2014.09.005. PMC   4254633 . PMID   25446899.
  38. Kamerkar S, LeBleu VS, Sugimoto H, Yang S, Ruivo CF, Melo SA, et al. (June 2017). "Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer". Nature. 546 (7659): 498–503. Bibcode:2017Natur.546..498K. doi:10.1038/nature22341. PMC   5538883 . PMID   28607485.
  39. Ostrowski M, Carmo NB, Krumeich S, Fanget I, Raposo G, Savina A, et al. (January 2010). "Rab27a and Rab27b control different steps of the exosome secretion pathway". Nature Cell Biology. 12 (1): 19–30, sup pp 1–13. doi:10.1038/ncb2000. hdl: 10044/1/19574 . PMID   19966785. S2CID   13935708.
  40. van Niel G, Porto-Carreiro I, Simoes S, Raposo G (July 2006). "Exosomes: a common pathway for a specialized function". Journal of Biochemistry. 140 (1): 13–21. doi: 10.1093/jb/mvj128 . PMID   16877764. S2CID   43541754.
  41. Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, et al. (February 2016). "Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes". Proceedings of the National Academy of Sciences of the United States of America. 113 (8): E968-77. Bibcode:2016PNAS..113E.968K. doi: 10.1073/pnas.1521230113 . PMC   4776515 . PMID   26858453.
  42. Tkach M, Kowal J, Théry C (January 2018). "Why the need and how to approach the functional diversity of extracellular vesicles". Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences. 373 (1737): 20160479. doi:10.1098/rstb.2016.0479. PMC   5717434 . PMID   29158309.
  43. Leslie M (August 2013). "Cell Biology. NIH effort gambles on mysterious extracellular RNAs". Science. 341 (6149): 947. doi:10.1126/science.341.6149.947. PMID   23990535.
  44. 1 2 Gutierrez BC, Ancarola ME, Volpato-Rossi I, Marcilla A, Ramirez MI, Rosenzvit MC, et al. (2022). "Extracellular vesicles from Trypanosoma cruzi-dendritic cell interaction show modulatory properties and confer resistance to lethal infection as a cell-free based therapy strategy". Frontiers in Cellular and Infection Microbiology. 12: 980817. doi: 10.3389/fcimb.2022.980817 . PMC   9710384 . PMID   36467728.
  45. Bazzan E, Tinè M, Casara A, Biondini D, Semenzato U, Cocconcelli E, et al. (June 2021). "Critical Review of the Evolution of Extracellular Vesicles' Knowledge: From 1946 to Today". International Journal of Molecular Sciences. 22 (12): 6417. doi: 10.3390/ijms22126417 . PMC   8232679 . PMID   34203956.
  46. Gurunathan S, Kang MH, Jeyaraj M, Qasim M, Kim JH (April 2019). "Review of the Isolation, Characterization, Biological Function, and Multifarious Therapeutic Approaches of Exosomes". Cells. 8 (4): 307. doi: 10.3390/cells8040307 . PMC   6523673 . PMID   30987213.
  47. 1 2 3 4 5 6 7 Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. (2018). "Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines". Journal of Extracellular Vesicles. 7 (1): 1535750. doi:10.1080/20013078.2018.1535750. PMC   6322352 . PMID   30637094.
  48. Trams EG, Lauter CJ, Salem N, Heine U (July 1981). "Exfoliation of membrane ecto-enzymes in the form of micro-vesicles". Biochimica et Biophysica Acta (BBA) - Biomembranes. 645 (1): 63–70. doi:10.1016/0005-2736(81)90512-5. PMID   6266476.
  49. Morello M, Minciacchi VR, de Candia P, Yang J, Posadas E, Kim H, et al. (November 2013). "Large oncosomes mediate intercellular transfer of functional microRNA". Cell Cycle. 12 (22): 3526–36. doi:10.4161/cc.26539. PMC   3906338 . PMID   24091630.
  50. Meehan B, Rak J, Di Vizio D (2016). "Oncosomes - large and small: what are they, where they came from?". Journal of Extracellular Vesicles. 5: 33109. doi:10.3402/jev.v5.33109. PMC   5040817 . PMID   27680302.
  51. Minciacchi VR, Spinelli C, Reis-Sobreiro M, Cavallini L, You S, Zandian M, Li X, Mishra R, Chiarugi P, Adam RM, Posadas EM, Viglietto G, Freeman MR, Cocucci E, Bhowmick NA, Di Vizio D (2017). "MYC Mediates Large Oncosome-Induced Fibroblast Reprogramming in Prostate Cancer". Cancer Research. 77 (9): 2306–2317. doi: 10.1158/0008-5472.CAN-16-2942 . PMID   28202510.
  52. Bertolini I, Terrasi A, Martelli C, Gaudioso G, Di Cristofori A, Storaci AM, Formica M, Braidotti P, Todoerti K, Ferrero S, Caroli M, Ottobrini L, Vaccari T, Vaira V (2019). "A GBM-like V-ATPase signature directs cell-cell tumor signaling and reprogramming via large oncosomes". eBioMedicine. 41: 225–235. doi:10.1016/j.ebiom.2019.01.051. PMC   6441844 . PMID   30737083.
  53. 1 2 3 Melentijevic I, Toth ML, Arnold ML, Guasp RJ, Harinath G, Nguyen KC, et al. (February 2017). "C. elegans neurons jettison protein aggregates and mitochondria under neurotoxic stress". Nature. 542 (7641): 367–371. Bibcode:2017Natur.542..367M. doi:10.1038/nature21362. PMC   5336134 . PMID   28178240.
  54. Nicolás-Ávila JA, Lechuga-Vieco AV, Esteban-Martínez L, Sánchez-Díaz M, Díaz-García E, Santiago DJ, et al. (2020). "A Network of Macrophages Supports Mitochondrial Homeostasis in the Heart". Cell. 183 (1): 94–109. doi: 10.1016/j.cell.2020.08.031 . hdl: 10261/226682 . PMID   32937105. S2CID   221716195.
  55. Ma L, Li Y, Peng J, Wu D, Zhao X, Cui Y, Chen L, Yan X, Du Y, Yu L (2015). "Discovery of the migrasome, an organelle mediating release of cytoplasmic contents during cell migration". Cell Research. 25 (1): 24–38. doi:10.1038/cr.2014.135. PMC   4650581 . PMID   25342562.
  56. Jiao H, Jiang D, Hu X, Du W, Ji L, Yang Y, Li X, Sho T, Wang X, Li Y, Wu YT, Wei YH, Hu X, Yu L (2021). "Mitocytosis, a migrasome-mediated mitochondrial quality-control process". Cell. 184 (11): 2896–2910. doi: 10.1016/j.cell.2021.04.027 . PMID   34048705. S2CID   235226529.
  57. Nolte-'t Hoen E, Cremer T, Gallo RC, Margolis LB (August 2016). "Extracellular vesicles and viruses: Are they close relatives?". Proceedings of the National Academy of Sciences of the United States of America. 113 (33): 9155–61. Bibcode:2016PNAS..113.9155N. doi: 10.1073/pnas.1605146113 . PMC   4995926 . PMID   27432966.
  58. Mateescu B, Kowal EJ, van Balkom BW, Bartel S, Bhattacharyya SN, Buzás EI, et al. (2017). "Obstacles and opportunities in the functional analysis of extracellular vesicle RNA - an ISEV position paper". Journal of Extracellular Vesicles. 6 (1): 1286095. doi:10.1080/20013078.2017.1286095. PMC   5345583 . PMID   28326170.
  59. Multia E, Tear CJ, Palviainen M, Siljander P, Riekkola ML (December 2019). "Fast isolation of highly specific population of platelet-derived extracellular vesicles from blood plasma by affinity monolithic column, immobilized with anti-human CD61 antibody". Analytica Chimica Acta. 1091: 160–168. Bibcode:2019AcAC.1091..160M. doi:10.1016/j.aca.2019.09.022. hdl: 10138/321264 . PMID   31679569. S2CID   203147714.
  60. Multia E, Liangsupree T, Jussila M, Ruiz-Jimenez J, Kemell M, Riekkola ML (October 2020). "Automated On-Line Isolation and Fractionation System for Nanosized Biomacromolecules from Human Plasma". Analytical Chemistry. 92 (19): 13058–13065. doi:10.1021/acs.analchem.0c01986. PMC   7586295 . PMID   32893620.
  61. Morani M, Mai TD, Krupova Z, Defrenaix P, Multia E, Riekkola ML, Taverna M (September 2020). "Electrokinetic characterization of extracellular vesicles with capillary electrophoresis: A new tool for their identification and quantification". Analytica Chimica Acta. 1128: 42–51. Bibcode:2020AcAC.1128...42M. doi:10.1016/j.aca.2020.06.073. hdl: 10138/332354 . PMID   32825911. S2CID   221238347.
  62. Lacroix R, Judicone C, Poncelet P, Robert S, Arnaud L, Sampol J, Dignat-George F (March 2012). "Impact of pre-analytical parameters on the measurement of circulating microparticles: towards standardization of protocol". Journal of Thrombosis and Haemostasis. 10 (3): 437–46. doi: 10.1111/j.1538-7836.2011.04610.x . PMID   22212198. S2CID   46519893.
  63. Witwer KW, Buzás EI, Bemis LT, Bora A, Lässer C, Lötvall J, et al. (2013). "Standardization of sample collection, isolation and analysis methods in extracellular vesicle research". Journal of Extracellular Vesicles. 2: 20360. doi:10.3402/jev.v2i0.20360. PMC   3760646 . PMID   24009894.
  64. Coumans FA, Brisson AR, Buzas EI, Dignat-George F, Drees EE, El-Andaloussi S, et al. (May 2017). "Methodological Guidelines to Study Extracellular Vesicles". Circulation Research. 120 (10): 1632–1648. doi: 10.1161/CIRCRESAHA.117.309417 . PMID   28495994.
  65. Liangsupree T, Multia E, Riekkola ML (January 2021). "Modern isolation and separation techniques for extracellular vesicles". Journal of Chromatography A. 1636: 461773. doi: 10.1016/j.chroma.2020.461773 . ISSN   0021-9673. PMID   33316564.
  66. Lötvall J, Hill AF, Hochberg F, Buzás EI, Di Vizio D, Gardiner C, et al. (2014). "Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles". Journal of Extracellular Vesicles. 3: 26913. doi:10.3402/jev.v3.26913. PMC   4275645 . PMID   25536934.
  67. Van Deun J, Mestdagh P, Agostinis P, Akay Ö, Anand S, Anckaert J, et al. (February 2017). "EV-TRACK: transparent reporting and centralizing knowledge in extracellular vesicle research". Nature Methods. 14 (3): 228–232. doi:10.1038/nmeth.4185. PMID   28245209. S2CID   205425936.
  68. Dhondt B, Geeurickx E, Tulkens J, Van Deun J, Vergauwen G, Lippens L, et al. (11 March 2020). "Unravelling the proteomic landscape of extracellular vesicles in prostate cancer by density-based fractionation of urine". Journal of Extracellular Vesicles. 9 (1): 1736935. doi: 10.1080/20013078.2020.1736935 . PMC   7144211 . PMID   32284825.
  69. Yin Y, Chen H, Wang Y, Zhang L, Wang X (October 2021). "Roles of extracellular vesicles in the aging microenvironment and age-related diseases". Journal of Extracellular Vesicles. 10 (12): e12154. doi:10.1002/jev2.12154. PMC   8491204 . PMID   34609061.
  70. Xu D, Tahara H (March 2013). "The role of exosomes and microRNAs in senescence and aging". Advanced Drug Delivery Reviews. 65 (3): 368–375. doi:10.1016/j.addr.2012.07.010. PMID   22820533.
  71. Suh N (October 2018). "MicroRNA controls of cellular senescence". BMB Reports. 51 (10): 493–499. doi:10.5483/BMBRep.2018.51.10.209. PMC   6235093 . PMID   30269742.
  72. Jiang F, Chen Q, Wang W, Ling Y, Yan Y, Xia P (January 2020). "Hepatocyte-derived extracellular vesicles promote endothelial inflammation and atherogenesis via microRNA-1". Journal of Hepatology. 72 (1): 156–166. doi:10.1016/j.jhep.2019.09.014. PMID   31568800. S2CID   203622470.
  73. Grigorian Shamagian L, Rogers RG, Luther K, Angert D, Echavez A, Liu W, et al. (July 2023). "Rejuvenating effects of young extracellular vesicles in aged rats and in cellular models of human senescence". Scientific Reports. 13 (1): 12240. Bibcode:2023NatSR..1312240G. doi:10.1038/s41598-023-39370-5. PMC   10382547 . PMID   37507448.
  74. Owens, A. Phillip; Mackman, Nigel (2011-05-13). Weber, Christian; Mause, Sebastian (eds.). "Microparticles in Hemostasis and Thrombosis". Circulation Research. 108 (10): 1284–1297. doi:10.1161/CIRCRESAHA.110.233056. ISSN   0009-7330. PMC   3144708 .
  75. Yamamoto S, Azuma E, Muramatsu M, Hamashima T, Ishii Y, Sasahara M (November 2016). "Significance of Extracellular Vesicles: Pathobiological Roles in Disease". Cell Structure and Function. 41 (2): 137–143. doi: 10.1247/csf.16014 . PMID   27679938.
  76. Yim K, AlHrout A, Borgoni S, Chahwan R (December 2020). "Extracellular Vesicles Orchestrate Immune and Tumor Interaction Networks". Cancers. 12 (12): 3696. doi: 10.3390/cancers12123696 . PMC   7763968 . PMID   33317058.
  77. Cappariello A, Rucci N (September 2019). "Tumour-Derived Extracellular Vesicles (EVs): A Dangerous "Message in A Bottle" for Bone". International Journal of Molecular Sciences. 20 (19): 4805. doi: 10.3390/ijms20194805 . PMC   6802008 . PMID   31569680.
  78. Söllvander S, Nikitidou E, Brolin R, Söderberg L, Sehlin D, Lannfelt L, Erlandsson A (May 2016). "Accumulation of amyloid-β by astrocytes result in enlarged endosomes and microvesicle-induced apoptosis of neurons". Molecular Neurodegeneration. 11 (1): 38. doi: 10.1186/s13024-016-0098-z . PMC   4865996 . PMID   27176225.
  79. Nikitidou E, Khoonsari PE, Shevchenko G, Ingelsson M, Kultima K, Erlandsson A (2017). "Increased Release of Apolipoprotein E in Extracellular Vesicles Following Amyloid-β Protofibril Exposure of Neuroglial Co-Cultures". Journal of Alzheimer's Disease. 60 (1): 305–321. doi:10.3233/JAD-170278. PMC   5676865 . PMID   28826183.
  80. Georgievski A, Michel A, Thomas C, Mlamla Z, Pais de Barros JP, Lemaire-Ewing S, et al. (2022). "Acute lymphoblastic leukemia-derived extracellular vesicles affect quiescence of hematopoietic stem and progenitor cells". Cell Death Dis. 12 (4): 337. doi:10.1038/s41419-022-04761-5. PMC   9005650 . PMID   35414137.
  81. Lanna A, Vaz B, D'Ambra C, Valvo S, Vuotto C, Chiurchiù V, et al. (October 2022). "An intercellular transfer of telomeres rescues T cells from senescence and promotes long-term immunological memory". Nature Cell Biology. 24 (10): 1461–1474. doi:10.1038/s41556-022-00991-z. PMC   7613731 . PMID   36109671.
  82. Agnati LF, Guidolin D, Guescini M, Genedani S, Fuxe K (September 2010). "Understanding wiring and volume transmission". Brain Research Reviews. 64 (1): 137–159. doi:10.1016/j.brainresrev.2010.03.003. PMID   20347870. S2CID   36665895.
  83. Koniusz S, Andrzejewska A, Muraca M, Srivastava AK, Janowski M, Lukomska B (2016). "Extracellular Vesicles in Physiology, Pathology, and Therapy of the Immune and Central Nervous System, with Focus on Extracellular Vesicles Derived from Mesenchymal Stem Cells as Therapeutic Tools". Frontiers in Cellular Neuroscience. 10: 109. doi: 10.3389/fncel.2016.00109 . PMC   4852177 . PMID   27199663.
  84. Verderio C, Muzio L, Turola E, Bergami A, Novellino L, Ruffini F, et al. (October 2012). "Myeloid microvesicles are a marker and therapeutic target for neuroinflammation". Annals of Neurology. 72 (4): 610–624. doi:10.1002/ana.23627. PMID   23109155. S2CID   35702508.
  85. Urbanelli L, Buratta S, Sagini K, Ferrara G, Lanni M, Emiliani C (2015). "Exosome-based strategies for Diagnosis and Therapy". Recent Patents on CNS Drug Discovery. 10 (1): 10–27. doi:10.2174/1574889810666150702124059. PMID   26133463.
  86. Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. (December 2008). "Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers". Nature Cell Biology. 10 (12): 1470–1476. doi:10.1038/ncb1800. PMC   3423894 . PMID   19011622.