Biology:Extracellular vesicle

From HandWiki
Short description: Lipid bilayer-delimited particles

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] They carry a cargo of proteins,[3] nucleic acids, lipids, metabolites, and even organelles from the parent cell. EVs carry distinct proteo-transcriptomic signatures that are different from their cancer cell of origin[clarification needed].[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.

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

Main page: Biology:Exosome (vesicle)

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

Main page: Biology:Exopher

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 signficance

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]

Disease

EVs are believed to play a role in the spreading of different diseases.[74][44][75] Studies have shown that tumor cells send EVs to send signal to target resident cells, which can lead to tumor invasion and metastasis.[76] In vitro studies of Alzheimer's disease have shown that astrocytes that accumulate amyloid beta release EVs that cause neuronal apoptosis.[77] 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.[78] 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.[79]

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.[80]

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

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.[85]

References

  1. 1.0 1.1 "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. 2023. doi:10.3389/fmolb.2023.1156821. PMID 37266331. 
  2. "Characterizing Extracellular Vesicles and Their Diverse RNA Contents". Frontiers in Genetics 11: 700. 2020-07-17. doi:10.3389/fgene.2020.00700. PMID 32765582. 
  3. "Comparison of methods to isolate proteins from extracellular vesicles for mass spectrometry-based proteomic analyses". Analytical Biochemistry 584: 113390. November 2019. doi:10.1016/j.ab.2019.113390. PMID 31401005. 
  4. "Extracellular vesicles carry distinct proteo-transcriptomic signatures that are different from their cancer cell of origin". iScience 25 (6): 104414. June 2022. doi:10.1016/j.isci.2022.104414. PMID 35663013. Bibcode2022iSci...25j4414C. 
  5. "Colorectal Cancer Cell-Derived Small Extracellular Vesicles Educate Human Fibroblasts to Stimulate Migratory Capacity.". Frontiers in Cell and Developmental Biology 9: 696373700. 2021-07-15. doi:10.3389/fcell.2021.696373. PMID 34336845. 
  6. 6.0 6.1 6.2 6.3 6.4 6.5 6.6 6.7 "Biological properties of extracellular vesicles and their physiological functions". Journal of Extracellular Vesicles 4: 27066. 2015. doi:10.3402/jev.v4.27066. PMID 25979354. 
  7. "The biological significance of the thromboplastic protein of blood". The Journal of Biological Chemistry 166 (1): 189–97. November 1946. doi:10.1016/S0021-9258(17)34997-9. PMID 20273687. 
  8. "The nature and significance of platelet products in human plasma". British Journal of Haematology 13 (3): 269–88. May 1967. doi:10.1111/j.1365-2141.1967.tb08741.x. PMID 6025241. 
  9. "Vesicles associated with calcification in the matrix of epiphyseal cartilage". The Journal of Cell Biology 41 (1): 59–72. April 1969. doi:10.1083/jcb.41.1.59. PMID 5775794. 
  10. 10.0 10.1 "Fine structure and histochemistry of "calcifying globules" in epiphyseal cartilage". Zeitschrift für Zellforschung und Mikroskopische Anatomie 103 (2): 192–217. 1970. doi:10.1007/BF00337312. PMID 5412827. 
  11. "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. October 1974. doi:10.1002/aja.1001410203. PMID 4415703. 
  12. "Letter: Particles associated with microvillous border of intestinal mucosa". Lancet 2 (7941): 931–2. November 1975. doi:10.1016/s0140-6736(75)92175-3. PMID 53415. 
  13. "Letter: Membrane fragments with koinozymic properties released from villous adenoma of the rectum". Lancet 2 (7946): 1214–5. December 1975. doi:10.1016/s0140-6736(75)92709-9. PMID 53703. 
  14. 14.0 14.1 "Small, virus-like particles detected in bovine sera by electron microscopy". Journal of the National Cancer Institute 52 (6): 1931–4. June 1974. doi:10.1093/jnci/52.6.1931. PMID 4834422. 
  15. 15.0 15.1 15.2 "Microvesicles and vesicles of multivesicular bodies versus "virus-like" particles". Journal of the National Cancer Institute 54 (5): 1137–48. May 1975. doi:10.1093/jnci/54.5.1137. PMID 165305. 
  16. "Letter: Serum extracellular vesicles profiling is associated with COVID-19 progression and immune responses". J Extracell Biol 1 (4): e37. April 2022. doi:10.1002/jex2.37. PMID 35574251. 
  17. "Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor". Cell 33 (3): 967–78. July 1983. doi:10.1016/0092-8674(83)90040-5. PMID 6307529. 
  18. "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. November 1984. PMID 6151502. 
  19. "Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes)". The Journal of Biological Chemistry 262 (19): 9412–20. July 1987. doi:10.1016/S0021-9258(18)48095-7. PMID 3597417. 
  20. "Tumor shedding and coagulation". Science 212 (4497): 923–4. May 1981. doi:10.1126/science.7195067. PMID 7195067. Bibcode1981Sci...212..923D. 
  21. "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. November 1970. doi:10.1073/pnas.67.3.1608. PMID 5274483. Bibcode1970PNAS...67.1608F. 
  22. "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. December 1973. doi:10.1073/pnas.70.12.3875. PMID 4521213. Bibcode1973PNAS...70.3875M. 
  23. "Promotive effect on human sperm progressive motility by prostasomes". Urological Research 10 (5): 253–7. 1982. doi:10.1007/bf00255932. PMID 6219486. 
  24. "B lymphocytes secrete antigen-presenting vesicles". The Journal of Experimental Medicine 183 (3): 1161–72. March 1996. doi:10.1084/jem.183.3.1161. PMID 8642258. 
  25. "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. July 2000. doi:10.1038/77498. PMID 10888925. 
  26. "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. January 2003. doi:10.1097/00002030-200301030-00006. PMID 12478067. 
  27. "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. July 2006. doi:10.1007/s00262-005-0075-9. PMID 16283305. 
  28. "Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication". Leukemia 20 (9): 1487–95. September 2006. doi:10.1038/sj.leu.2404296. PMID 16791265. 
  29. "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. September 2007. doi:10.1634/stemcells.2007-0128. PMID 17556595. 
  30. "Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells". Nature Cell Biology 9 (6): 654–9. June 2007. doi:10.1038/ncb1596. PMID 17486113. 
  31. "Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers". Nature Cell Biology 10 (12): 1470–6. December 2008. doi:10.1038/ncb1800. PMID 19011622. 
  32. "Functional delivery of viral miRNAs via exosomes". Proceedings of the National Academy of Sciences of the United States of America 107 (14): 6328–33. April 2010. doi:10.1073/pnas.0914843107. PMID 20304794. Bibcode2010PNAS..107.6328P. 
  33. 33.0 33.1 "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. April 2022. doi:10.3390/cancers14092068. PMID 35565197. 
  34. "Microvesicles: messengers and mediators of tumor progression". Cell Cycle 8 (13): 2014–8. July 2009. doi:10.4161/cc.8.13.8988. PMID 19535896. 
  35. "Tumour exosome integrins determine organotropic metastasis". Nature 527 (7578): 329–35. November 2015. doi:10.1038/nature15756. PMID 26524530. Bibcode2015Natur.527..329H. 
  36. "Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET". Nature Medicine 18 (6): 883–91. June 2012. doi:10.1038/nm.2753. PMID 22635005. 
  37. "Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis". Cancer Cell 26 (5): 707–21. November 2014. doi:10.1016/j.ccell.2014.09.005. PMID 25446899. 
  38. "Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer". Nature 546 (7659): 498–503. June 2017. doi:10.1038/nature22341. PMID 28607485. Bibcode2017Natur.546..498K. 
  39. "Rab27a and Rab27b control different steps of the exosome secretion pathway". Nature Cell Biology 12 (1): 19–30; sup pp 1–13. January 2010. doi:10.1038/ncb2000. PMID 19966785. 
  40. "Exosomes: a common pathway for a specialized function". Journal of Biochemistry 140 (1): 13–21. July 2006. doi:10.1093/jb/mvj128. PMID 16877764. 
  41. "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. February 2016. doi:10.1073/pnas.1521230113. PMID 26858453. Bibcode2016PNAS..113E.968K. 
  42. "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. January 2018. doi:10.1098/rstb.2016.0479. PMID 29158309. 
  43. "Cell Biology. NIH effort gambles on mysterious extracellular RNAs". Science 341 (6149): 947. August 2013. doi:10.1126/science.341.6149.947. PMID 23990535. 
  44. 44.0 44.1 "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. 2022. doi:10.3389/fcimb.2022.980817. PMID 36467728. 
  45. "Critical Review of the Evolution of Extracellular Vesicles' Knowledge: From 1946 to Today". International Journal of Molecular Sciences 22 (12): 6417. June 2021. doi:10.3390/ijms22126417. PMID 34203956. 
  46. "Review of the Isolation, Characterization, Biological Function, and Multifarious Therapeutic Approaches of Exosomes". Cells 8 (4): 307. April 2019. doi:10.3390/cells8040307. PMID 30987213. 
  47. 47.0 47.1 47.2 47.3 47.4 47.5 47.6 "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. 2018. doi:10.1080/20013078.2018.1535750. PMID 30637094. 
  48. "Exfoliation of membrane ecto-enzymes in the form of micro-vesicles". Biochimica et Biophysica Acta (BBA) - Biomembranes 645 (1): 63–70. July 1981. doi:10.1016/0005-2736(81)90512-5. PMID 6266476. 
  49. "Large oncosomes mediate intercellular transfer of functional microRNA". Cell Cycle 12 (22): 3526–36. November 2013. doi:10.4161/cc.26539. PMID 24091630. 
  50. "Oncosomes - large and small: what are they, where they came from?". Journal of Extracellular Vesicles 5: 33109. 2016. doi:10.3402/jev.v5.33109. PMID 27680302. 
  51. "MYC Mediates Large Oncosome-Induced Fibroblast Reprogramming in Prostate Cancer". Cancer Research 77 (9): 2306–2317. 2017. doi:10.1158/0008-5472.CAN-16-2942. PMID 28202510. 
  52. "A GBM-like V-ATPase signature directs cell-cell tumor signaling and reprogramming via large oncosomes". eBioMedicine 41: 225–235. 2019. doi:10.1016/j.ebiom.2019.01.051. PMID 30737083. 
  53. 53.0 53.1 53.2 "C. elegans neurons jettison protein aggregates and mitochondria under neurotoxic stress". Nature 542 (7641): 367–371. February 2017. doi:10.1038/nature21362. PMID 28178240. Bibcode2017Natur.542..367M. 
  54. "A Network of Macrophages Supports Mitochondrial Homeostasis in the Heart". Cell 183 (1): 94–109. 2020. doi:10.1016/j.cell.2020.08.031. PMID 32937105. 
  55. "Discovery of the migrasome, an organelle mediating release of cytoplasmic contents during cell migration". Cell Research 25 (1): 24–38. 2015. doi:10.1038/cr.2014.135. PMID 25342562. 
  56. "Mitocytosis, a migrasome-mediated mitochondrial quality-control process". Cell 184 (11): 2896–2910. 2021. doi:10.1016/j.cell.2021.04.027. PMID 34048705. 
  57. "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. August 2016. doi:10.1073/pnas.1605146113. PMID 27432966. Bibcode2016PNAS..113.9155N. 
  58. "Obstacles and opportunities in the functional analysis of extracellular vesicle RNA - an ISEV position paper". Journal of Extracellular Vesicles 6 (1): 1286095. 2017. doi:10.1080/20013078.2017.1286095. PMID 28326170. 
  59. "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. December 2019. doi:10.1016/j.aca.2019.09.022. PMID 31679569. Bibcode2019AcAC.1091..160M. 
  60. "Automated On-Line Isolation and Fractionation System for Nanosized Biomacromolecules from Human Plasma". Analytical Chemistry 92 (19): 13058–13065. October 2020. doi:10.1021/acs.analchem.0c01986. PMID 32893620. 
  61. "Electrokinetic characterization of extracellular vesicles with capillary electrophoresis: A new tool for their identification and quantification". Analytica Chimica Acta 1128: 42–51. September 2020. doi:10.1016/j.aca.2020.06.073. PMID 32825911. Bibcode2020AcAC.1128...42M. 
  62. "Impact of pre-analytical parameters on the measurement of circulating microparticles: towards standardization of protocol". Journal of Thrombosis and Haemostasis 10 (3): 437–46. March 2012. doi:10.1111/j.1538-7836.2011.04610.x. PMID 22212198. 
  63. "Standardization of sample collection, isolation and analysis methods in extracellular vesicle research". Journal of Extracellular Vesicles 2: 20360. 2013. doi:10.3402/jev.v2i0.20360. PMID 24009894. 
  64. "Methodological Guidelines to Study Extracellular Vesicles". Circulation Research 120 (10): 1632–1648. May 2017. doi:10.1161/CIRCRESAHA.117.309417. PMID 28495994. 
  65. "Modern isolation and separation techniques for extracellular vesicles". Journal of Chromatography A 1636: 461773. January 2021. doi:10.1016/j.chroma.2020.461773. ISSN 0021-9673. PMID 33316564. 
  66. "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. 2014. doi:10.3402/jev.v3.26913. PMID 25536934. 
  67. "EV-TRACK: transparent reporting and centralizing knowledge in extracellular vesicle research". Nature Methods 14 (3): 228–232. February 2017. doi:10.1038/nmeth.4185. PMID 28245209. 
  68. "Unravelling the proteomic landscape of extracellular vesicles in prostate cancer by density-based fractionation of urine". Journal of Extracellular Vesicles 9 (1): 1736935. 11 March 2020. doi:10.1080/20013078.2020.1736935. PMID 32284825. 
  69. "Roles of extracellular vesicles in the aging microenvironment and age-related diseases". Journal of Extracellular Vesicles 10 (12): e12154. October 2021. doi:10.1002/jev2.12154. PMID 34609061. 
  70. "The role of exosomes and microRNAs in senescence and aging". Advanced Drug Delivery Reviews 65 (3): 368–375. March 2013. doi:10.1016/j.addr.2012.07.010. PMID 22820533. 
  71. "MicroRNA controls of cellular senescence". BMB Reports 51 (10): 493–499. October 2018. doi:10.5483/BMBRep.2018.51.10.209. PMID 30269742. 
  72. "Hepatocyte-derived extracellular vesicles promote endothelial inflammation and atherogenesis via microRNA-1". Journal of Hepatology 72 (1): 156–166. January 2020. doi:10.1016/j.jhep.2019.09.014. PMID 31568800. 
  73. "Rejuvenating effects of young extracellular vesicles in aged rats and in cellular models of human senescence". Scientific Reports 13 (1): 12240. July 2023. doi:10.1038/s41598-023-39370-5. PMID 37507448. Bibcode2023NatSR..1312240G. 
  74. "Significance of Extracellular Vesicles: Pathobiological Roles in Disease". Cell Structure and Function 41 (2): 137–143. November 2016. doi:10.1247/csf.16014. PMID 27679938. 
  75. "Extracellular Vesicles Orchestrate Immune and Tumor Interaction Networks". Cancers 12 (12): 3696. December 2020. doi:10.3390/cancers12123696. PMID 33317058. 
  76. "Tumour-Derived Extracellular Vesicles (EVs): A Dangerous "Message in A Bottle" for Bone". International Journal of Molecular Sciences 20 (19): 4805. September 2019. doi:10.3390/ijms20194805. PMID 31569680. 
  77. "Accumulation of amyloid-β by astrocytes result in enlarged endosomes and microvesicle-induced apoptosis of neurons". Molecular Neurodegeneration 11 (1): 38. May 2016. doi:10.1186/s13024-016-0098-z. PMID 27176225. 
  78. "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. 2017. doi:10.3233/JAD-170278. PMID 28826183. 
  79. "Acute lymphoblastic leukemia-derived extracellular vesicles affect quiescence of hematopoietic stem and progenitor cells". Cell Death Dis. 12 (4): 337. 2022. doi:10.1038/s41419-022-04761-5. PMID 35414137. 
  80. "An intercellular transfer of telomeres rescues T cells from senescence and promotes long-term immunological memory". Nature Cell Biology 24 (10): 1461–1474. October 2022. doi:10.1038/s41556-022-00991-z. PMID 36109671. 
  81. "Understanding wiring and volume transmission". Brain Research Reviews 64 (1): 137–159. September 2010. doi:10.1016/j.brainresrev.2010.03.003. PMID 20347870. 
  82. "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. 2016. doi:10.3389/fncel.2016.00109. PMID 27199663. 
  83. "Myeloid microvesicles are a marker and therapeutic target for neuroinflammation". Annals of Neurology 72 (4): 610–624. October 2012. doi:10.1002/ana.23627. PMID 23109155. https://www.openaccessrepository.it/record/21830. 
  84. "Exosome-based strategies for Diagnosis and Therapy". Recent Patents on CNS Drug Discovery 10 (1): 10–27. 2015. doi:10.2174/1574889810666150702124059. PMID 26133463. 
  85. "Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers". Nature Cell Biology 10 (12): 1470–1476. December 2008. doi:10.1038/ncb1800. PMID 19011622. 

External links