Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2614 word(s) 2614 2021-03-09 04:40:00 |
2 format correct Meta information modification 2614 2021-03-25 11:12:30 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
J. Blanco Prieto, M. Extracellular Vesicle. Encyclopedia. Available online: https://encyclopedia.pub/entry/8262 (accessed on 21 July 2024).
J. Blanco Prieto M. Extracellular Vesicle. Encyclopedia. Available at: https://encyclopedia.pub/entry/8262. Accessed July 21, 2024.
J. Blanco Prieto, María. "Extracellular Vesicle" Encyclopedia, https://encyclopedia.pub/entry/8262 (accessed July 21, 2024).
J. Blanco Prieto, M. (2021, March 25). Extracellular Vesicle. In Encyclopedia. https://encyclopedia.pub/entry/8262
J. Blanco Prieto, María. "Extracellular Vesicle." Encyclopedia. Web. 25 March, 2021.
Extracellular Vesicle
Edit

Extracellular vesicles (EVs) are constituted by a group of heterogeneous membrane vesicles secreted by most cell types that play a crucial role in cell–cell communication.

cardiovascular diseases myocardial infarction cardiac repair extracellular vesicles exosomes drug delivery cargo loading targeting

1. Extracellular Vesicles

1.1. Classification

EVs comprise submicron particles heterogeneous in size, delimited by a lipid bilayer that cannot replicate. Traditionally, they have been classified according to their size and biogenesis, distinguishing: small particles or exosomes of endosomal origin with diameters ranging from 30 to 150 nm; ectosomes or microvesicles directly shed from the plasma membrane and polydisperse in size (100–1000 nm); and apoptotic bodies generated as a consequence of programmed cell death (1000–5000 nm) [1][2]. However, in recent years it has become apparent that the picture is more complex than expected. Assigning an EV to a particular biogenesis pathway still remains extraordinarily difficult given the overlap in size-distribution and protein-expression patterns among different EV types, especially when referring to exosomes and microvesicles, challenging the attempts to define a more precise nomenclature for EV classes [3]. Consequently, the latest recommendations of the ISEV encourage authors to define EV subtypes considering their physical characteristics—attending to: (a) size (small, medium/large) or density (low, medium, high) according to a defined range, (b) biochemical composition relaying on specific markers (e.g., CD63, CD81, annexin V, etc.), (c) isolation conditions (e.g., hypoxia, serum conditioning), and/or d) cellular origin (platelet, endothelial, cardiomyocytes, etc.)—rather than by the use of the traditional terms exosomes or microvesicles [1]. A summary of EVs’ physical and biochemical properties as well as parental cell conditions is provided in Table 1. In this review, the term EVs will refer to both exosomes and microvesicles.

Table 1. Requirements for extracellular vesicles’ separation and classification based on their physical and biochemical properties as well as donor cell conditions.

Physical properties Size
[1]
Small EVs <100 nm
Small/medium EVs <200 nm
Medium/large EVs >200 nm
Density
(in sucrose) [4]
Low 1.13–1.19 g/mL
Medium 1.16–1.28 g/mL
High >1.28 g/mL
Biochemical composition Surface antigens
[1]
Tetraspanins
MHC class I
Integrins
Transferrin receptor
LAMP1/2
Heparan sulfate
Proteoglycans
EMMPRIN
ADAM10
GPI-anchored 5ʹnucleotidase CD73
Complement-binding proteins CD55 and CD59
Sonic hedgehog
Lipids
[1][5]
Phosphatidylserine
Phosphatidylinositol
Phosphatidylethanolamine
Phosphatidylcholine
Cholesterol
Ceramide
Diacylglycerol
Glycosphingolipids
Internal cargo
[1][6][7]
Proteins TSG101
ALIX
VPS4A/B
ARRDC1
Flotillins-1 and 2
Caveolins
Annexins
Heat shock proteins HSC70 and HSP84
Syntenin
Cardiac-related miRNAs let-7
miR-16
miR-17-92
miR-19b
miR-20a/b
miR-21a
miR-24
miR-26a
miR-34
miR-93
miR-94a
miR-107a
miR-125b
miR-126
miR-130a/b
miR-132
miR-143
miR-145
miR-146a
miR-181b
miR-182
miR-208a
miR-210
miR-214
miR-294
miR-302a
miR-451
Conditions at EVs harvest Cell culture conditions
[1]
Normoxia
Hypoxia
Surface coating
Treatment
Grade of confluency
Passage number
Donor status
[1]
Age
Biological sex
Circadian variation
Body mass index
Pathological/healthy condition
Exercise level
Diet
Medication

ADAM10: ADAM metallopeptidase domain 10; ALIX: ALG-2-interacting protein X; ARRDC1: arrestin domain-containing protein 1; EMMPRIN: extracellular matrix metalloproteinase inducer; EVs: extracellular vesicles; GPI: glycosylphosphatidylinositol; LAMP: lysosomal-associated membrane protein; MHC: major histocompatibility complex; miRNA: microRNA; TSG101: tumor susceptibility gene 101; VPS4A/B: Vacuolar Protein Sorting 4 Homolog A/B.

EVs are released into the extracellular space by most cell types and can be found in a wide range of body fluids as reservoirs of lipids, proteins, nucleic acids, and carbohydrates of their parental cells. Current knowledge supports the view that each cell type tunes EVs’ biogenesis, depending on its activation status. Moreover, their cargo is particular to the stimulus or biological condition triggering their formation and release, suggesting the existence of intracellular selective cargo-sorting mechanisms. Subsequently, EVs’ composition will directly affect their fate and function [2][8][9]. There is a consensus about two major EV biogenesis pathways, giving rise to the most widely studied subpopulations, exosomes and microvesicles. Exosomes, generated within the endosomal system, are intraluminal vesicles formed by the inward budding of the endosomal membrane during maturation of multivesicular endosomes (MVEs) and are released to the extracellular space by fusion of MVEs with the cell membrane [10]. As for microvesicles, they originate by outward budding of the plasma membrane [11]. Microvesicle biogenesis determines the expression of surface-specific antigens from the cell origin, as well as the externalization of phosphatidylserine on the outer membrane leaflet [12], although the latter is not a prerequisite in all microvesicles [13]. Despite the aforementioned differences in biogenesis, the overlap in size, density, or composition, together with the lack of appropriate technology, hampers the possibility of distinguishing EV subpopulations once released to the extracellular medium, favoring the use of the generic term EVs, instead of a more specific nomenclature [1].

1.3. Mechanism of Action

The lipid bilayer protects EVs’ content from degradation by nucleases and proteinases present in biofluids, enabling the transfer of proteins, lipids, or nucleic acids from parental cells to recipient cells [14]. As such, EVs contribute to normal homeostasis, but also to the progression of several pathologies, including CVDs [15][16]. The mechanisms by which EVs mediate intercellular communication are not completely understood but are supposed to involve specific interactions between proteins or lipids enriched at the EVs surface (e.g., tetraspanins, integrins, lectins, phosphatidylserine) and receptors at the plasma membrane of recipient cells (e.g., intercellular adhesion molecules (ICAMs), annexin V, galectin 5) [2][17][18][19][20]. After docking at the cell membrane, EVs can remain at the binding site eliciting functional responses in recipient cells by activating downstream molecular pathways, or by direct interaction with extracellular matrix components [21] (Figure 1). They can also be internalized by endocytosis or by fusion with the plasma membrane undergoing different fates. For instance, endocytosed EVs can reach the MVEs and be targeted for degradation by lysosomes, they can escape digestion by back fusion with the MVEs’ membrane, or they can be re-secreted to the extracellular space via the early endocytic recycling pathway [21][22][23][24][25]. Either by direct fusion with the plasma membrane or after escaping lysosomal degradation, EVs can release their content into the cytoplasm of recipient cells and regulate cellular processes [26] (Figure 1).

Figure 1. Mechanism of action of EVs. After released from donor cells, EVs may induce a response in recipient cells by different mechanisms. First, EVs may remain at the binding site on the cell membrane eliciting functional responses by activating downstream molecular pathways. Alternatively, EVs may be internalized by endocytosis or fusion with the cell membrane undergoing different intracellular fates. They can be targeted for degradation by lysosomes, they can escape degradation and modulate cell behavior, or they can be re-secreted to the extracellular space.

1.4. Separation and Characterization of Extracellular Vesicles

EVs can be separated from the cell culture medium and most body fluids (liquid biopsy), blood being the most frequently studied source of EVs. Before EV separation, some preanalytical parameters should be considered [27][28], such as the use of serum-free media or EV-depleted serum for EV separation from conditioned medium [1][29]. Moreover, differences in the physicochemical and biochemical properties of the selected separation methods can impact the enriched EV subpopulations [30][31][32][33][34] and do not enable an absolute purification of EVs from other contaminants [35].

Ultracentrifugation (UC) is the most commonly used EV separation and enrichment technique based on particle density, involving multiple centrifugation and ultracentrifugation steps [30]. Speeds of 10,000–20,000 g enable the separation of medium/large vesicles, while small-sized vesicles are recovered at higher speeds (100,000 g).

Size exclusion techniques include ultrafiltration and chromatography. Ultrafiltration is usually based on cellulose filters defined by molecular mass and size exclusion range [31], while size exclusion chromatography (SEC), separates fractions by elution with phosphate buffered saline (PBS) and has been proven to be reliable and scalable for various applications [36].

Immune affinity isolation is based on the immunolabeling of proteins on the surface of EVs, enabling the separation of specific particle subpopulations from other EV classes, contaminant protein aggregates or lipoproteins. Usually, specific antibodies are conjugated to magnetic beads and EVs are separated using magnets [37][38].

A range of commercial kits are also available, some based on polymer precipitation- methods [31][32][33] and others on non-precipitation alternatives, for instance, those selective for phosphatidylserine positive vesicles [29]. Alternative or complementary techniques to classical procedures are also emerging, including microfluidics, asymmetric flow field-flow fractionation, or high-resolution flow cytometry [35].

After separation, EVs’ purity should be tested by the use of multiple complementary methods: (i) western blotting to analyze EVs markers (e.g., CD63, Alix, etc.) and co-isolated contaminants [1]; (ii) nanoparticle tracking analysis (NTA), which can determine particle size and concentration [39]; (iii) conventional transmission electron microscopy (TEM) and the more strongly recommended cryo-TEM [40], or (iv) nanoflow cytometry, that enables the determination of cell surface antigens, the quantification of EV subpopulations based on parental cell markers [41][42], and the lipid nature of the studied particles with cell-permeant, non-fluorescent pro-dyes . Also, current advances in EV-adapted proteomic, lipidomic, and genomic technologies will greatly help to delimit the molecular signature of the EV subpopulations under research.

2. Potential Applications of Extracellular Vesicles as Therapeutic Agents in Myocardial Infarction

Driven by the drawbacks associated with cell transplantation as well as the key role of stem cell paracrine secretion in cardiac repair, EVs have emerged recently as a next-generation cell-free regenerative therapy. Several studies have been performed in the last five years, aiming to test EVs’ potential as cell substitutes in the cardiac regenerative field, with significant preclinical success (Figure 2). All these studies collect different cell sources, isolation techniques, therapeutic doses, or administration routes, reflecting the heterogeneity and immature nature of the field. Here, we group the most relevant findings from these studies as well as a brief compendium of the EV-associated molecules involved in heart repair, based on the parental cell type. A summary of these can be found in Table 2.

Figure 2. Summary of the beneficial effects of EVs in cardiac repair. Administration of EVs in MI preclinical models showed that EVs modulate a regenerative response in several cardiac cells, including cardiomyocytes, macrophages, endothelial cells, and fibroblasts. Together, these cell-level effects result in the reduction of infarct size and the improvement of cardiac function after MI.

Table 2. Representative preclinical efficacy studies from the last five years, using extracellular vesicles as therapeutic agents for myocardial infarction.

Cell Source Isolation Method Animal Model Dose Administration Route and Time Post-MI Reparative Effect Molecule/Mechanism Involved Ref
MSCs
Rat BM-MSCs Total Exosome Isolation Kit (Invitrogen) Rat, permanent 20 µg IM; immediate
  • Improved cardiac function
  • Reduced fibrosis
  • Reduced inflammation
-  
Mouse BM-MSCs Density- gradient UC Mouse, I/R 50 µg IM; immediate after reperfusion
  • Reduced infarct size
  • Alleviated inflammation (polarization of macrophages to M2 phenotype)
Inhibition of TLR4 by miR-182  
Proinflammatory rat BM-MSCs Density-gradient UC Mouse, permanent 50 µg IM; immediate
  • Reduced inflammation
  • Anti-inflammatory macrophage polarization
  • Reduced cardiomyocyte apoptosis
Suppression of NF-κB and regulation of AKT1/AKT2  
BM-MSCs UC Rat, permanent 10 µg EVs (and 2×106 BM-MSCs) IM; at 30 min
  • Improved cardiac function
  • Reduced infarct size and fibrosis
  • Increased vascularization
  • Reduced inflammation
  • Enhanced recruitment of IV-infused MSCs
-  
ATV-pre-treated rat BM-MSCs UC Rat, permanent 10 µg IM; immediate
  • Improved cardiac function
  • Reduced infarct size
  • Decreased cardiomyocyte apoptosis
  • Increased angiogenesis
lncRNA H19 and miR-675  
Mouse BM-MSCs UC Mouse, permanent - IV; immediate and
day 6
  • Improved cardiac function
  • Angiogenesis
  • Reduced fibrosis
miR-210 and Efna3 gene suppression  
Mouse BM-MSCs UC Mouse, permanent EVs derived from 2×107 cells IM; immediate
  • Improved cardiac function
  • Angiogenesis
  • Decreased scar size
  • Reduced cardiomyocyte survival
  • Activation of resident CPCs
miR-210  
Rat BM-MSCs Total Exosome Isolation Kit (Invitrogen) Rat, I/R 5 µg IM; prior to reperfusion
  • Decreased cardiomyocyte apoptosis
  • Reduced infarct size
  • Improved heart function by an enhanced autophagy
AMPK and AKT pathways  
Mouse BM-MSCs UC Mouse, I/R 12.5 µg/ 5.62×105 EVs IM; 24h prior to ischemia • Decreased infarct size Reduced expression of pro-apoptotic genes PDCD4, PTEN, Peli1 and FasL via miR-21a-5p  
Mouse BM-MSCs UC Mouse, permanent 200 µg IM; immediate
  • Improved cardiac function
  • Reduced infarct size
miR-125b  
BM-MSCs ExoQuick Rat, permanent - IM; immediate
  • Reduced infarct size
  • Alleviated cardiomyocyte apoptosis
  • Improved cardiac function
miR-24  
Rat ADSCs UC Rat, permanent 2.5×1012 particles IV; at 1h
  • Decreased fibrosis
  • Decreased cell apoptosis
  • Attenuated inflammation via anti-inflammatory macrophage polarization
  • Improved cardiac function
S1P/SK1/S1PR1 activation  
Rat ADSCs Ultrafiltration and UC Rat, I/R 400 µg IV; at reperfusion
  • Reduced infarct area
  • Attenuated apoptosis
  • Reduced serum levels of cardiac damage markers
Wnt/β-catenin activation  
Human umbilical cord MSCs Density-gradient UC Rat, permanent 400 µg and 800 µg IV; once daily for 7 days • Safety: no effect on hemolysis, no vascular and muscle stimulation, no side effects on hematology indexes, liver and renal function, and protective effect on weight loss -  
Human umbilical cord MSCs ExoQuick-TC (System Biosciences) Rat, permanent 400 µg IM; immediate
  • Increased density of myofibroblasts
  • Attenuated inflammation
  • Reduced cardiomyocyte apoptosis
-  
Human umbilical cord MSCs Density-gradient UC Rat, permanent 400 µg IV; immediate
  • Improved cardiac function
  • Increased cardiomyocyte survival
Upregulation of Smad7 by inhibition of miR-125b-5p  
Cardiac MSCs Precipitacion with PEG Mouse, permanent 50 µg IM; immediate
  • Improved cardiac function
  • Increased scar thickness
  • Angiogenesis
  • Cardiomyocyte proliferation
-  
CDCs
Human CDCs Ultrafiltration and precipitation with PEG Pig, I/R 7.5 mg IC; 30 min after reperfusion
IM; 30 min after reperfusion
  • Decreased infarct size and preserved LV function
  • Reduced leukocyte infiltration
  • Reduced fibrotic mass
  • Higher arteriolar density
-  
Porcine CDCs Ultrafiltration followed by Field-Flow Fractionation Pig, I/R 9.16 mg IM; at 72h after reperfusion
  • Inconclusive; tendency to reduce infarct size and increase cardiac function
  • Increased M2 macrophages
-  
Human CDCs Ultrafiltration and PEG precipitation Pig, I/R 7.5 mg IM; at 20 min after reperfusion
  • Preserved cardiac function
  • Reduced microvascular occlusion
  • Attenuated infarct size
  • Reduced CD68+ macrophages infiltration
Regulation of gene expression by miRNA  
Human CDCs Ultrafiltration and precipitation with PEG Rat, I/R 350 µg IM; at 30 min after reperfusion
  • Preserved cardiac function
  • Reduced infarcted area
-  
Human CDCs ExoQuick (precipitation) Rat, permanent 250 µg IM; at 4 weeks
  • Improved cardiac function
  • Reduced scar mass
  • Increased wall thickness
  • Increased capillary and microvessel density
Regulation of gene expression by miRNA  
CPCs
Human CPCs Density-gradient UC Mice, permanent 8 µg IM; at 15 min
  • Reduced infarct size
  • Increased proliferation of cardiomyocytes and endothelial cells
Activation of endoglin in endothelial cells  
Rat CPCs UC Rat, I/R 5 µg/kg IM; during reperfusion
  • Reduced infarct size
  • Increased cardiac contractility
Decreased levels of collagen I, collagen III, vimentin and CTGF
Regulation of gene expression via miRNA
 
Human CPCs UC Rat, permanent and I/R 1011 particles IM; at 1h after permanent ligation or at reperfusion
  • Increased cardiac function
  • Reduced scar size
  • Increased blood vessel density
  • Decreased CD68+ macrophages
miR-146a-3p, miR-132, and miR-181a
PAPP-A
IGF-1
 
iPS
Human iPS UC Mouse, permanent 3×1010 particles Transcutaneous echo-guided IM; at 3 weeks • Increased cardiac function Regulation of gene expression via miRNA  
Human iPS UC Mouse, permanent 100 µg (1010 particles) IM; at 2 days or 3 weeks
  • No detectable humoral or immune response
  • Decreased pro-inflammatory monocytes and cytokines
-  
Mouse iPS UC Mouse, I/R 100 µg IM; at 48h after reperfusion
  • Preserved cardiac function
  • Improved systolic infarct wall thickness
  • Smaller LV end-systolic volume
  • Reduced apoptosis in myocytes
  • Increased capillary density
  • No tumor formation
Regulation of gene expression via miRNA and metabolic regulation via protein delivery
(in silico analysis)
 
ESC
Human ESC UC Mouse, permanent 20 µg IM; immediate
  • Improved cardiac function and LV systolic dimension
  • Reduced scar size
  • Decreased cardiomyocyte apoptosis
  • Higher number of endothelial cells
Targeting miR-497 by lncRNA MALAT1  
Human ESC UC Mouse, permanent - Transcutaneous echo-guided IM; at 2-3 weeks
  • Decreased LV end-systolic and diastolic volume
  • Reduced fibrosis
  • Smaller cardiomyocytes
Gene regulation of DNA repair, cell survival, cell cycle progression and cardiomyocyte contractility (in silico)  
Mouse ESC UC Mouse, permanent - IM; immediate
  • Enhanced contractility and decreased LV end-systolic diameter
  • Increased capillary density
  • Reduced apoptosis
  • Elevated cardiomyocyte proliferation
Regulation of CPC cell cycle and association with proliferation and survival mediated by miR-294  

ADSCs: adipose tissue-derived mesenchymal stem cells; ATV: atorvastatin; BM-MSCs: bone marrow-derived mesenchymal stromal cells; CDCs: cardiosphere-derived cells; CPCs: cardiac progenitor cells; CTGF: connective tissue growth factor; ESC: embryonic stem cells; EVs: extracellular vesicles; I/R: ischemia/reperfusion; IC: intracoronary; IGF-1: insulin-like growth factor-1; IM: intramyocardially; iPS: induced pluripotent stem cells; IV: intravenously; lncRNA: long non-coding RNA; LV: left ventricle; MALAT1: metastasis-associated lung adenocarcinoma transcript 1; MI: myocardial infarction; miRNA: microRNA; MSCs: mesenchymal stromal cells; NF-κB: nuclear factor kappa B; PAPP-A: pregnancy-associated plasma protein A; PEG: polyethylene glycol; S1P: sphingosine 1-phosphate; S1PR1: sphingosine-1-phosphate receptor 1; SK1: sphingosine kinase 1; Smad7: mothers against decapentaplegic homolog 7; TLR4: toll-like receptor 4; UC: ultracentrifugation.

 

References

  1. World Health Organization Cardiovascular Diseases (CVDs). Available online: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds) (accessed on 4 December 2020).
  2. Andreadou, I.; Cabrera-Fuentes, H.A.; Devaux, Y.; Frangogiannis, N.G.; Frantz, S.; Guzik, T.; Liehn, E.; Pedrosa da Costa Gomes, C.; Schulz, R.; Hausenloy, D.J. Immune cells as targets for cardioprotection: New players and novel therapeutic opportunities. Res. 2019, 115, 1117–1130.
  3. Abbafati, C.; Machado, D.B.; Cislaghi, B.; Salman, O.M.; Karanikolos, M.; McKee, M.; Abbas, K.M.; Brady, O.J.; Larson, H.J.; Trias-Llimós, S.; et al. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2020, 396, 1204–1222, doi:10.1016/S0140-6736(20)30925-9.
  4. Burden, G.; Roth, G.A.; Mensah, G.A.; Johnson, C.O.; Addolorato, G.; Ammirati, E.; Baddour, L.M.; Banach, M.; Barengo, N.C.; Beaton, A.Z.; et al. Global Burden of Cardiovascular Diseases and Risk Factors, 1990–2019: Update From the GBD 2019 Study. Am. Coll. Cardiol. 2020, 76, 2982–3021, doi:10.1016/j.jacc.2020.11.010.
  5. de Abreu, R.C.; Fernandes, H.; da Costa Martins, P.A.; Sahoo, S.; Emanueli, C.; Ferreira, L. Native and bioengineered extracellular vesicles for cardiovascular therapeutics. Rev. Cardiol. 2020, 17, 685–697, doi:10.1038/s41569-020-0389-5.
  6. Marbán, E. A mechanistic roadmap for the clinical application of cardiac cell therapies. Biomed. Eng. 2018, 2, 353–361, doi:10.1038/s41551-018-0216-z.A.
  7. Marbán, E. The Secret Life of Exosomes: What Bees Can Teach Us About Next-Generation Therapeutics. Am. Coll. Cardiol. 2018, 71, 193–200, doi:10.1016/j.jacc.2017.11.013.
  8. Grigorian Shamagian, L.; Madonna, R.; Taylor, D.; Climent, A.M.; Prosper, F.; Bras-Rosario, L.; Bayes-Genis, A.; Ferdinandy, P.; Fernández-Avilés, F.; Izpisua Belmonte, J.C.; et al. Perspectives on Directions and Priorities for Future Preclinical Studies in Regenerative Medicine. Res. 2019, 124, 938–951, doi:10.1161/CIRCRESAHA.118.313795.
  9. Menasché, P. Cell therapy trials for heart regeneration—Lessons learned and future directions. Rev. Cardiol. 2018, 15, 659–671.
  10. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. 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. Extracell. Vesicles 2018, 7, 1535750, doi:10.1080/20013078.2018.1535750.
  11. Whittaker, T.E.; Nagelkerke, A.; Nele, V.; Kauscher, U.; Stevens, M.M. Experimental artefacts can lead to misattribution of bioactivity from soluble mesenchymal stem cell paracrine factors to extracellular vesicles. Extracell. Vesicles 2020, 9, 1807674, doi:10.1080/20013078.2020.1807674.
  12. Sluijter, J.P.G.; Davidson, S.M.; Boulanger, C.M.; Buzás, E.I.; De Kleijn, D.P.V.; Engel, F.B.; Giricz, Z.; Hausenloy, D.J.; Kishore, R.; Lecour, S.; et al. Extracellular vesicles in diagnostics and therapy of the ischaemic heart: Position Paper from the Working Group on Cellular Biology of the Heart of the European Society of Cardiology. Res. 2018, 114, 19–34, doi:10.1093/cvr/cvx211.
  13. Chong, S.Y.; Lee, C.K.; Huang, C.; Ou, Y.H.; Charles, C.J.; Richards, A.M.; Neupane, Y.R.; Pavon, M.V.; Zharkova, O.; Pastorin, G.; et al. Extracellular vesicles in cardiovascular diseases: Alternative biomarker sources, therapeutic agents, and drug delivery carriers. J. Mol. Sci. 2019, 20, 3272, doi:10.3390/ijms20133272.
  14. Martins-Marques, T.; Hausenloy, D.J.; Sluijter, J.P.G.; Leybaert, L.; Girao, H. Intercellular Communication in the Heart: Therapeutic Opportunities for Cardiac Ischemia. Trends Mol. Med. 2020, S1471-4914, 30263–X, doi:10.1016/j.molmed.2020.10.002.
  15. Tkach, M.; Théry, C. Communication by Extracellular Vesicles: Where We Are and Where We Need to Go. Cell 2016, 164, 1226–1232, doi:10.1016/j.cell.2016.01.043.
  16. Ghafarian, F.; Pashirzad, M.; Khazaei, M.; Rezayi, M.; Hassanian, S.M.; Ferns, G.A.; Avan, A. The clinical impact of exosomes in cardiovascular disorders: From basic science to clinical application. Cell. Physiol. 2019, 234, 12226–12236, doi:10.1002/jcp.27964.
  17. Saludas, L.; Pascual-Gil, S.; Roli, F.; Garbayo, E.; Blanco-Prieto, M.J. Heart tissue repair and cardioprotection using drug delivery systems. Maturitas 2018, 110, 1–9, doi:10.1016/J.MATURITAS.2018.01.011.
  18. Rezaie, J.; Rahbarghazi, R.; Pezeshki, M.; Mazhar, M.; Yekani, F.; Khaksar, M.; Shokrollahi, E.; Amini, H.; Hashemzadeh, S.; Sokullu, S.E.; et al. Cardioprotective role of extracellular vesicles: A highlight on exosome beneficial effects in cardiovascular diseases. Cell. Physiol. 2019, 234, 21732–21745, doi:10.1002/jcp.28894.
  19. Tikhomirov, R.; Donnell, B.R.O.; Catapano, F.; Faggian, G.; Gorelik, J.; Martelli, F.; Emanueli, C. Exosomes: From Potential Culprits to New Therapeutic Promise in the Setting of Cardiac Fibrosis. Cells 2020, 9, 592, doi:10.3390/cells9030592.
  20. Deb, A.; Gupta, S.; Mazumder, P.B. Exosomes: A new horizon in modern medicine. Life Sci. 2020, 118623, doi:10.1016/j.lfs.2020.118623.
  21. Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Rev. Mol. Cell Biol. 2018, 19, 213–228.
  22. Russell, A.E.; Sneider, A.; Witwer, K.W.; Bergese, P.; Bhattacharyya, S.N.; Cocks, A.; Cocucci, E.; Erdbrügger, U.; Falcon-Perez, J.M.; Freeman, D.W.; et al. Biological membranes in EV biogenesis, stability, uptake, and cargo transfer: An ISEV position paper arising from the ISEV membranes and EVs workshop. Extracell. Vesicles 2019, 8, 1684862, doi:10.1080/20013078.2019.1684862.
  23. Konoshenko, M.Y.; Lekchnov, E.A.; Vlassov, A.V.; Laktionov, P.P. Isolation of Extracellular Vesicles: General Methodologies and Latest Trends. Biomed Res. Int. 2018, 2018, doi:10.1155/2018/8545347.
  24. Skotland, T.; Sagini, K.; Sandvig, K.; Llorente, A. An emerging focus on lipids in extracellular vesicles. Drug Deliv. Rev. 2020, 159, 308–321, doi:10.1016/j.addr.2020.03.002.
  25. Femminò, S.; Penna, C.; Margarita, S.; Comità, S.; Brizzi, M.F.; Pagliaro, P. Extracellular vesicles and cardiovascular system: Biomarkers and Cardioprotective Effectors. Pharmacol. 2020, 135, 106790, doi:10.1016/j.vph.2020.106790.
  26. Kesidou, D.; da Costa Martins, P.A.; de Windt, L.J.; Brittan, M.; Beqqali, A.; Baker, A.H. Extracellular Vesicle miRNAs in the Promotion of Cardiac Neovascularisation. Physiol. 2020, 11, doi:10.3389/fphys.2020.579892.
  27. Pinheiro, A.; Silva, A.M.; Teixeira, J.H.; Gonçalves, R.M.; Almeida, M.I.; Barbosa, M.A.; Santos, S.G. Extracellular vesicles: Intelligent delivery strategies for therapeutic applications. Control. Release 2018, 289, 56–69.
  28. De Jong, B.; Barros, E.R.; Hoenderop, J.G.J.; Rigalli, J.P. Recent advances in extracellular vesicles as drug delivery systems and their potential in precision medicine. Pharmaceutics 2020, 12, 1006.
  29. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Rev. Cell Dev. Biol. 2014, 30, 255–289.
  30. Taylor, J.; Azimi, I.; Monteith, G.; Bebawy, M. Ca2+ mediates extracellular vesicle biogenesis through alternate pathways in malignancy. Extracell. Vesicles 2020, 9, doi:10.1080/20013078.2020.1734326.
  31. Boulanger, C.M.; Loyer, X.; Rautou, P.E.; Amabile, N. Extracellular vesicles in coronary artery disease. Rev. Cardiol. 2017, 14, 259–272.
  32. Connor, D.E.; Exner, T.; Ma, D.D.F.; Joseph, J.E. The majority of circulating platelet-derived microparticles fail to bind annexin V, lack phospholipid-dependent procoagulant activity and demonstrate greater expression of glycoprotein Ib. Haemost. 2010, 103, 1044–1052, doi:10.1160/TH09-09-0644.
  33. Momen-Heravi, F.; Getting, S.J.; Moschos, S.A. Extracellular vesicles and their nucleic acids for biomarker discovery. Ther. 2018, 192, 170–187, doi:10.1016/j.pharmthera.2018.08.002.
  34. Cesselli, D.; Parisse, P.; Aleksova, A.; Veneziano, C.; Cervellin, C.; Zanello, A.; Beltrami, A.P. Extracellular vesicles: How drug and pathology interfere with their biogenesis and function. Physiol. 2018, 9, 1394.
  35. Morelli, A.E.; Larregina, A.T.; Shufesky, W.J.; Sullivan, M.L.G.; Stolz, D.B.; Papworth, G.D.; Zahorchak, A.F.; Logar, A.J.; Wang, Z.; Watkins, S.C.; et al. Endocytosis, intracellular sorting, and processing of exosomes by dendritic cells. Blood 2004, 104, 3257–3266, doi:10.1182/blood-2004-03-0824.
  36. Nazarenko, I.; Rana, S.; Baumann, A.; McAlear, J.; Hellwig, A.; Trendelenburg, M.; Lochnit, G.; Preissner, K.T.; Zöller, M. Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Cancer Res. 2010, 70, 1668–1678, doi:10.1158/0008-5472.CAN-09-2470.
  37. Christianson, H.C.; Svensson, K.J.; Van Kuppevelt, T.H.; Li, J.P.; Belting, M. Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their internalization and functional activity. Natl. Acad. Sci. USA 2013, 110, 17380–17385, doi:10.1073/pnas.1304266110.
  38. Fuentes, P.; Sesé, M.; Guijarro, P.J.; Emperador, M.; Sánchez-Redondo, S.; Peinado, H.; Hümmer, S.; Ramón y. Cajal, S. ITGB3-mediated uptake of small extracellular vesicles facilitates intercellular communication in breast cancer cells. Commun. 2020, 11, doi:10.1038/s41467-020-18081-9.
  39. Gonda, A.; Kabagwira, J.; Senthil, G.N.; Wall, N.R. Internalization of exosomes through receptor-mediated endocytosis. Cancer Res. 2019, 17, 337–347.
  40. Feng, D.; Zhao, W.L.; Ye, Y.Y.; Bai, X.C.; Liu, R.Q.; Chang, L.F.; Zhou, Q.; Sui, S.F. Cellular internalization of exosomes occurs through phagocytosis. Traffic 2010, 11, 675–687, doi:10.1111/j.1600-0854.2010.01041.x.
  41. Ares, G.R.; Ortiz, P.A. Dynamin2, clathrin,andlipid rafts mediate endocytosis of the apical Na/K/2Cl cotransporter NKCC2 in thick ascending limbs. Biol. Chem. 2012, 287, 37824–37834, doi:10.1074/jbc.M112.386425.
  42. Nanbo, A.; Kawanishi, E.; Yoshida, R.; Yoshiyama, H. Exosomes Derived from Epstein-Barr Virus-Infected Cells Are Internalized via Caveola-Dependent Endocytosis and Promote Phenotypic Modulation in Target Cells. Virol. 2013, 87, 10334–10347, doi:10.1128/jvi.01310-13.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 1.1K
Revisions: 2 times (View History)
Update Date: 25 Mar 2021
1000/1000
Video Production Service