Next Article in Journal
Generation of Human Stomach Cancer iPSC-Derived Organoids Induced by Helicobacter pylori Infection and Their Application to Gastric Cancer Research
Next Article in Special Issue
Different Sourced Extracellular Vesicles and Their Potential Applications in Clinical Treatments
Previous Article in Journal
The Role of the Dynamic Lung Extracellular Matrix Environment on Fibroblast Morphology and Inflammation
Previous Article in Special Issue
Mesenchymal Stem Cell-Derived Exosomes: Applications in Regenerative Medicine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Methodologies to Isolate and Purify Clinical Grade Extracellular Vesicles for Medical Applications

1
Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
2
Department of Cancer Immunology, Genentech Inc., South San Francisco, CA 94080, USA
3
Molecular Medicine Department of Medicine, Stanford University, Palo Alto, CA 94304, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2022, 11(2), 186; https://doi.org/10.3390/cells11020186
Submission received: 15 November 2021 / Revised: 3 January 2022 / Accepted: 4 January 2022 / Published: 6 January 2022
(This article belongs to the Special Issue Stem Cells and Extracellular Vesicles)

Abstract

:
The use of extracellular vesicles (EV) in nano drug delivery has been demonstrated in many previous studies. In this study, we discuss the sources of extracellular vesicles, including plant, salivary and urinary sources which are easily available but less sought after compared with blood and tissue. Extensive research in the past decade has established that the breadth of EV applications is wide. However, the efforts on standardizing the isolation and purification methods have not brought us to a point that can match the potential of extracellular vesicles for clinical use. The standardization can open doors for many researchers and clinicians alike to experiment with the proposed clinical uses with lesser concerns regarding untraceable side effects. It can make it easier to identify the mechanism of therapeutic benefits and to track the mechanism of any unforeseen effects observed.

1. Introduction

Cell development and maintenance of homeostasis are related to intracellular communication, both in a specific site or between various tissues. Cells communicate via cell junctions, secretions and electrical stimuli [1,2]. Similarly, EVs are another mode of communication between cells and tissues. They carry cargos containing proteins, lipids, receptors and genetic molecules. Based on their cellular origin, different types of EVs have been identified: apoptotic bodies, micro vesicles and exosomes, to name a few [3,4,5].
Nomenclature standardization efforts have been made since the first International society for Extracellular Vesicles meeting (ISEV), but the umbrella term “extracellular vesicles” remains [6,7].
In the past couple of decades, extracellular vesicles have attracted the attention of the scientific community as a source of versatile communication mediators. Numerous studies are being performed to study them. Their characteristics, including small size, less toxicity and immunogenicity and being modifiable, make them suitable biomarkers and drug delivery vehicles [8,9,10,11].
The main challenges for studying extracellular vesicles are their isolation and characterization. There are some conventional methods including ultracentrifugation and ultrafiltration, and several novel techniques including microfluidic chips and immunoaffinity precipitation kits for this purpose [12,13,14]. The clinical use of EV can be affected by the isolation method [14,15]. Therefore, trying to choose the best protocol and customize it based on the study seems necessary.
Moreover, to study EVs, it is crucial to determine their characteristics. Various properties of EVs should be identified, such as size, density, concentration, protein and nucleotide content, surface protein and lipid structure [12,16,17]. Extracellular vesicles inherit their characteristics and content from their parent cells. Therefore, clarifying their characteristics can be used for the diagnosis and prognosis of various conditions [18,19,20,21].
In this study, sources of the extracellular vesicle are described, and various isolation and characterization techniques have been discussed.

2. Different Methods for EV Isolation and Purification

In the past few decades, there has been considerable attention on using extracellular vesicles as biomarkers for various conditions and as drug delivery vehicles. One of the challenges encountered for wide application is choosing an optimum, efficient and reliable isolation method [11,22]. Filtration, ultracentrifugation and affinity separation are the most common isolation methods [23,24,25]. To isolate well-purified and healthy extracellular vesicles, a suitable combination of isolation and purification methods is necessary, Figure 1 [26,27,28].
Various peptides, proteins, lipids and cell debris contaminants are present in the source samples, some of which are similar to EVs in structure and composition, whereas some interact with EVs, preventing extraction [22,29,30].
In the following paragraphs, EV isolation methods are discussed in further detail, Figure 1.

2.1. Centrifugation-Ultracentrifugation-Density Gradient

A centrifuge is a device for separating particles from a solution according to their size, shape and density, and the viscosity of the medium. It causes denser elements (cells, particles, proteins) to separate at the bottom of a tube. The greater the difference in density, the faster they separate [31,32].
Ultracentrifugation is the gold standard method for extracellular vesicle isolation. The different types of ultracentrifugation are differential ultracentrifugation, density gradient centrifugation and rate-zonal centrifugation techniques [23,33].
Differential ultracentrifugation was the first technique that was used for extracellular vesicle isolation. This method is based on density, size and shape of the EVs [23,34,35,36,37,38]. The duration of centrifugation, temperature and sample dilution play pivotal role in separation efficiency [39,40]. It is easy to use and requires no or slight sample pretreatment, but it takes longer time, needs more labor and a large sample [23,41,42]. In addition, various types of EVs cannot be separated via this technique [38]. Separation via density gradient could be considered for reaching higher purification as described below [23].
Density gradient centrifugation (DGC) is another ultracentrifugation method. The difference between UC and DGC is, centrifugation occurs in a tube that contains preconstructed density gradient medium in case of DGC [23,31,35]. Sucrose and iodixanol (OptiPrep®) are the most used media. Through this technique, extracellular vesicles can be separated from proteins [23]. Furthermore, various kinds of EVs could be separated based on their density [14]. Longer cycle durations, low yield rate and requirement of larger volume of sample (in comparison with UC) are the drawbacks of this technique [43,44].
Gradient centrifugation is rate-zonal centrifugation based on density gradient and sedimentation rate. The sample is added to the top of the tube and through centrifugation, compounds with higher density go through the dense layer, easier that lighter compounds. The duration of centrifugation should be controlled to avoid pellet construction at the bottom of the tube [23,45]. In addition, via this technique, particles with the same density and different diameter (size) can be separated [46]. This technique causes more extracellular recovery in comparison with density gradient centrifugation [23].

2.2. Precipitation

This method works based on dispersibility alteration [23,31]. A water-excluding compound is first added to the sample. Polyethylene glycol (PEG), a polymer, is the commonly used compound for this purpose. After adding the polymer, centrifugation or filtration is needed for separation. The polymer dries the sample and leads to the precipitation of other molecules. [30,31,47].
Precipitation is considered a quick and easy method for EV isolation and can be used for small or large volume samples. In addition, this method requires little proficiency and not a specialized apparatus. The selectivity and quality of the isolate (unspecific co-precipitation) in this method is poor, and it must be combined with other method(s) [23,31,35,48,49]. To overcome this, filtration or ultracentrifugation can be carried out before treatment with PEG [23,31]. In addition, some commercial precipitation kits have been developed [23,50]. These kits are fast, easy to use and do not require a specific apparatus. However, they are expensive, not applicable for large samples and are not efficient to separate different types of EVs [51,52,53]. Other compounds that can be used for precipitation are Acetat salt and protamine [51,54,55,56].
Precipitation should always be followed by centrifugation and filtration to eliminate contaminants [51,54,56].
Lectin is another chemical precipitant. In this technique, the sample is pretreated via centrifugation for separating cell debris and lectin is added to the sample and incubated overnight. Lectin conjugates with the carbohydrate of the exosome membrane, changes its solubility and causes precipitation, following which exosomes/EVs will be separated via centrifugation [23,31,57]. Chemical precipitations methods are simple, cost less, and are suitable for different sample sizes [51,54,56,58,59,60].

2.3. Size Based Approaches

As the title suggests, EV isolation here is based on size differentiation. Various techniques purify EVs based on size, including ultrafiltration, isolation kits, sequential filtration, size-exclusion chromatography (SEC), field-flow fractionation (FFF) and hydrostatic filtration dialysis (HFD) [23].
Ultrafiltration is the most common size-based isolation method. In this technique, sample goes through membrane filters with different pore sizes. EVs are then separated based on size and molecular weight [23,31]. One of the limitations of ultrafiltration is EV clogging and trapping in the membrane. This can be prevented by initial filtration using large pore filters, followed by filtration through small pore filters [23,48,61]. The other drawbacks of ultrafiltration are poor efficiency and filter plugging [23,48,62,63]. Ultrafiltration also leads to deformation of EVs due to the pre membrane pressure (this disadvantage can be reduced by forcing lower pressure). However, the technique is still popular as it is less time-consuming and does not require expensive instruments [23,48,50,63].
In recent times, isolation kits based on size differentiation have been developed. One of them is the ExoMir Kit (Bioo Scientific; Austin, TX, USA) that contains two different membranes (upper membrane: 200 nm and downer: 20 nm) in a syringe [23].
In addition, ExoTIC (exosome total isolation chip) technology is the other kit that could purify EVs by passing the sample through different filters. These kits are easy to use, have a high yield rate and can be used for different types of bio-fluid samples [64,65]. Other methods are tangential flow filtration (TFF), direct filtration, and cyclic TFF [66].
Sequential filtration is another technique where the sample is passed through different filters. In each step particles with larger size than membrane pores are trapped, and smaller particles go through it. It is a semi-automated technique. Therefore, it is easy to use and less time-consuming [62]. Filter trapping is a limitation of sequential filtration that leads to membrane plugging and yield rate reduction [62,67,68].
Size-exclusion chromatography is another method that isolates EVs based on size. It consists of a column that allows penetration of smaller particles. This causes bigger particles to exit the column earlier. This protects the structure, integrity and biological function of EVs [23,62,69,70,71,72]. In this method, the sample does not rely on extensive pretreatment [62,70].
The first time that this method was developed, starch and water were used to form pores in the column, but through time, various other compounds such as dextran polymer (Sephadex), agarose (Sepharose) and polyacrylamide (Sephacryl or BioGel) have been used [23,62,73,74,75].
The other innovation for EV isolation based on the size differentiation is field-flow fractionation (FFF). In this method, sample is injected into a chamber that is affected via a cross flow, whereas bigger particles will be pushed to the walls of the chamber due to the cross flow, smaller particles elute earlier [23,76]. This method provides an opportunity to isolate various types of EVs and even very tiny compounds. It is faster, highly efficient, label-free and has higher sample recovery [77].
Another technique called hydrostatic dialysis isolation (HDI) uses hydrostatic forces for isolation. Small particles diffuse through the membrane and larger ones stay in the tube [23,78]. Via this method, the Tamm–Horsfall Protein, one of the abundant proteins in urinary EVs, is eliminated. After HDI, centrifugation is performed for further purification [79].

2.4. Affinity

Affinity based EV isolation is based on the antigens present on the EV surface. Antigens on the EV membrane are considered as markers to distinguish their sources [23,80,81,82]. These antigens are captured via specific antibodies [23,35,83]. This method provides highly purified EVs, but the harvest rate is low [23,49]. Pretreatment of samples, especially plasma, with ultracentrifugation or ultrafiltration is necessary [23,81]. In the study conducted by Tauro BJ et al., the efficacy and the results of three different methods including ultracentrifugation, density gradient isolation and immunoaffinity capture method indicate that immunoaffinity causes the highest purification of EV [80]. The limitation of this method is related to the availability of antibodies of the identified antigen. Masking of the antigens on the EV surface can prevent isolation via immunoaffinity capture methods [14]. Enzyme-linked immunosorbent assay (ELISA) is the most common immunoaffinity-based isolation and identification method [23,84]. Samples should be pretreated with ultracentrifugation before affinity capture [23,31].
One of the most effective methods to elevate EV harvest through immunoaffinity is increasing the surface area of presenting antibodies. Magneto-immunoprecipitation is a technique used for this. In this, a biotinylated antibody specific to the presenting antigen is attached to the surface of magnetic beads coated with streptavidin. Isolated EVs are then detached and used for other purposes while preserving the activity of EV protein [23,31,85]. This technique is easy and fast, but a high affinity between antigen and antibody can prevent the detachment of EVs [86].
In a study that was performed by Zhang J et.al, a combination of three methods was used to reach the optimum level of EV purification. Their protocol contains tangential flow filtration, centrifugation and immunomagnetic affinity technique; the first and second steps produce purified EVs and with immunomagnetic affinity; EVs that contain specific markers are isolated [87,88].

2.5. Micro-, Nano-Fluidics, Chips

Micro-, nano-fluidic chips isolate EVs based on their biochemical properties using acoustic, electrophoretic, and electromagnetic technology. This method is fast, inexpensive, efficient and can be used on small samples [23,31,89,90,91].
Microchips have been developed to isolate EVs with different approaches, including immunoaffinity, size and density-based separations. Through immunoaffinity capture, markers on the EV membrane bind to their specific antibody on the beads or inner surfaces modified by antibodies. The major limitation is the need for appearance of specific antigen on the EV surface. Developing size based microchips can surpass this limitation [91].
For size-based isolation of microchips, pressure and electrophoresis techniques are used. Electrophoresis is preferred in comparison with pressure as it prevents pore blockage [90,91].
In addition, nanowires, nano-sized deterministic lateral displacement (nano-DLD) and viscoelastic flow are the other techniques that can be used. The mechanism using nanowires is similar to SEC and contains micro-porous silicon. Nano-DLD is a pillar-array-based microfluidic method that categorizes elements in an incessant stream [91], whereas viscoelastic flow is a novel passive and label-free technique in this category that separate particles based on variance among elastic lift forces executed on compounds with different sizes in a viscoelastic medium [91,92].
Acoustic separation is one of the techniques that is used via micro-fluidic chips. In this technique, the sample is exposed to ultrasonic waves. The larger particles are affected via heavier radiation and transferred to the pressure node faster. The ultrasonic wave frequencies are controlled to separate specific particles based on the size range [23,89]. Furthermore, development of this technique produces highly purified EVs and can separate them from very low density lipoproteins with remarkable efficiency [93].
The other technique in this group is immuno-based microfluidic isolation. The mechanism is similar to ELISA. Compared with ELISA, smaller samples can be used in this method (microliter). The specificity of the method is related to the specificity of antibodies that are immobilized on the chip [23,94,95]. As mentioned before, antibodies can be loaded on the beads located on the inner surface of the microchannel [96]. To reach the mentioned specificity, Exochip has been developed recently and due to the anti-CD68 antibodies (conjugate with CD68 that is expressed on the exosomes are released via various cell types) that are fixed on the microfluidic chips, the specific EV are isolated [23,97]. ExoSearch is the other microfluidic chip for EV isolation that can be used for smaller samples and consumes lesser time [23,94]. The modified magnetic beads via specific antibodies identify CA-125, EpCAM and CD24 on the EVs of ovarian cancer [94,98].

3. Comparison of Different Methodological Isolation Procedures

The optimal isolation method is one of the greatest challenges for the clinical use of EVs. As mentioned before, various types of isolation methods have been developed. Each method had its own advantages and disadvantages. When considering different methods, “an ideal method for isolation of EVs should be relatively simple, inexpensive, should not require a complex or expensive equipment, should be relatively fast and allow for isolation of EVs from a large volume of samples” [31,35].
The pros and cons of each method have been described in summary in Table 1.
The percentages of research studies published describing each isolation method are described in pie chart, Figure 2. The comparison studies have stated that UC is the primary isolation method for 41.5% of the evaluations (for source volumes from <1 to >100 mL) [99,100].

4. Clinical Application of EVs

4.1. EVs as Diagnosis Biomarkers

Extracellular vesicles are sacs that are secreted by almost all types of cells and are responsible for intracellular communication. They inherit their content and characteristics from their donor cells [10,112,113]. Pathological and physiological characteristics of donor cells are reflected in the appearance of specific nucleotide and proteins (on the EV surface or in their content) [10,114]. In addition, the rate of EV secretion can be changed in various conditions. Therefore, EVs are precise markers for the diagnosis, prognosis and monitoring of different pathologies [113,115].
Extracellular vesicles are desirable markers for Alzheimer’s disease (AD). For instance, increased levels of Aβ1-42, total tau, p-T181 tau and p-S396 tau have been determined in the exosomes that are isolated from the plasma of AD patients compared with healthy candidates [116,117]. Additionally, lysosomal protein such as cathepsin D and LAMP1 and synaptic proteins such as synaptophysin, synaptopodin, synaptotagmin-2 and neurogranin are increased and decreased in the exosome content of plasma in AD [118,119,120]. miRNA are the other molecules that can be searched in AD patient exosomes. miR-29b, miR-181c, miR-15b, miR-146a and miR-107 are examples of miRNA that increased in AD [120].
Cancers can also be diagnosed via EV markers. Increasing in the level of some miRNAs (let-7a, miR-1,229, miR-1,246, miR-150, miR-21, miR-223, and miR-23a) in exosomes have been found in early stage of colon cancer that can be considered as biomarkers [121,122,123]. In addition, high presentation of miRNA141 and miRNA195 have been detected in circulating exosomes of early stage breast cancer patients [124]. lncRNA content of exosomes can be the other candidate for cancer diagnosis, for instance lincRNA-p21 in the urine exosomes of prostate cancer patients [20,125,126]. The main challenge for considering a specific marker on the EV is overlapping markers of different conditions. For example, upregulation of CD95L on macro vesicles have been found to be related to oral squamous cell carcinoma; however, its elevated expression is also identified in pregnancy [114,127,128]. Changes that occurred in several protein expression including psoriasin, kertain-14, galectin-7, epidermal fatty acid binding protein (E-FABP), migration inhibitor factor-related protein (MRP8) and 14 and stratifin can be considered as bladder cancer biomarkers [129,130].
Extracellular can be used for cardiovascular disease diagnosis. miRNA-133a, miRNA-143/145, miRNA-150, miRNA-155, miRNA-214, miRNA-223, and miRNA320b in exosomes are considered as markers of arthrosclerosis (AS) [131,132]. Several proteins are suggested as biomarkers of AS, including integrins derived from exosomes secreted via macrophage foam cell [133], PSMA6 (prostate-specific membrane antigen), PSMA7, and annexin A2 in blood exosomes originated from innate immune system cells [21,134] and VCAM-1 (vascular cell adhesion molecule 1) and eNOS (endothelial constitutive nitric oxide synthase) identified in plasma endothelial cell-derived exosome [131,135].
miRNA-1 and miRNA-133a alter in circulation of Acute Coronary Syndromes (ACS) and Myocardial Infarction (MI) patients [136,137,138]. Extracellular vesicle miRNA content such as miRNA-133b, miRNA-208b, and miRNA-499 are considered as MI biomarkers [138,139]. In addition, miRNA-233 is the other exosomal miRNA that can be used for diagnosis of ischemic stroke [138,140]. The other examples of exosomal miRNA with diagnostic potential are miRNA-34a, miRNA-146a, MiRNA-92, miRNA-192, and miRNA-194 are associated with heart failure [138,141,142,143].
Furthermore, proteins of exosomes can be changed in cardiovascular disease that can be considered as biomarkers. TNF-a increases in hypoxia condition [144,145], angiotensin II (AngII) type 1 receptor is overexpressed in high pressure [145], polygenic immunoglobulin receptor (pIgR), complement factor C5a (C5a) and cystatin C are upregulated in Acute Coronary syndrome [146] and Complement C1q subcomponent subunit A (C1QA) and Complement C5 (C5), Apolipoprotein D (APOD) and Apolipoprotein C-III (APOCC3) and Platelet glycoprotein Ib alpha chain (GP1BA) and platelet basic protein (PPBP) are associated with MI [138,144,147].
Moreover, extracellular vesicles can be used for diagnosis and prognosis of some infectious diseases. Some examples are as follows: Akt and CD9 in exosomes increase during urinary tract infection and can be identified as biomarkers [148]. Exosomal lncRNA-HEIH increases in Chronic hepatitis C (CHC) related hepatocellular carcinoma and can be introduced as a marker [149]. Upregulation of neurofilament-light (NF-L), high mobility group box 1 (HMGB1) and amyloid β in circulating exosomes of HIV-infected patients can indicate neuronal damage via this virus [150,151].
Therefore, extracellular vesicles are potential biomarkers of various conditions. Their content and their release can be changed via a variety of diseases. to use EVs as biomarkers, optimal isolation methods and standardized characterization protocols are necessary for this purpose [120].

4.2. EVs as Therapeutic Vehicle

In addition to being markers, EVs are identified as biological vehicles. They can carry various kinds of compounds including peptides, proteins, lipids, nucleic acids (RNA, DNA) and carbohydrates. In addition, extracellular vesicles, especially exosomes, can be modified and loaded with various cargoes [152,153,154]. There are different types of extracellular vesicle loading methods that provide appropriate situation to load and deliver different compounds, these methods include, incubation, transfection, physical treatment (sonication, extrusion, freeze–thaw treatment, electroporation and surfactant treatment and dialysis), in situ association and synthesis [152,155].
Currently, the potential of different kinds of nucleic acids (including miRNA, siRNA, lncRNA, mRNA, DNA, etc.) against various conditions has been cleared [156,157,158]. Their clinical application is faced with various challenges such as degradability in blood that cause short half-life, immunogenicity, accumulation in kidney and live and disability to cross hydrophilic membrane [159,160,161,162,163,164]. Therefore, it seems crucial to develop an appropriate and efficient delivery vehicle [157]. A wide variety of studies have been performed using exosomes as delivery vehicle for nucleic acids. Among various types of EVs, micro vesicles are the most appropriate vehicle for them [165]. The summary of studies that deliver miRNA and siRNA via exosomes have been described in Table 2 and Table 3.
The evaluation of effects of exosomes derived from embryonic stem cells reveals that exosomes can transfer their mRNA content to the target cells and protect their functional activities [194,195]. In addition, exosomes derived from mesenchymal stem cells contain IGF-1 m-RNA that causes expression of IGF-1 and leads to renoprotective effects [196].
In addition, some studies have evaluated EVs to deliver peptides and proteins. For instance, exosomes loaded with Survivin-T34A cause growth limitation of prostate tumors via apoptosis induction [197]. Catalase, a redox enzyme, was loaded in macrophage-derived exosomes and injected intravenously to the mouse model of acute brain injury that lead to neuroprotective effects [198]. In the other study, human MUC1 (hMUC1) has been expressed in two MHC type-distinct mouse cell lines. The result of the study indicate that the engineered exosomes can stimulate immune system and suppress hMUC1-expressing tumor growth, specifically and efficiently [199]. In the other study, performed by Admyre C, a combination of 23 immunogenic peptides from EBV, CMV and influenza virus were loaded into monocyte-derived DC exosomes and exhibited immune system induction via stimulation of CD8+ T cell [200].
Furthermore, extracellular vesicles can carry some other compounds including synthetic and natural molecules. Doxorubicin [201,202], paclitaxel [203] and rhodamine 123 [204] delivery to the cancer cells and dopamine transferring into the brain [205] are the examples of the synthetic molecules. Curcumin and celasterol are the compounds that, when divided from plant extracts, have also been loaded into exosomes and used against inflammation and cancer, respectively [57,206,207,208].
Overall, it seems that extracellular vesicles are successful in embracing different types of cargoes. Furthermore, Extracellular vesicles can be modified for target therapy. Different approaches have been investigated for this purpose that was mentioned before [209,210]. The strategy for EV modification is related to the condition or characteristics of target cells [155]. The most common modification approach is designing exosomes via specific ligands to bind to target receptors. For this aim, there are two different techniques: 1. Direct assembling, 2. Transfection. Her2 and EGFR are located using these techniques and targeted in breast cancer and colorectal cancer, respectively [155,211,212]. The other approach for EV engineering is using pH-sensitive peptides on their surface, which in acidic environments causes deformation releasing the cargo [155,213]. In addition, magnetic directing can be used to direct therapy towards the target. EVs that are designed by magnetic compounds are directed by outer magnetic force to the target area [155,214]. This revealed the potential of EVs to load various cargoes and then modify as an appropriate vehicle for different molecules [155].

5. Conclusions

Isolation and characterization of EVs needs more attention to reach a high efficacy and specificity. For this, the characteristics of sample and study should be considered. In addition, advantages and disadvantages of methods as well as their properties and conditions help us to choose the most effective method [215,216]. Standardization of purification methods would be a great step to understand the requirements. Clear classification of these methods based on the characteristics of EVs, prognostic, diagnostic and clinical requirements, cost-effectiveness would be prudent. The main areas to focus on with regard to isolation and purification are high selectivity and greater efficiency [114,217,218]. This will increase the reach of EVs for clinicians and scientists to try the EVs as the need arises. This in turn would increase the knowledge regarding their behavior in clinically relevant scenarios.

Author Contributions

A.A., F.M., N.B., S.P.K., M.U. wrote, edited, and approved the published manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rasmussen, H. Cell communication, calcium ion, and cyclic adenosine monophosphate. Science 1970, 170, 404–412. [Google Scholar] [CrossRef]
  2. Krutovskikh, V.A.; Piccoli, C.; Yamasaki, H. Gap junction intercellular communication propagates cell death in cancerous cells. Oncogene 2002, 21, 1989–1999. [Google Scholar] [CrossRef] [Green Version]
  3. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef] [PubMed]
  4. Paolicelli, R.C.; Bergamini, G.; Rajendran, L. Cell-to-cell Communication by Extracellular Vesicles: Focus on Microglia. Neuroscience 2019, 405, 148–157. [Google Scholar] [CrossRef]
  5. Ratajczak, J.; Wysoczynski, M.; Hayek, F.; Janowskawieczorek, A.; Ratajczak, M.Z. Membrane-derived microvesicles: Important and underappreciated mediators of cell-to-cell communication. Leukemia 2006, 20, 1487–1495. [Google Scholar] [CrossRef] [PubMed]
  6. Witwer, K.W.; Théry, C. Extracellular vesicles or exosomes? On primacy, precision, and popularity influencing a choice of nomenclature. J. Extracell. Vesicles 2019, 8, 1648167. [Google Scholar] [CrossRef] [PubMed]
  7. Merchant, M.L.; Rood, I.M.; Deegens, J.K.J.; Klein, J.B. Isolation and characterization of urinary extracellular vesicles: Implications for biomarker discovery. Nat. Rev. Nephrol. 2017, 13, 731–749. [Google Scholar] [CrossRef] [Green Version]
  8. Andaloussi, S.E.; Mäger, I.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 2013, 12, 347–357. [Google Scholar] [CrossRef]
  9. Nawaz, M.; Camussi, G.; Valadi, H.; Nazarenko, I.; Ekström, K.; Wang, X.; Principe, S.; Shah, N.; Ashraf, N.M.; Fatima, F.; et al. The emerging role of extracellular vesicles as biomarkers for urogenital cancers. Nat. Rev. Urol. 2014, 11, 688–701. [Google Scholar] [CrossRef]
  10. Bernardi, S.; Balbi, C. Extracellular Vesicles: From Biomarkers to Therapeutic Tools. Biology 2020, 9, 258. [Google Scholar] [CrossRef]
  11. Kodam, S.P.; Ullah, M. Diagnostic and Therapeutic Potential of Extracellular Vesicles. Technol. Cancer Res. Treat. 2021, 20. [Google Scholar] [CrossRef]
  12. Xu, R.; Greening, D.W.; Zhu, H.-J.; Takahashi, N.; Simpson, R.J. Extracellular vesicle isolation and characterization: Toward clinical application. J. Clin. Investig. 2016, 126, 1152–1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Furi, I.; Momen-Heravi, F.; Szabo, G. Extracellular vesicle isolation: Present and future. Ann. Transl. Med. 2017, 5, 263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Taylor, D.D.; Shah, S. Methods of isolating extracellular vesicles impact down-stream analyses of their cargoes. Methods 2015, 87, 3–10. [Google Scholar] [CrossRef] [PubMed]
  15. Skovronova, R.; Grange, C.; Dimuccio, V.; Deregibus, M.C.; Camussi, G.; Bussolati, B. Surface Marker Expression in Small and Medium/Large Mesenchymal Stromal Cell-Derived Extracellular Vesicles in Naive or Apoptotic Condition Using Orthogonal Techniques. Cells 2021, 10, 2948. [Google Scholar] [CrossRef]
  16. Hartjes, T.A.; Mytnyk, S.; Jenster, G.W.; Van Steijn, V.; Van Royen, M.E. Extracellular Vesicle Quantification and Characterization: Common Methods and Emerging Approaches. Bioengineering 2019, 6, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Corso, G.; Heusermann, W.; Trojer, D.; Görgens, A.; Steib, E.; Voshol, J.; Graff, A.; Genoud, C.; Lee, Y.; Hean, J.; et al. Systematic characterization of extracellular vesicle sorting domains and quantification at the single molecule–single vesicle level by fluorescence correlation spectroscopy and single particle imaging. J. Extracell. Vesicles 2019, 8, 1663043. [Google Scholar] [CrossRef] [PubMed]
  18. Hoshino, A.; Kim, H.S.; Bojmar, L.; Gyan, K.E.; Cioffi, M.; Hernandez, J.; Zambirinis, C.P.; Rodrigues, G.; Molina, H.; Heissel, S.; et al. Extracellular vesicle and particle biomarkers define multiple human cancers. Cell 2020, 182, 1044–1061.e18. [Google Scholar] [CrossRef]
  19. Yekula, A.; Muralidharan, K.; Kang, K.M.; Wang, L.; Balaj, L.; Carter, B.S. From laboratory to clinic: Translation of extracellular vesicle based cancer biomarkers. Methods 2020, 177, 58–66. [Google Scholar] [CrossRef]
  20. Yousafzai, N.A.; Jin, H.; Ullah, M.; Wang, X. Recent advances of SIRT1 and implications in chemotherapeutics resistance in cancer. Am. J. Cancer Res. 2021, 11, 5233–5248. [Google Scholar]
  21. Ullah, A.; Mabood, N.; Maqbool, M.; Khan, L.; Ullah, M. Cytidine deamination-induced perpetual immunity to SAR-CoV-2 infection is a potential new therapeutic target. Int. J. Med. Sci. 2021, 18, 3788–3793. [Google Scholar] [CrossRef]
  22. Monguió-Tortajada, M.; Gálvez-Montón, C.; Bayes-Genis, A.; Roura, S.; Borràs, F.E. Extracellular vesicle isolation methods: Rising impact of size-exclusion chromatography. Experientia 2019, 76, 2369–2382. [Google Scholar] [CrossRef] [PubMed]
  23. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef] [Green Version]
  25. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [Green Version]
  26. Witwer, K.W.; Buzás, E.I.; Bemis, L.T.; Bora, A.; Lässer, C.; Lötvall, J.; Nolte-’t Hoen, E.N.; Piper, M.G.; Sivaraman, S.; Skog, J.; et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J. Extracell. Vesicles 2013, 2, 20360. [Google Scholar] [CrossRef]
  27. Ullah, M.; Qian, N.P.M.; Yannarelli, G.; Akbar, A. Heat shock protein 20 promotes sirtuin 1-dependent cell proliferation in induced pluripotent stem cells. World J. Stem Cells 2021, 13, 659–669. [Google Scholar] [CrossRef]
  28. Akbar, A.; Pillalamarri, N.; Jonnakuti, S.; Ullah, M. Artificial intelligence and guidance of medicine in the bubble. Cell Biosci. 2021, 11, 108. [Google Scholar] [CrossRef]
  29. Tian, Y.; Gong, M.; Hu, Y.; Liu, H.; Zhang, W.; Zhang, M.; Hu, X.; Aubert, D.; Zhu, S.; Wu, L.; et al. Quality and efficiency assessment of six extracellular vesicle isolation methods by nano-flow cytometry. J. Extracell. Vesicles 2019, 9, 1697028. [Google Scholar] [CrossRef]
  30. Ullah, A.; Mabood, N.; Maqbool, M.; Khan, L.; Khan, M.; Ullah, M. SAR-CoV-2 infection, emerging new variants and the role of activation induced cytidine deaminase (AID) in lasting immunity. Saudi Pharm. J. 2021, 29, 1181–1184. [Google Scholar] [CrossRef]
  31. Li, P.; Kaslan, M.; Lee, S.H.; Yao, J.; Gao, Z. Progress in Exosome Isolation Techniques. Theranostics 2017, 7, 789–804. [Google Scholar] [CrossRef]
  32. Ullah, M.; Qian, N.P.M.; Yannarelli, G. Advances in innovative exosome-technology for real time monitoring of viable drugs in clinical translation, prognosis and treatment response. Oncotarget 2021, 12, 1029–1031. [Google Scholar] [CrossRef] [PubMed]
  33. Pillalamarri, N.; Abdullah; Ren, G.; Khan, L.; Ullah, A.; Jonnakuti, S.; Ullah, M. Exploring the utility of extracellular vesicles in ameliorating viral infection-associated inflammation, cytokine storm and tissue damage. Transl. Oncol. 2021, 14, 101095. [Google Scholar] [CrossRef] [PubMed]
  34. Zarovni, N.; Corrado, A.; Guazzi, P.; Zocco, D.; Lari, E.; Radano, G.; Muhhina, J.; Fondelli, C.; Gavrilova, J.; Chiesi, A. Integrated isolation and quantitative analysis of exosome shuttled proteins and nucleic acids using immunocapture approaches. Methods 2015, 87, 46–58. [Google Scholar] [CrossRef]
  35. Zhang, M.; Jin, K.; Gao, L.; Zhang, Z.; Li, F.; Zhou, F.; Zhang, L. Methods and Technologies for Exosome Isolation and Characterization. Small Methods 2018, 2, 1800021. [Google Scholar] [CrossRef]
  36. Muller, L.; Hong, C.-S.; Stolz, D.B.; Watkins, S.C.; Whiteside, T.L. Isolation of biologically-active exosomes from human plasma. J. Immunol. Methods 2014, 411, 55–65. [Google Scholar] [CrossRef] [Green Version]
  37. Hiemstra, T.F.; Charles, P.D.; Gracia, T.; Hester, S.S.; Gatto, L.; Al-Lamki, R.; Floto, R.A.; Su, Y.; Skepper, J.N.; Lilley, K.S.; et al. Human Urinary Exosomes as Innate Immune Effectors. J. Am. Soc. Nephrol. 2014, 25, 2017–2027. [Google Scholar] [CrossRef] [Green Version]
  38. Livshits, M.A.; Khomyakova, E.; Evtushenko, E.; Lazarev, V.N.; Kulemin, N.; Semina, S.E.; Generozov, E.; Govorun, V.M. Isolation of exosomes by differential centrifugation: Theoretical analysis of a commonly used protocol. Sci. Rep. 2015, 5, 17319. [Google Scholar] [CrossRef] [PubMed]
  39. Baranyai, T.; Herczeg, K.; Onódi, Z.; Voszka, I.; Módos, K.; Marton, N.; Nagy, G.; Mäger, I.; Wood, M.J.; El Andaloussi, S.; et al. Isolation of Exosomes from Blood Plasma: Qualitative and Quantitative Comparison of Ultracentrifugation and Size Exclusion Chromatography Methods. PLoS ONE 2015, 10, e0145686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Ullah, M.; Kodam, S.P.; Mu, Q.; Akbar, A. Microbubbles versus Extracellular Vesicles as Therapeutic Cargo for Targeting Drug Delivery. ACS Nano 2021, 15, 3612–3620. [Google Scholar] [CrossRef]
  41. Soares Martins, T.; Catita, J.; Martins Rosa, I.; AB da Cruz e Silva, O.; Henriques, A.G. Exosome isolation from distinct biofluids using precipitation and column-based approaches. PLoS ONE 2018, 13, e0198820. [Google Scholar] [CrossRef] [Green Version]
  42. Serrano-Pertierra, E.; Oliveira-Rodríguez, M.; Rivas, M.; Oliva, P.; Villafani, J.; Navarro, A.; Blanco-López, M.C.; Cernuda-Morollón, E. Characterization of Plasma-Derived Extracellular Vesicles Isolated by Different Methods: A Comparison Study. Bioengineering 2019, 6, 8. [Google Scholar] [CrossRef] [Green Version]
  43. Salih, M.; Zietse, R.; Hoorn, E.J. Urinary extracellular vesicles and the kidney: Biomarkers and beyond. Am. J. Physiol.-Ren. Physiol. 2014, 306, F1251–F1259. [Google Scholar] [CrossRef] [Green Version]
  44. Van Deun, J.; Mestdagh, P.; Sormunen, R.; Cocquyt, V.; Vermaelen, K.; Vandesompele, J.; Bracke, M.; De Wever, O.; Hendrix, A. The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling. J. Extracell. Vesicles 2014, 3, 24858. [Google Scholar] [CrossRef] [Green Version]
  45. Anderson, N.G. An introduction to particle separations in zonal centrifuges. Natl. Cancer Inst. Monogr. 1966, 21, 9–39. [Google Scholar]
  46. Rikkert, L.G.; Engelaer, M.; Hau, C.M.; Terstappen, L.W.M.M.; Nieuwland, R.; Coumans, F.A. Rate zonal centrifugation can partially separate platelets from platelet-derived vesicles. Res. Pract. Thromb. Haemost. 2020, 4, 1053–1059. [Google Scholar] [CrossRef]
  47. Kim, D.-K.; Nishida, H.; An, S.Y.; Shetty, A.K.; Bartosh, T.J.; Prockop, D.J. Chromatographically isolated CD63+CD81+ extracellular vesicles from mesenchymal stromal cells rescue cognitive impairments after TBI. Proc. Natl. Acad. Sci. USA 2015, 113, 170–175. [Google Scholar] [CrossRef] [Green Version]
  48. Zeringer, E.; Barta, T.; Li, M.; Vlassov, A.V. Strategies for Isolation of Exosomes. Cold Spring Harb. Protoc. 2015, 2015, 319–323. [Google Scholar] [CrossRef] [Green Version]
  49. Batrakova, E.V.; Kim, M. Using exosomes, naturally-equipped nanocarriers, for drug delivery. J. Control. Release 2015, 219, 396–405. [Google Scholar] [CrossRef] [Green Version]
  50. Amarnath, S.; Foley, J.E.; Farthing, D.E.; Gress, R.E.; Laurence, A.; Eckhaus, M.A.; Métais, J.; Rose, J.J.; Hakim, F.T.; Felizardo, T.C.; et al. Bone Marrow-Derived Mesenchymal Stromal Cells Harness Purinergenic Signaling to Tolerize Human T h1 Cells In Vivo. Stem Cells 2014, 33, 1200–1212. [Google Scholar] [CrossRef] [Green Version]
  51. Carnino, J.M.; Lee, H.; Jin, Y. Isolation and characterization of extracellular vesicles from Broncho-alveolar lavage fluid: A review and comparison of different methods. Respir. Res. 2019, 20, 240. [Google Scholar] [CrossRef] [Green Version]
  52. Sunkara, V.; Woo, H.-K.; Cho, Y.-K. Emerging techniques in the isolation and characterization of extracellular vesicles and their roles in cancer diagnostics and prognostics. Analyst 2015, 141, 371–381. [Google Scholar] [CrossRef] [Green Version]
  53. Yamamoto, K.R.; Alberts, B.M.; Benzinger, R.; Lawhorne, L.; Treiber, G. Rapid bacteriophage sedimentation in the presence of polyethylene glycol and its application to large-scale virus purification. Virology 1970, 40, 734–744. [Google Scholar] [CrossRef]
  54. 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, 8545347. [Google Scholar] [CrossRef] [PubMed]
  55. Keller, S.; König, A.-K.; Marmé, F.; Runz, S.; Wolterink, S.; Koensgen, D.; Mustea, A.; Sehouli, J.; Altevogt, P. Systemic presence and tumor-growth promoting effect of ovarian carcinoma released exosomes. Cancer Lett. 2009, 278, 73–81. [Google Scholar] [CrossRef]
  56. Deregibus, M.C.; Figliolini, F.; D’Antico, S.; Manzini, P.M.; Pasquino, C.; De Lena, M.; Tetta, C.; Brizzi, M.F.; Camussi, G. Charge-based precipitation of extracellular vesicles. Int. J. Mol. Med. 2016, 38, 1359–1366. [Google Scholar] [CrossRef] [Green Version]
  57. Ullah, M. Need for Specialized Therapeutic Stem Cells Banks Equipped with Tumor Regression Enzymes and Anti-Tumor Genes. J. Biomed. Allied Res. 2020, 2, 13. [Google Scholar]
  58. Brownlee, Z.; Lynn, K.D.; Thorpe, P.E.; Schroit, A.J. A novel “salting-out” procedure for the isolation of tumor-derived exosomes. J. Immunol. Methods 2014, 407, 120–126. [Google Scholar] [CrossRef] [Green Version]
  59. Rider, M.A.; Hurwitz, S.N.; Meckes, D.G., Jr. ExtraPEG: A Polyethylene Glycol-Based Method for Enrichment of Extracellular Vesicles. Sci. Rep. 2016, 6, 23978. [Google Scholar] [CrossRef] [PubMed]
  60. Crescitelli, R.; Lässer, C.; Lötvall, J. Isolation and characterization of extracellular vesicle subpopulations from tissues. Nat. Protoc. 2021, 16, 1548–1580. [Google Scholar] [CrossRef] [PubMed]
  61. Liga, A.; Vliegenthart, A.D.B.; Oosthuyzen, W.; Dear, J.W.; Kersaudy-Kerhoas, M. Exosome isolation: A microfluidic road-map. Lab Chip 2015, 15, 2388–2394. [Google Scholar] [CrossRef] [Green Version]
  62. Yang, D.; Zhang, W.; Zhang, H.; Zhang, F.; Chen, L.; Ma, L.; Larcher, L.M.; Chen, S.; Liu, N.; Zhao, Q. Progress, opportunity, and perspective on exosome isolation-efforts for efficient exosome-based theranostics. Theranostics 2020, 10, 3684. [Google Scholar] [CrossRef]
  63. Lobb, R.J.; Becker, M.; Wen, S.W.; Wong, C.S.F.; Wiegmans, A.P.; Leimgruber, A.; Möller, A. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J. Extracell. Vesicles 2015, 4, 27031. [Google Scholar] [CrossRef] [PubMed]
  64. Jeppesen, D.; Hvam, M.L.; Primdahl-Bengtson, B.; Boysen, A.T.; Whitehead, B.; Dyrskjøt, L.; Ørntoft, T.F.; Howard, K.A.; Ostenfeld, M.S. Comparative analysis of discrete exosome fractions obtained by differential centrifugation. J. Extracell. Vesicles 2014, 3, 25011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Liu, F.; Vermesh, O.; Mani, V.; Ge, T.J.; Madsen, S.J.; Sabour, A.; Hsu, E.-C.; Gowrishankar, G.; Kanada, M.; Jokerst, J.V.; et al. The Exosome Total Isolation Chip. ACS Nano 2017, 11, 10712–10723. [Google Scholar] [CrossRef] [PubMed]
  66. Kim, K.; Park, J.; Jung, J.-H.; Lee, R.; Park, J.-H.; Yuk, J.M.; Hwang, H.; Yeon, J.H. Cyclic tangential flow filtration system for isolation of extracellular vesicles. APL Bioeng. 2021, 5, 016103. [Google Scholar] [CrossRef]
  67. Soda, N.; Rehm, B.H.A.; Sonar, P.; Nguyen, N.-T.; Shiddiky, M.J.A. Advanced liquid biopsy technologies for circulating biomarker detection. J. Mater. Chem. B 2019, 7, 6670–6704. [Google Scholar] [CrossRef]
  68. Peterson, M.F.; Otoc, N.; Sethi, J.K.; Gupta, A.; Antes, T.J. Integrated systems for exosome investigation. Methods 2015, 87, 31–45. [Google Scholar] [CrossRef]
  69. Gheinani, A.H.; Vögeli, M.; Baumgartner, U.; Vassella, E.; Draeger, A.; Burkhard, F.C.; Monastyrskaya, K. Improved isolation strategies to increase the yield and purity of human urinary exosomes for biomarker discovery. Sci. Rep. 2018, 8, 3945. [Google Scholar] [CrossRef]
  70. Gámez-Valero, A.; Monguió-Tortajada, M.; Carreras-Planella, L.; Franquesa, M.; Beyer, K.; Borràs, F.E. Size-Exclusion Chromatography-based isolation minimally alters Extracellular Vesicles’ characteristics compared to precipitating agents. Sci. Rep. 2016, 6, srep33641. [Google Scholar] [CrossRef] [Green Version]
  71. Navajas, R.; Corrales, F.J.; Paradela, A. Serum Exosome Isolation by Size-Exclusion Chromatography for the Discovery and Validation of Preeclampsia-Associated Biomarkers. In Proteomics for Biomarker Discovery; Humana Press: New York, NY, USA, 2019; pp. 39–50. [Google Scholar] [CrossRef]
  72. Stranska, R.; Gysbrechts, L.; Wouters, J.; Vermeersch, P.; Bloch, K.; Dierickx, D.; Andrei, G.; Snoeck, R. Comparison of membrane affinity-based method with size-exclusion chromatography for isolation of exosome-like vesicles from human plasma. J. Transl. Med. 2018, 16, 1. [Google Scholar] [CrossRef] [PubMed]
  73. GH, L.; CR, R. The separation of substances on the basis of their molecular weights, using columns of starch and water. Biochem. J. 1955, 60, xxxiv. [Google Scholar]
  74. Ruysschaert, T.; Marque, A.; Duteyrat, J.-L.; Lesieur, S.; Winterhalter, M.; Fournier, D. Liposome retention in size exclusion chromatography. BMC Biotechnol. 2005, 5, 11. [Google Scholar] [CrossRef] [Green Version]
  75. Vogel, R.; Coumans, F.A.W.; Maltesen, R.G.; Böing, A.N.; Bonnington, K.E.; Broekman, M.L.; Broom, M.F.; Buzás, E.I.; Christiansen, G.; Hajji, N.; et al. A standardized method to determine the concentration of extracellular vesicles using tunable resistive pulse sensing. J. Extracell. Vesicles 2016, 5, 31242. [Google Scholar] [CrossRef]
  76. Kang, D.; Oh, S.; Ahn, S.-M.; Lee, B.-H.; Moon, M.H. Proteomic Analysis of Exosomes from Human Neural Stem Cells by Flow Field-Flow Fractionation and Nanoflow Liquid Chromatography−Tandem Mass Spectrometry. J. Proteome Res. 2008, 7, 3475–3480. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, H.; Lyden, D. Asymmetric-flow field-flow fractionation technology for exomere and small extracellular vesicle separation and characterization. Nat. Protoc. 2019, 14, 1027–1053. [Google Scholar] [CrossRef]
  78. Musante, L.; Tataruch, D.; Gu, D.; Martin, A.B.; Calzaferri, G.; Aherne, S.; Holthofer, H. A Simplified Method to Recover Urinary Vesicles for Clinical Applications and Sample Banking. Sci. Rep. 2014, 4, 7532. [Google Scholar] [CrossRef] [PubMed]
  79. Musante, L.; Tataruch, D.E.; Holthofer, H. Use and Isolation of Urinary Exosomes as Biomarkers for Diabetic Nephropathy. Front. Endocrinol. 2014, 5, 149. [Google Scholar] [CrossRef] [Green Version]
  80. Tauro, B.J.; Greening, D.W.; Mathias, R.A.; Ji, H.; Mathivanan, S.; Scott, A.M.; Simpson, R.J. Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes. Methods 2012, 56, 293–304. [Google Scholar] [CrossRef]
  81. Mathivanan, S.; Lim, J.W.E.; Tauro, B.J.; Ji, H.; Moritz, R.L.; Simpson, R.J. Proteomics Analysis of A33 Immunoaffinity-purified Exosomes Released from the Human Colon Tumor Cell Line LIM1215 Reveals a Tissue-specific Protein Signature. Mol. Cell. Proteom. 2010, 9, 197–208. [Google Scholar] [CrossRef] [Green Version]
  82. Taylor, D.D.; Gercel-Taylor, C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 2010, 110, 13–21, Erratum in 2010, 116, 153. [Google Scholar] [CrossRef] [PubMed]
  83. Cui, Y.; Gao, J.; He, Y.; Jiang, L. Plant extracellular vesicles. Protoplasma 2019, 257, 3–12. [Google Scholar] [CrossRef] [PubMed]
  84. Hosseini, S.; Vázquez-Villegas, P.; Rito-Palomares, M.; Martinez-Chapa, S.O. Enzyme-Linked Immunosorbent Assay (ELISA): From A to Z; Springer: Singapore, 2018. [Google Scholar]
  85. Hong, C.S.; Muller, L.; Boyiadzis, M.; Whiteside, T.L. Isolation and Characterization of CD34+ Blast-Derived Exosomes in Acute Myeloid Leukemia. PLoS ONE 2014, 9, e103310. [Google Scholar] [CrossRef] [Green Version]
  86. Shao, H.; Im, H.; Castro, C.M.; Breakefield, X.; Weissleder, R.; Lee, H. New Technologies for Analysis of Extracellular Vesicles. Chem. Rev. 2018, 118, 1917–1950. [Google Scholar] [CrossRef]
  87. Zhang, J.; Nguyen, L.T.H.; Hickey, R.; Walters, N.; Wang, X.; Kwak, K.J.; Lee, L.J.; Palmer, A.F.; Reátegui, E. Immunomagnetic sequential ultrafiltration (iSUF) platform for enrichment and purification of extracellular vesicles from biofluids. Sci. Rep. 2021, 11, 8034. [Google Scholar] [CrossRef]
  88. Shih, C.-L.; Chong, K.-Y.; Hsu, S.-C.; Chien, H.-J.; Ma, C.-T.; Chang, J.W.-C.; Yu, C.-J.; Chiou, C.-C. Development of a magnetic bead-based method for the collection of circulating extracellular vesicles. New Biotechnol. 2016, 33, 116–122. [Google Scholar] [CrossRef] [PubMed]
  89. Lee, K.; Shao, H.; Weissleder, R.; Lee, H. Acoustic Purification of Extracellular Microvesicles. ACS Nano 2015, 9, 2321–2327. [Google Scholar] [CrossRef] [Green Version]
  90. Davies, R.T.; Kim, J.; Jang, S.C.; Choi, E.-J.; Gho, Y.S.; Park, J. Microfluidic filtration system to isolate extracellular vesicles from blood. Lab Chip 2012, 12, 5202–5210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Guo, S.-C.; Tao, S.-C.; Dawn, H. Microfluidics-based on-a-chip systems for isolating and analysing extracellular vesicles. J. Extracell. Vesicles 2018, 7, 1508271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Liu, C.; Guo, J.; Tian, F.; Yang, N.; Yan, F.; Ding, Y.; Wei, J.; Hu, G.; Nie, G.; Sun, J. Field-Free Isolation of Exosomes from Extracellular Vesicles by Microfluidic Viscoelastic Flows. ACS Nano 2017, 11, 6968–6976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Wu, M.; Chen, C.; Wang, Z.; Bachman, H.; Ouyang, Y.; Huang, P.-H.; Sadovsky, Y.; Huang, T.J. Separating extracellular vesicles and lipoproteins via acoustofluidics. Lab Chip 2019, 19, 1174–1182. [Google Scholar] [CrossRef]
  94. Zhao, Z.; Yang, Y.; Zeng, Y.; He, M. A microfluidic ExoSearch chip for multiplexed exosome detection towards blood-based ovarian cancer diagnosis. Lab Chip 2016, 16, 489–496. [Google Scholar] [CrossRef] [Green Version]
  95. Jiang, C.; Fu, Y.; Liu, G.; Shu, B.; Davis, J.; Tofaris, G.K. Multiplexed Profiling of Extracellular Vesicles for Biomarker Development. Nanomicro Lett. 2021, 14, 3. [Google Scholar] [CrossRef]
  96. Talebjedi, B.; Tasnim, N.; Hoorfar, M.; Mastromonaco, G.F.; De Almeida Monteiro Melo Ferraz, M. Exploiting Microfluidics for Extracellular Vesicle Isolation and Characterization: Potential Use for Standardized Embryo Quality Assessment. Front. Vet. Sci. 2021, 7, 1139. [Google Scholar] [CrossRef]
  97. Kanwar, S.S.; Dunlay, C.J.; Simeone, D.M.; Nagrath, S. Microfluidic device (ExoChip) for on-chip isolation, quantification and characterization of circulating exosomes. Lab Chip 2014, 14, 1891–1900. [Google Scholar] [CrossRef]
  98. Hisey, C.L.; Dorayappan, K.D.P.; Cohn, D.E.; Selvendiran, K.; Hansford, D.J. Microfluidic affinity separation chip for selective capture and release of label-free ovarian cancer exosomes. Lab Chip 2018, 18, 3144–3153. [Google Scholar] [CrossRef] [PubMed]
  99. Guerreiro, E.M.; Vestad, B.; Steffensen, L.A.; Aass, H.C.D.; Saeed, M.; Øvstebø, R.; Costea, D.-E.; Galtung, H.K.; Søland, T.M. Efficient extracellular vesicle isolation by combining cell media modifications, ultrafiltration, and size-exclusion chromatography. PLoS ONE 2018, 13, e0204276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Deville, S.; Berckmans, P.; Van Hoof, R.; Lambrichts, I.; Salvati, A.; Nelissen, I. Comparison of extracellular vesicle isolation and storage methods using high-sensitivity flow cytometry. PLoS ONE 2021, 16, e0245835. [Google Scholar] [CrossRef] [PubMed]
  101. Zhang, Z.; Wang, C.; Li, T.; Liu, Z.; Li, L. Comparison of ultracentrifugation and density gradient separation methods for isolating Tca8113 human tongue cancer cell line-derived exosomes. Oncol. Lett. 2014, 8, 1701–1706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Heinemann, M.L.; Ilmer, M.; Silva, L.P.; Hawke, D.H.; Recio, A.; Vorontsova, M.A.; Alt, E.; Vykoukal, J. Benchtop isolation and characterization of functional exosomes by sequential filtration. J. Chromatogr. A 2014, 1371, 125–135. [Google Scholar] [CrossRef] [PubMed]
  103. Mao, W.; Wen, Y.; Lei, H.; Lu, R.; Wang, S.; Wang, Y.; Chen, R.; Gu, Y.; Zhu, L.; Abhange, K.K.; et al. Isolation and Retrieval of Extracellular Vesicles for Liquid Biopsy of Malignant Ground-Glass Opacity. Anal. Chem. 2019, 91, 13729–13736. [Google Scholar] [CrossRef]
  104. Knol, J.C.; de Reus, I.; Schelfhorst, T.; Beekhof, R.; de Wit, M.; Piersma, S.R.; Pham, T.V.; Smit, E.F.; Verheul, H.M.; Jiménez, C.R. Peptide-mediated ‘miniprep’ isolation of extracellular vesicles is suitable for high-throughput proteomics. EuPA Open Proteom. 2016, 11, 11–15. [Google Scholar] [CrossRef] [Green Version]
  105. Bathini, S.; Pakkiriswami, S.; Ouellette, R.J.; Ghosh, A.; Packirisamy, M. Magnetic particle based liquid biopsy chip for isolation of extracellular vesicles and characterization by gene amplification. Biosens. Bioelectron. 2021, 194, 113585. [Google Scholar] [CrossRef]
  106. He, M.; Crow, J.; Roth, M.; Zeng, Y.; Godwin, A.K. Integrated immunoisolation and protein analysis of circulating exosomes using microfluidic technology. Lab Chip 2014, 14, 3773–3780. [Google Scholar] [CrossRef] [Green Version]
  107. Sancho-Albero, M.; Sebastián, V.; Sesé, J.; Pazo-Cid, R.; Mendoza, G.; Arruebo, M.; Martín-Duque, P.; Santamaría, J. Isolation of exosomes from whole blood by a new microfluidic device: Proof of concept application in the diagnosis and monitoring of pancreatic cancer. J. Nanobiotechnol. 2020, 18, 150. [Google Scholar] [CrossRef]
  108. Liang, L.-G.; Kong, M.-Q.; Zhou, S.; Sheng, Y.-F.; Wang, P.; Yu, T.; Inci, F.; Kuo, W.P.; Li, L.-J.; Demirci, U.; et al. An integrated double-filtration microfluidic device for isolation, enrichment and quantification of urinary extracellular vesicles for detection of bladder cancer. Sci. Rep. 2017, 7, srep46224. [Google Scholar] [CrossRef] [Green Version]
  109. Wu, M.; Ouyang, Y.; Wang, Z.; Zhang, R.; Huang, P.-H.; Chen, C.; Li, H.; Li, P.; Quinn, D.; Dao, M.; et al. Isolation of exosomes from whole blood by integrating acoustics and microfluidics. Proc. Natl. Acad. Sci. USA 2017, 114, 10584–10589. [Google Scholar] [CrossRef] [Green Version]
  110. Casadei, L.; Choudhury, A.; Sarchet, P.; Sundaram, P.M.; Lopez, G.; Braggio, D.; Balakirsky, G.; Pollock, R.; Prakash, S. Cross-flow microfiltration for isolation, selective capture and release of liposarcoma extracellular vesicles. J. Extracell. Vesicles 2021, 10, e12062. [Google Scholar] [CrossRef] [PubMed]
  111. Yeo, J.C.; Kenry; Zhao, Z.; Zhang, P.; Wang, Z.; Lim, C.T. Label-free extraction of extracellular vesicles using centrifugal microfluidics. Biomicrofluidics 2018, 12, 024103. [Google Scholar] [CrossRef] [PubMed]
  112. Schey, K.L.; Luther, J.; Rose, K.L. Proteomics characterization of exosome cargo. Methods 2015, 87, 75–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Rikkert, L.G.; Beekman, P.; Caro, J.; Coumans, F.A.W.; Enciso-Martinez, A.; Jenster, G.; Le Gac, S.; Lee, W.; Van Leeuwen, T.G.; Loozen, G.B.; et al. Cancer-ID: Toward Identification of Cancer by Tumor-Derived Extracellular Vesicles in Blood. Front. Oncol. 2020, 10, 608. [Google Scholar] [CrossRef]
  114. Julich, H.; Willms, A.; Lukacs-Kornek, V.; Kornek, M. Extracellular vesicle profiling and their use as potential disease specific biomarker. Front. Immunol. 2014, 5, 413. [Google Scholar] [CrossRef] [Green Version]
  115. Castellani, C.; Burrello, J.; Fedrigo, M.; Burrello, A.; Bolis, S.; Di Silvestre, D.; Tona, F.; Bottio, T.; Biemmi, V.; Toscano, G.; et al. Circulating extracellular vesicles as non-invasive biomarker of rejection in heart transplant. J. Heart Lung Transplant. 2020, 39, 1136–1148. [Google Scholar] [CrossRef]
  116. Eren, E.; Hunt, J.F.; Shardell, M.; Chawla, S.; Tran, J.; Gu, J.; Vogt, N.M.; Johnson, S.C.; Bendlin, B.B.; Kapogiannis, D. Extracellular vesicle biomarkers of Alzheimer’s disease associated with sub-clinical cognitive decline in late middle age. Alzheimer’s Dement. 2020, 16, 1293–1304. [Google Scholar] [CrossRef]
  117. Fiandaca, M.S.; Kapogiannis, D.; Mapstone, M.; Boxer, A.; Eitan, E.; Schwartz, J.B.; Abner, E.L.; Petersen, R.C.; Federoff, H.J.; Miller, B.L. Identification of preclinical Alzheimer’s disease by a profile of pathogenic proteins in neurally derived blood exosomes: A case-control study. Alzheimer’s Dement. 2015, 11, 600–607.e1. [Google Scholar]
  118. Goetzl, E.J.; Boxer, A.; Schwartz, J.B.; Abner, E.L.; Petersen, R.C.; Miller, B.L.; Kapogiannis, D. Altered lysosomal proteins in neural-derived plasma exosomes in preclinical Alzheimer disease. Neurology 2015, 85, 40–47. [Google Scholar] [CrossRef] [Green Version]
  119. Goetzl, E.J.; Kapogiannis, D.; Schwartz, J.B.; Lobach, I.V.; Goetzl, L.; Abner, E.L.; Jicha, G.A.; Karydas, A.M.; Boxer, A.; Miller, B.L. Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer’s disease. FASEB J. 2016, 30, 4141–4148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Lee, S.; Mankhong, S.; Kang, J.-H. Extracellular Vesicle as a Source of Alzheimer’s Biomarkers: Opportunities and Challenges. Int. J. Mol. Sci. 2019, 20, 1728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Ogata-Kawata, H.; Izumiya, M.; Kurioka, D.; Honma, Y.; Yamada, Y.; Furuta, K.; Gunji, T.; Ohta, H.; Okamoto, H.; Sonoda, H.; et al. Circulating Exosomal microRNAs as Biomarkers of Colon Cancer. PLoS ONE 2014, 9, e92921. [Google Scholar] [CrossRef] [PubMed]
  122. Mohammadi, S.; Yousefi, F.; Shabaninejad, Z.; Movahedpour, A.; Mahjoubin-Tehran, M.; Shafiee, A.; Moradizarmehri, S.; Hajighadimi, S.; Savardashtaki, A.; Mirzaei, H. Exosomes and cancer: From oncogenic roles to therapeutic applications. IUBMB Life 2019, 72, 724–748. [Google Scholar] [CrossRef]
  123. Ullah, M.; Akbar, A. Clinical Relevance of RNA Editing to Early Detection of Cancer in Human. Int. J. Stem Cell Res. Ther. 2020, 7, 66. [Google Scholar]
  124. Corcoran, C.; Friel, A.; Duffy, M.J.; Crown, J.; O’Driscoll, L. Intracellular and Extracellular MicroRNAs in Breast Cancer. Clin. Chem. 2011, 57, 18–32. [Google Scholar] [CrossRef] [Green Version]
  125. Işın, M.; Uysaler, E.; Özgür, E.; Köseoğlu, H.; Şanlı, Ö.; Yücel, Ö.B.; Gezer, U.; Dalay, N. Exosomal lncRNA-p21 levels may help to distinguish prostate cancer from benign disease. Front. Genet. 2015, 6, 168. [Google Scholar]
  126. Ullah, M.; Akbar, A.; Yannarelli, G. Clinical Applications of RNA Editing Technology for the Early Detection of Cancer and Future Directions. Technol. Cancer Res. Treat. 2020, 19, 1533033820964194. [Google Scholar] [CrossRef]
  127. Abrahams, V.M.; Chavez, S.; Guller, S.; Mor, G. First trimester trophoblast cells secrete Fas ligand which induces immune cell apoptosis. Mol. Hum. Reprod. 2004, 10, 55–63. [Google Scholar] [CrossRef] [PubMed]
  128. Kim, J.W.; Wieckowski, E.; Taylor, D.D.; Reichert, T.E.; Watkins, S.; Whiteside, T.L. Fas ligand-positive membranous vesicles isolated from sera of patients with oral cancer induce apoptosis of activated T lymphocytes. Clin. Cancer Res. 2005, 11, 1010–1020. [Google Scholar]
  129. Pisitkun, T.; Johnstone, R.; Knepper, M.A. Discovery of Urinary Biomarkers. Mol. Cell. Proteom. 2006, 5, 1760–1771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Schorey, J.S.; Bhatnagar, S. Exosome Function: From Tumor Immunology to Pathogen Biology. Traffic 2008, 9, 871–881. [Google Scholar] [CrossRef] [Green Version]
  131. Lu, M.; Yuan, S.; Li, S.; Li, L.; Liu, M.; Wan, S. The Exosome-Derived Biomarker in Atherosclerosis and Its Clinical Application. J. Cardiovasc. Transl. Res. 2018, 12, 68–74. [Google Scholar] [CrossRef] [PubMed]
  132. Cervio, E.; Barile, L.; Moccetti, T.; Vassalli, G. Exosomes for Intramyocardial Intercellular Communication. Stem Cells Int. 2015, 2015, 482171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Niu, C.; Wang, X.; Zhao, M.; Cai, T.; Liu, P.; Li, J.; Willard, B.; Zu, L.; Zhou, E.; Li, Y.; et al. Macrophage Foam Cell–Derived Extracellular Vesicles Promote Vascular Smooth Muscle Cell Migration and Adhesion. J. Am. Heart Assoc. 2016, 5, e004099. [Google Scholar] [CrossRef] [Green Version]
  134. Wang, C.; Li, Z.; Liu, Y.; Yuan, L. Exosomes in atherosclerosis: Performers, bystanders, biomarkers, and therapeutic targets. Theranostics 2021, 11, 3996–4010. [Google Scholar] [CrossRef]
  135. Goetzl, E.J.; Schwartz, J.B.; Mustapic, M.; Lobach, I.V.; Daneman, R.; Abner, E.L.; Jicha, G.A. Altered cargo proteins of human plasma endothelial cell–derived exosomes in atherosclerotic cerebrovascular disease. FASEB J. 2017, 31, 3689–3694. [Google Scholar] [CrossRef] [Green Version]
  136. Li, C.; Pei, F.; Zhu, X.; Duan, D.D.; Zeng, C. Circulating microRNAs as novel and sensitive biomarkers of acute myocardial Infarction. Clin. Biochem. 2012, 45, 727–732. [Google Scholar] [CrossRef] [Green Version]
  137. Kuwabara, Y.; Ono, K.; Horie, T.; Nishi, H.; Watanabe, S.; Kinoshita, M.; Baba, O.; Chujo, Y.; Nagao, K.; Kojima, Y.; et al. MicroRNA-1 and MicroRNA-133a Elevations in Serum of Patients with Cardiovascular Disease Indicate the Existence of Myocardial Damage. Am. Heart Assoc. 2011, 124, A11460. [Google Scholar]
  138. Zamani, P.; Fereydouni, N.; Butler, A.E.; Navashenaq, J.G.; Sahebkar, A. The therapeutic and diagnostic role of exosomes in cardiovascular diseases. Trends Cardiovasc. Med. 2018, 29, 313–323. [Google Scholar] [CrossRef]
  139. Deddens, J.C.; Vrijsen, K.R.; Colijn, J.M.; Oerlemans, M.I.; Metz, C.H.G.; Van Der Vlist, E.J.; Nolte-’t Hoen, E.N.M.; den Ouden, K.; Jansen Of Lorkeers, S.J.; Van Der Spoel, T.I.G.; et al. Circulating Extracellular Vesicles Contain miRNAs and are Released as Early Biomarkers for Cardiac Injury. J. Cardiovasc. Transl. Res. 2016, 9, 291–301. [Google Scholar] [CrossRef] [Green Version]
  140. Chen, Y.; Song, Y.; Huang, J.; Qu, M.; Zhang, Y.; Geng, J.; Zhang, Z.; Liu, J.; Yang, G.-Y. Increased Circulating Exosomal miRNA-223 Is Associated with Acute Ischemic Stroke. Front. Neurol. 2017, 8, 57. [Google Scholar] [CrossRef] [Green Version]
  141. Matsumoto, S.; Sakata, Y.; Suna, S.; Nakatani, D.; Usami, M.; Hara, M.; Kitamura, T.; Hamasaki, T.; Nanto, S.; Kawahara, Y.; et al. Circulating p53-Responsive MicroRNAs Are Predictive Indicators of Heart Failure After Acute Myocardial Infarction. Circ. Res. 2013, 113, 322–326. [Google Scholar] [CrossRef] [Green Version]
  142. Goren, Y.; Kushnir, M.; Zafrir, B.; Tabak, S.; Lewis, B.S.; Amir, O. Serum levels of microRNAs in patients with heart failure. Eur. J. Heart Fail. 2012, 14, 147–154. [Google Scholar] [CrossRef]
  143. Halkein, J.; Tabruyn, S.P.; Ricke-Hoch, M.; Haghikia, A.; Nguyen, N.-Q.; Scherr, M.; Castermans, K.; Malvaux, L.; Lambert, V.; Thiry, M.; et al. MicroRNA-146a is a therapeutic target and biomarker for peripartum cardiomyopathy. J. Clin. Investig. 2013, 123, 2143–2154. [Google Scholar] [CrossRef] [PubMed]
  144. Yu, X.; Deng, L.; Wang, D.; Li, N.; Chen, X.; Cheng, X.; Yuan, J.; Gao, X.; Liao, M.; Wang, M.; et al. Mechanism of TNF-α autocrine effects in hypoxic cardiomyocytes: Initiated by hypoxia inducible factor 1α, presented by exosomes. J. Mol. Cell. Cardiol. 2012, 53, 848–857. [Google Scholar] [CrossRef] [PubMed]
  145. Pironti, G.; Strachan, R.T.; Abraham, D.; Yu, S.M.-W.; Chen, M.; Chen, W.; Hanada, K.; Mao, L.; Watson, L.J.; Rockman, H.A. Circulating Exosomes Induced by Cardiac Pressure Overload Contain Functional Angiotensin II Type 1 Receptors. Circulation 2015, 131, 2120–2130. [Google Scholar] [CrossRef] [PubMed]
  146. de Hoog, V.C.; Timmers, L.; Schoneveld, A.H.; Wang, J.W.; van de Weg, S.M.; Sze, S.K.; van Keulen, J.K.; Hoes, A.W.; den Ruijter, H.M.; de Kleijn, D.P.; et al. Serum extracellular vesicle protein levels are associated with acute coronary syndrome. Eur. Heart J. Acute Cardiovasc. Care 2013, 2, 53–60. [Google Scholar] [CrossRef] [Green Version]
  147. Cheow, E.S.H.; Cheng, W.C.; Lee, C.N.; de Kleijn, D.; Sorokin, V.; Sze, S.K. Plasma-derived Extracellular Vesicles Contain Predictive Biomarkers and Potential Therapeutic Targets for Myocardial Ischemic (MI) Injury. Mol. Cell. Proteom. 2016, 15, 2628–2640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Mizutani, K.; Kawakami, K.; Horie, K.; Fujita, Y.; Kameyama, K.; Kato, T.; Nakane, K.; Tsuchiya, T.; Yasuda, M.; Masunaga, K.; et al. Urinary exosome as a potential biomarker for urinary tract infection. Cell. Microbiol. 2019, 21, e13020. [Google Scholar] [CrossRef] [PubMed]
  149. Zhang, C.; Yang, X.; Qi, Q.; Gao, Y.; Wei, Q.; Han, S. lncRNA-HEIH in serum and exosomes as a potential biomarker in the HCV-related hepatocellular carcinoma. Cancer Biomark. 2018, 21, 651–659. [Google Scholar] [CrossRef]
  150. Sun, B.; Dalvi, P.; Abadjian, L.; Tang, N.; Pulliam, L. Blood neuron-derived exosomes as biomarkers of cognitive impairment in HIV. AIDS 2017, 31, F9–F17. [Google Scholar] [CrossRef]
  151. Ullah, M. Novel Coronavirus (COVID-19) Treatment Options. Biomed. J. Sci. Tech. Res. 2020, 27, 20872–20874. [Google Scholar] [CrossRef]
  152. Donoso-Quezada, J.; Ayala-Mar, S.; González-Valdez, J. State-of-the-art exosome loading and functionalization techniques for enhanced therapeutics: A review. Crit. Rev. Biotechnol. 2020, 40, 804–820. [Google Scholar] [CrossRef]
  153. Li, S.-P.; Lin, Z.-X.; Jiang, X.-Y.; Yu, X.-Y. Exosomal cargo-loading and synthetic exosome-mimics as potential therapeutic tools. Acta Pharmacol. Sin. 2018, 39, 542–551. [Google Scholar] [CrossRef] [Green Version]
  154. Keerthikumar, S.; Chisanga, D.; Ariyaratne, D.; Al Saffar, H.; Anand, S.; Zhao, K.; Samuel, M.; Pathan, M.; Jois, M.; Chilamkurti, N.; et al. ExoCarta: A Web-Based Compendium of Exosomal Cargo. J. Mol. Biol. 2015, 428, 688–692. [Google Scholar] [CrossRef] [Green Version]
  155. Fu, S.; Wang, Y.; Xia, X.; Zheng, J.C. Exosome engineering: Current progress in cargo loading and targeted delivery. NanoImpact 2020, 20, 100261. [Google Scholar] [CrossRef]
  156. Wang, F.; Zuroske, T.; Watts, J.K. RNA therapeutics on the rise. Nat. Rev. Drug Discov. 2020, 19, 441–442. [Google Scholar] [CrossRef]
  157. Dowdy, S.F. Overcoming cellular barriers for RNA therapeutics. Nat. Biotechnol. 2017, 35, 222–229. [Google Scholar] [CrossRef]
  158. Patil, S.D.; Rhodes, D.G.; Burgess, D.J. DNA-based therapeutics and DNA delivery systems: A comprehensive review. AAPS J. 2005, 7, E61–E77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Lässer, C. Exosomal RNA as biomarkers and the therapeutic potential of exosome vectors. Expert Opin. Biol. Ther. 2012, 12, S189–S197. [Google Scholar] [CrossRef]
  160. Chen, Y.; Gao, D.-Y.; Huang, L. In vivo delivery of miRNAs for cancer therapy: Challenges and strategies. Adv. Drug Deliv. Rev. 2014, 81, 128–141. [Google Scholar] [CrossRef] [Green Version]
  161. Garzon, R.; Marcucci, G.; Croce, C.M. Targeting microRNAs in cancer: Rationale, strategies and challenges. Nat. Rev. Drug Discov. 2010, 9, 775–789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Zhang, Y.; Wang, Z.; Gemeinhart, R.A. Progress in microRNA delivery. J. Control. Release 2013, 172, 962–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Aryani, A.; Denecke, B. Exosomes as a Nanodelivery System: A Key to the Future of Neuromedicine? Mol Neurobiol. 2016, 53, 818–834. [Google Scholar] [CrossRef] [Green Version]
  164. Van den Boorn, J.G.; Schlee, M.; Coch, C.; Hartmann, G. SiRNA delivery with exosome nanoparticles. Nat. Biotechnol. 2011, 29, 325–326. [Google Scholar] [CrossRef] [PubMed]
  165. Kanada, M.; Bachmann, M.; Hardy, J.W.; Frimannson, D.O.; Bronsart, L.; Wang, A.; Sylvester, M.D.; Schmidt, T.L.; Kaspar, R.L.; Butte, M.J.; et al. Differential fates of biomolecules delivered to target cells via extracellular vesicles. Proc. Natl. Acad. Sci. USA 2015, 112, E1433–E1442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Ohno, S.-I.; Takanashi, M.; Sudo, K.; Ueda, S.; Ishikawa, A.; Matsuyama, N.; Fujita, K.; Mizutani, T.; Ohgi, T.; Ochiya, T.; et al. Systemically Injected Exosomes Targeted to EGFR Deliver Antitumor MicroRNA to Breast Cancer Cells. Mol. Ther. 2013, 21, 185–191. [Google Scholar] [CrossRef] [Green Version]
  167. Katakowski, M.; Buller, B.; Zheng, X.; Lu, Y.; Rogers, T.; Osobamiro, O.; Shu, W.; Jiang, F.; Chopp, M. Exosomes from marrow stromal cells expressing miR-146b inhibit glioma growth. Cancer Lett. 2013, 335, 201–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Shimbo, K.; Miyaki, S.; Ishitobi, H.; Kato, Y.; Kubo, T.; Shimose, S.; Ochi, M. Exosome-formed synthetic microRNA-143 is transferred to osteosarcoma cells and inhibits their migration. Biochem. Biophys. Res. Commun. 2014, 445, 381–387. [Google Scholar] [CrossRef]
  169. Lou, G.; Song, X.; Yang, F.; Wu, S.; Wang, J.; Chen, Z.; Liu, Y. Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. J. Hematol. Oncol. 2015, 8, 122. [Google Scholar] [CrossRef] [Green Version]
  170. O’Brien, K.; Lowry, M.C.; Corcoran, C.; Martinez, V.G.; Daly, M.; Rani, S.; Gallagher, W.; Radomski, M.W.; MacLeod, R.A.; O’Driscoll, L. miR-134 in extracellular vesicles reduces triple-negative breast cancer aggression and increases drug sensitivity. Oncotarget 2015, 6, 32774–32789. [Google Scholar] [CrossRef] [Green Version]
  171. Munoz, J.L.; Bliss, S.A.; Greco, S.J.; Ramkissoon, S.H.; Ligon, K.L.; Rameshwar, P. Delivery of functional anti-miR-9 by mesenchymal stem cell–derived exosomes to glioblastoma multiforme cells conferred chemosensitivity. Mol. Ther.-Nucleic Acids 2013, 2, e126. [Google Scholar] [CrossRef]
  172. Pusic, A.D.; Pusic, K.M.; Clayton, B.; Kraig, R.P. IFNγ-stimulated dendritic cell exosomes as a potential therapeutic for remyelination. J. Neuroimmunol. 2013, 266, 12–23. [Google Scholar] [CrossRef] [Green Version]
  173. Xin, H.; Katakowski, M.; Wang, F.; Qian, J.-Y.; Liu, X.S.; Ali, M.M.; Buller, B.; Zhang, Z.G.; Chopp, M. MicroRNA-17–92 Cluster in Exosomes Enhance Neuroplasticity and Functional Recovery After Stroke in Rats. Stroke 2017, 48, 747–753. [Google Scholar] [CrossRef]
  174. Xin, H.; Li, Y.; Buller, B.; Katakowski, M.; Zhang, Y.; Wang, X.; Shang, X.; Zhang, Z.G.; Chopp, M. Exosome-Mediated Transfer of miR-133b from Multipotent Mesenchymal Stromal Cells to Neural Cells Contributes to Neurite Outgrowth. Stem Cells 2012, 30, 1556–1564. [Google Scholar] [CrossRef] [Green Version]
  175. Yang, J.; Zhang, X.; Chen, X.; Wang, L.; Yang, G. Exosome Mediated Delivery of miR-124 Promotes Neurogenesis after Ischemia. Mol. Ther.-Nucleic Acids 2017, 7, 278–287. [Google Scholar] [CrossRef] [Green Version]
  176. Jiang, M.; Wang, H.; Jin, M.; Yang, X.; Ji, H.; Jiang, Y.; Zhang, H.; Wu, F.; Wu, G.; Lai, X.; et al. Exosomes from MiR-30d-5p-ADSCs Reverse Acute Ischemic Stroke-Induced, Autophagy-Mediated Brain Injury by Promoting M2 Microglial/Macrophage Polarization. Cell. Physiol. Biochem. 2018, 47, 864–878. [Google Scholar] [CrossRef] [PubMed]
  177. Gartz, M.; Strande, J.L. Examining the Paracrine Effects of Exosomes in Cardiovascular Disease and Repair. J. Am. Heart Assoc. 2018, 7, e007954. [Google Scholar] [CrossRef] [Green Version]
  178. Yu, B.; Kim, H.W.; Gong, M.; Wang, J.; Millard, R.W.; Wang, Y.; Ashraf, M.; Xu, M. Exosomes secreted from GATA-4 overexpressing mesenchymal stem cells serve as a reservoir of anti-apoptotic microRNAs for cardioprotection. Int. J. Cardiol. 2014, 182, 349–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Ibrahim, A.G.-E.; Cheng, K.; Marbán, E. Exosomes as Critical Agents of Cardiac Regeneration Triggered by Cell Therapy. Stem Cell Rep. 2014, 2, 606–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Xiao, J.; Pan, Y.; Li, X.H.; Yang, X.Y.; Feng, Y.L.; Tan, H.H.; Jiang, L.; Feng, J.; Yu, X.Y. Cardiac progenitor cell-derived exosomes prevent cardiomyocytes apoptosis through exosomal miR-21 by targeting PDCD4. Cell Death Dis. 2016, 7, e2277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Greco, K.A.; Franzen, C.A.; Foreman, K.E.; Flanigan, R.C.; Kuo, P.C.; Gupta, G.N. PLK-1 Silencing in Bladder Cancer by siRNA Delivered With Exosomes. Urology 2016, 91, 241.e1–241.e7. [Google Scholar] [CrossRef]
  182. Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341. [Google Scholar] [CrossRef]
  183. Shtam, T.A.; Kovalev, R.A.; Varfolomeeva, E.Y.; Makarov, E.M.; Kil, Y.V.; Filatov, M.V. Exosomes are natural carriers of exogenous siRNA to human cells in vitro. Cell Commun. Signal. 2013, 11, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Wang, J.; Zheng, Y.; Zhao, M. Exosome-Based Cancer Therapy: Implication for Targeting Cancer Stem Cells. Front. Pharmacol. 2017, 7, 533. [Google Scholar] [CrossRef] [Green Version]
  185. Lunavat, T.R.; Jang, S.C.; Nilsson, L.; Park, H.T.; Repiska, G.; Lässer, C.; Nilsson, J.A.; Gho, Y.S.; Lötvall, J. RNAi delivery by exosome-mimetic nanovesicles–Implications for targeting c-Myc in cancer. Biomaterials 2016, 102, 231–238. [Google Scholar] [CrossRef]
  186. Limoni, S.K.; Moghadam, M.F.; Moazzeni, S.M.; Gomari, H.; Salimi, F. Engineered Exosomes for Targeted Transfer of siRNA to HER2 Positive Breast Cancer Cells. Appl. Biochem. Biotechnol. 2018, 187, 352–364. [Google Scholar] [CrossRef]
  187. Bellavia, D.; Raimondo, S.; Calabrese, G.; Forte, S.; Cristaldi, M.; Patinella, A.; Memeo, L.; Manno, M.; Raccosta, S.; Diana, P.; et al. Interleukin 3- receptor targeted exosomes inhibit in vitro and in vivo Chronic Myelogenous Leukemia cell growth. Theranostics 2017, 7, 1333–1345. [Google Scholar] [CrossRef]
  188. Kamerkar, S.; LeBleu, V.S.; Sugimoto, H.; Yang, S.; Ruivo, C.; Melo, S.; Lee, J.J.; Kalluri, R. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature 2017, 546, 498–503. [Google Scholar] [CrossRef]
  189. Ju, Z.; Ma, J.; Wang, C.; Yu, J.; Qiao, Y.; Hei, F. Exosomes from iPSCs Delivering siRNA Attenuate Intracellular Adhesion Molecule-1 Expression and Neutrophils Adhesion in Pulmonary Microvascular Endothelial Cells. Inflammation 2016, 40, 486–496. [Google Scholar] [CrossRef] [PubMed]
  190. Liu, Y.; Li, D.; Liu, Z.; Zhou, Y.; Chu, D.; Li, X.; Jiang, X.; Hou, D.; Chen, X.; Chen, Y.; et al. Targeted exosome-mediated delivery of opioid receptor Mu siRNA for the treatment of morphine relapse. Sci. Rep. 2015, 5, 17543. [Google Scholar] [CrossRef]
  191. Didiot, M.-C.; Hall, L.M.; Coles, A.H.; Haraszti, R.A.; Godinho, B.M.; Chase, K.; Sapp, E.; Ly, S.; Alterman, J.F.; Hassler, M.R.; et al. Exosome-mediated Delivery of Hydrophobically Modified siRNA for Huntingtin mRNA Silencing. Mol. Ther. 2016, 24, 1836–1847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Vaccari, J.P.D.R.; Brand, F.; Adamczak, S.; Lee, S.W.; Perez-Barcena, J.; Wang, M.Y.; Bullock, M.R.; Dietrich, W.D.; Keane, R.W. Exosome-mediated inflammasome signaling after central nervous system injury. J. Neurochem. 2015, 136, 39–48. [Google Scholar] [CrossRef]
  193. Pan, Q.; Ramakrishnaiah, V.; Henry, S.; Fouraschen, S.; de Ruiter, P.E.; Kwekkeboom, J.; Tilanus, H.W.; Janssen, H.L.A.; van der Laan, L.J. Hepatic cell-to-cell transmission of small silencing RNA can extend the therapeutic reach of RNA interference (RNAi). Gut 2011, 61, 1330–1339. [Google Scholar] [CrossRef] [PubMed]
  194. Ratajczak, J.; Miękus, K.; Kucia, M.; Zhang, J.; Reca, R.; Dvorak, P.; Ratajczak, M.Z. Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: Evidence for horizontal transfer of mRNA and protein delivery. Leukemia 2006, 20, 847–856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Ullah, M.; Liu, D.D.; Rai, S.; Razavi, M.; Concepcion, W.; Thakor, A.S. Pulsed focused ultrasound enhances the therapeutic effect of mesenchymal stromal cell-derived extracellular vesicles in acute kidney injury. Stem Cell Res. Ther. 2020, 11, 398. [Google Scholar] [CrossRef]
  196. Tomasoni, S.; Longaretti, L.; Rota, C.; Morigi, M.; Conti, S.; Gotti, E.; Capelli, C.; Introna, M.; Remuzzi, G.; Benigni, A. Transfer of Growth Factor Receptor mRNA Via Exosomes Unravels the Regenerative Effect of Mesenchymal Stem Cells. Stem Cells Dev. 2013, 22, 772–780. [Google Scholar] [CrossRef] [Green Version]
  197. Aspe, J.R.; Osterman, C.D.; Jutzy, J.; Deshields, S.; Whang, S.; Wall, N.R. Enhancement of Gemcitabine sensitivity in pancreatic adenocarcinoma by novel exosome-mediated delivery of the Survivin-T34A mutant. J. Extracell. Vesicles 2014, 3, 23244. [Google Scholar] [CrossRef]
  198. Haney, M.J.; Klyachko, N.L.; Zhao, Y.; Gupta, R.; Plotnikova, E.G.; He, Z.; Patel, T.; Piroyan, A.; Sokolsky, M.; Kabanov, A.V.; et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J. Control. Release 2015, 207, 18–30. [Google Scholar] [CrossRef] [Green Version]
  199. Cho, J.-A.; Yeo, D.-J.; Son, H.-Y.; Kim, H.-W.; Jung, D.-S.; Ko, J.-K.; Koh, J.S.; Kim, Y.-N.; Kim, C.-W. Exosomes: A new delivery system for tumor antigens in cancer immunotherapy. Int. J. Cancer 2004, 114, 613–622. [Google Scholar] [CrossRef] [PubMed]
  200. Admyre, C.; Johansson, S.M.; Paulie, S.; Gabrielsson, S. Direct exosome stimulation of peripheral humanT cells detected by ELISPOT. Eur. J. Immunol. 2006, 36, 1772–1781. [Google Scholar] [CrossRef]
  201. Tian, Y.; Li, S.; Song, J.; Ji, T.; Zhu, M.; Anderson, G.J.; Wei, J.; Nie, G. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 2014, 35, 2383–2390. [Google Scholar] [CrossRef]
  202. Schindler, C.; Collinson, A.; Matthews, C.; Pointon, A.; Jenkinson, L.; Minter, R.R.; Vaughan, T.J.; Tigue, N.J. Exosomal delivery of doxorubicin enables rapid cell entry and enhanced in vitro potency. PLoS ONE 2019, 14, e0214545. [Google Scholar] [CrossRef] [Green Version]
  203. Kim, M.S.; Haney, M.J.; Zhao, Y.; Mahajan, V.; Deygen, I.; Klyachko, N.L.; Inskoe, E.; Piroyan, A.; Sokolsky, M.; Okolie, O.; et al. Development of exosome-encapsulated paclitaxel to overcome mdr in cancer cells. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 655–664. [Google Scholar] [CrossRef] [Green Version]
  204. Yang, T.; Martin, P.; Fogarty, B.; Brown, A.; Schurman, K.; Phipps, R.; Yin, V.P.; Lockman, P.; Bai, S. Exosome Delivered Anticancer Drugs Across the Blood-Brain Barrier for Brain Cancer Therapy in Danio Rerio. Pharm. Res. 2015, 32, 2003–2014. [Google Scholar] [CrossRef]
  205. Qu, M.; Lin, Q.; Huang, L.; Fu, Y.; Wang, L.; He, S.; Fu, Y.; Yang, S.; Zhang, Z.; Zhang, L.; et al. Dopamine-loaded blood exosomes targeted to brain for better treatment of Parkinson’s disease. J. Control. Release 2018, 287, 156–166. [Google Scholar] [CrossRef] [PubMed]
  206. Aqil, F.; Kausar, H.; Agrawal, A.; Jeyabalan, J.; Kyakulaga, A.-H.; Munagala, R.; Gupta, R. Exosomal formulation enhances therapeutic response of celastrol against lung cancer. Exp. Mol. Pathol. 2016, 101, 12–21. [Google Scholar] [CrossRef]
  207. Sun, D.; Zhuang, X.; Xiang, X.; Liu, Y.; Zhang, S.; Liu, C.; Barnes, S.; Grizzle, W.; Miller, D.; Zhang, H.-G. A Novel Nanoparticle Drug Delivery System: The Anti-inflammatory Activity of Curcumin Is Enhanced When Encapsulated in Exosomes. Mol. Ther. 2010, 18, 1606–1614. [Google Scholar] [CrossRef]
  208. Aqil, F.; Munagala, R.; Jeyabalan, J.; Agrawal, A.; Gupta, R. Exosomes for the Enhanced Tissue Bioavailability and Efficacy of Curcumin. AAPS J. 2017, 19, 1691–1702. [Google Scholar] [CrossRef] [PubMed]
  209. Das, C.K.; Jena, B.C.; Banerjee, I.; Das, S.; Parekh, A.; Bhutia, S.K.; Mandal, M. Exosome as a Novel Shuttle for Delivery of Therapeutics across Biological Barriers. Mol. Pharm. 2018, 16, 24–40. [Google Scholar] [CrossRef] [PubMed]
  210. Yim, N.; Ryu, S.-W.; Choi, K.; Lee, K.R.; Lee, S.; Choi, H.; Kim, J.; Shaker, M.R.; Sun, M.R.S.W.; Park, J.-H.; et al. Exosome engineering for efficient intracellular delivery of soluble proteins using optically reversible protein–protein interaction module. Nat. Commun. 2016, 7, 12277. [Google Scholar] [CrossRef] [PubMed]
  211. Kooijmans, S.A.A.; Gitz-Francois, J.J.J.M.; Schiffelers, R.M.; Vader, P. Recombinant phosphatidylserine-binding nanobodies for targeting of extracellular vesicles to tumor cells: A plug-and-play approach. Nanoscale 2018, 10, 2413–2426. [Google Scholar] [CrossRef] [Green Version]
  212. Gomari, H.; Moghadam, M.F.; Soleimani, M. Targeted cancer therapy using engineered exosome as a natural drug delivery vehicle. Onco Targets Ther. 2018, 11, 5753–5762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Lee, H.; Park, H.; Noh, G.J.; Lee, E.S. pH-responsive hyaluronate-anchored extracellular vesicles to promote tumor-targeted drug delivery. Carbohydr. Polym. 2018, 202, 323–333. [Google Scholar] [CrossRef] [PubMed]
  214. Zhuang, M.; Chen, X.; Du, D.; Shi, J.; Deng, M.; Long, Q.; Yin, X.; Wang, Y.; Rao, L. SPION decorated exosome delivery of TNF-α to cancer cell membranes through magnetism. Nanoscale 2019, 12, 173–188. [Google Scholar] [CrossRef] [PubMed]
  215. Sáenz-Cuesta, M.; Arbelaiz, A.; Oregi, A.; Irizar, H.; Osorio-Querejeta, I.; Muñoz-Culla, M.; Banales, J.; Falcón-Pérez, J.M.; Olascoaga, J.; Otaegui, D. Methods for extracellular vesicles isolation in a hospital setting. Front. Immunol. 2015, 6, 50. [Google Scholar] [CrossRef] [PubMed]
  216. Momen-Heravi, F.; Balaj, L.; Alian, S.; Mantel, P.-Y.; Halleck, A.E.; Trachtenberg, A.J.; Soria, C.E.; Oquin, S.; Bonebreak, C.M.; Saracoglu, E.; et al. Current methods for the isolation of extracellular vesicles. Biol. Chem. 2013, 394, 1253–1262. [Google Scholar] [CrossRef]
  217. Ohno, S.-I.; Drummen, G.P.C.; Kuroda, M. Focus on Extracellular Vesicles: Development of Extracellular Vesicle-Based Therapeutic Systems. Int. J. Mol. Sci. 2016, 17, 172. [Google Scholar] [CrossRef] [Green Version]
  218. Ullah, M.; Akbar, A.; Yannarelli, G. Applications of artificial intelligence in, early detection of cancer, clinical diagnosis and personalized medicine. Artif. Intell. Cancer 2020, 1, 39–44. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of different methods for extracellular isolation and purification.
Figure 1. Schematic representation of different methods for extracellular isolation and purification.
Cells 11 00186 g001
Figure 2. Comparison of different isolation methods for EVs purification. Flow chart data is based on published literature.
Figure 2. Comparison of different isolation methods for EVs purification. Flow chart data is based on published literature.
Cells 11 00186 g002
Table 1. Comparison of EV isolation techniques in terms of source, recovery, purity, sample volume and time.
Table 1. Comparison of EV isolation techniques in terms of source, recovery, purity, sample volume and time.
MethodSourcesTimeVolumeRecoveryPurityRef
UltracentrifugationMCF-7 cell line4 h1 mL-Moderate[44]
UltracentrifugationNon-Small-Cell Lung Cancer (NSCLC) SK-MES-1 cell line20 h500 µL70%<UF[63]
UltracentrifugationHuman colon carcinoma LIM1863 cells2h500 μL5–25%Low[80]
OptiPrep™ density gradient centrifugationHuman colon carcinoma LIM1863 cells>21 h500 μL5–25%>UC[80]
OptiPrep™ density gradient centrifugationhuman breast cancer cell line MCF-720 h1 mL-Very high[44]
Density Gradient centrifugationTca8113 human tongue squamous cell carcinoma cell line20 h>1 mL>UCSimilar to UC[101]
ExoQuick-TC™ precipitationhuman breast cancer cell line MCF-713 h1 mL-Low[44]
ExoChipBlood serum<2 h<400 μLLow-[97]
TEI precipitationhuman breast cancer cell line MCF-713 h1 mL-Low[44]
UltrafiltrationNon-Small-Cell Lung Cancer (NSCLC) SK-MES-1 cell line18 h500 µL90%>UC[63]
Sequential filtrationMDA231 breast cancer cells-150 mL<UCHigh[102]
heparin/polymer-coated microspheresPlasma1 h2 mL81%High[103]
Heat Shock Protein (HSP)-binding peptide Vn96HT-29 cell32 min2 mLPoorPoor[104]
Liquid biopsy chip + HSP-binding peptide Vn96MCF720 min0.2 mL99%-[105]
Enzyme-linked immunosorbent assayLNCaP cell line
HCT116 cell line
2 h100 μL75–80%-[34]
Integrated microfluidic platformPlasma2 h30 μL>99.9%-[106]
anti-EpCAM coated magnetic beadsHuman colon carcinoma LIM1863 cellsOvernight>1 mL5–25%>UC[80]
Acoustic NanofilterRed blood cells<30 min50 μL>80%-[89]
Microfluidic ExoSearch chipBlood>40 min20 μL42–97.3%-[94]
Immune-microfluidicCell line (ovarian cancer C30)~100 min30 μL>99.9%-[95]
Microfluidic affinity separation chipSerum20–40 min20–100 μL~60%-[98]
Micro fluidic viscoelastic flowsSerum<5 min<100 μL>80%>90%[92]
Microfluidic viscoelastic flowBlood∼25 min-> 99%∼98.4%[107]
Double-filtration microfluidic deviceUrine<10 min<100 μL74.2%-[108]
Modified acousticBlood25 min100 μL82%98%[109]
Crossflow microfiltrationLipo246 cell line30 min-32–76%Low[110]
Centrifugal microfluidicHuman breast adenocarcinoma cell line, MCF-7
Lung adenocarcinoma cell line, H1975
<4 min<10 μL90%85%[111]
Table 2. Therapy based on miRNA-loaded exosomes.
Table 2. Therapy based on miRNA-loaded exosomes.
CargoDonor CellTarget CellConditionLoading MethodIsolation MethodRoute of AdministrationResultRef
Cancer Therapy
Let-7aHEK293 cell expressing GE11EGFR-expressing breast cancerBreast cancerPre-transfectionultracentrifugationi.vTumor growth inhibition[166]
miR146-bMSCGliomaPrimery brain tumortransfection using electroporationExoQuick-TCintratumoralTumor growth inhibition[167]
miR-143Human bone-marrow-derivedOsteosarcoma cell line 143BOsteosarcomaTransfection using lipofectaminultracentrifugationNot applicableMigration inhibition[168]
miR-122MSCHepatocellular carcinoma cellsHepatocellular carcinomaTransfection by plasmidExoQuick-TCintratumoralEnhancing chemotherapeutic sensitivity, tumor growth inhibition[169]
miR-134Hs578T and Hs578Ts(i)8Hs578T and Hs578Ts(i)8Breast cancerTransfectionultracentrifugation and ExoQuick,Not applicableReducing migration and invasion, Enhancing chemotherapeutic sensitivity[170]
Anti-miR-9MSCDrug resistant glioblastoma multiforme (BT145and BT164)Brain tumorPre-overexpressionUltracentrifugation and Total Exosome Isolation kit from InvitrogenNot applicableEnhancing chemotherapeutic sensitivity[171]
Neurodegenerative diseases
miR-219Dendritic cellsoligodendrocytesmultiple sclerosis and dysmyelinating syndromes-ExoQuick and precipitation with centrifugation Intra nasalincrease baseline myelination, reduce oxidative stress, and improve remyelination [172]
miR-17-92MSCCerebral cellspost–middle cerebral artery occlusion (strock)Transfection with a miR-17–92 cluster plasmidmultistep centrifugationintravenousimprovement of neurological function and oligodendrogenesis, neurogenesis, and neuritis [173]
miR-133bMSCCerebral cellsmiddle cerebral artery occlusion (MCAO) (stroke)infected with lentivirus constructed with the vectors of LentimiRaGFP-hsa-miR-133b Vectormultistep centrifugationintra arteriallyenhancing neurological recovery and plasticity post stroke[174]
miR-124Bone marrow from adult male miceIschemic cellsstrokeelectroporationultracentrifugationintravenous injectionpromoted cortical neural progenitors to obtain neuronal identity and protect against ischemic injury[175]
miR-30d-5padiposederived stem cells (ADSCs)Cerebral cellsstrokeTransfection by Lipofectamine® 2000ultracentrifugationinjection through the tail veinsuppressing the inflammatory response and preventing cerebral injury by inhibiting outophagy-mediated microglia polarization to M1[176]
Cardiovascular diseases
miR-19a, miR-451mesenchymal stem cells (MSC) overexpressing GATA-4neonatal rat ventricles cardiomyocytesMINaturally exist in exosomes of donor cellsPrecipitation (CBI)intramyocardial injectionreduced apoptosis of cardyomyioyte and enhanced resistance to cardyomyocyte hypoxia[177,178]
miR-146ahuman CDCs (or normal human dermal fibroblasts [NHDFs]Neonatal rat cardiomyoctes (NRCMs)MINaturally exist in exosomes of donor cellsExoquick ExosomePrecipitation Solution (System Biosciences)intramyocardial injectionredevelop injured heart muscle[177,179]
miR-21cardiac progenitor cell (CPC)mouse cardiac endothelial cellsMINaturally exist in exosomes of CPCprecipitated with ExoQuick TC (System Biosciences)In vitroinhibiting role in the apoptosis pathway via downregulating programmed cell death 4 [177,180]
Table 3. Summary of studies conducted on siRNA as a cargo of exosomes.
Table 3. Summary of studies conducted on siRNA as a cargo of exosomes.
CargoDonor CellIsolation MethodLoading MethodConditionBiological EffectRef
PLK1 siRNAhuman embryonic kidney (HEK) cellultracentrifugationelectroporationBladder cancerEffective delivery of PLK1 siRNA[181]
GAPDH siRNAEngeneered self-dendritic cells to express Lamp2bultracentrifugationelectroporationAlzheimer’s diseaseknockdown of BACE1[182]
Alexa flour 488 labeled siRNAHeLa and ascites with the presence of exosomal marker proteins HLA-ABC and CD63 on the membrane of these exosomes by dot blot analysis.ultracentrifugationchemical treatment (lipofectamin)cancerSilencing of RAB51[183,184]
siRNANIH3T3 cellsultracentrifugationelectroporationlymphomaSilencing of c-Myc and stimulation of caspase-3[184,185]
siRNAHEK293T cell (transduced by a lentiviral vector bearing-LAMP2b-DARPin G3 chimeric gene)sequential centrifugationelectroporationHER2-positive breast cancer (SKBR3 cells)Down regulating the TPD52 gene[186]
BCR-ABL siRNAHEK293T (transfected with of IL3-Lamp2b plasmid DNA)ultracentrifugationTransfection with LipofectaminChronic Myeloid Leukemiainhibit Bcr-Abl and cancer cell growth[187]
siRNA or shRNA targeting KRASnormal fibroblast-like mesenchymal cellsultracentrifugationelectroporationPancreatic cancersuppressed cancer in multiple mouse models[188]
siRNAhuman induced pluripotent stem cells (huiPSCs)centrifugationelectroporationPulmonary inflammationEfficient delivery of the target siRNA into HMVECs, inhibiting the ICAM-1 protein expression[189]
opioid receptor mu (MOR) siRNAhuman embryonic kidney 293T cotransfected with siRNA and the RVG-Lamp2b plasmid using Lipofectamine 2000)using an exosome isolation kit (Invitrogen)transfectionmorphine addictionreduces MOR mRNA and protein levels in Neuro2A cells and the mouse brain[190]
hydrophobically modified small interfering RNAs(hsiRNAs)U87 glioblastoma cells ultracentrifugationco-incubationHuntington’s diseasebilateral silencing of Huntingtin mRNA[191]
SiRNAembryonic cortical neuronal cultureExoQuick (based on the production company’s instruction)electroporationSpinal cord-injuryknockdown of ASC protein transformation and significant decrease in caspase 1 activation[192]
siRNAHuman hepatoma cells HuH7ultracentrifugationTranfection (Lentiviral vectors LV-shCD81 and LV-shNS5b were constructed. LV-shNS5b contains expression cassettes of shRNA and targets the viral NS5b region)Hepatitis C virus infection and other liver diseasesuppression of CD81 expression in target hepatocytes[193]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Akbar, A.; Malekian, F.; Baghban, N.; Kodam, S.P.; Ullah, M. Methodologies to Isolate and Purify Clinical Grade Extracellular Vesicles for Medical Applications. Cells 2022, 11, 186. https://doi.org/10.3390/cells11020186

AMA Style

Akbar A, Malekian F, Baghban N, Kodam SP, Ullah M. Methodologies to Isolate and Purify Clinical Grade Extracellular Vesicles for Medical Applications. Cells. 2022; 11(2):186. https://doi.org/10.3390/cells11020186

Chicago/Turabian Style

Akbar, Asma, Farzaneh Malekian, Neda Baghban, Sai Priyanka Kodam, and Mujib Ullah. 2022. "Methodologies to Isolate and Purify Clinical Grade Extracellular Vesicles for Medical Applications" Cells 11, no. 2: 186. https://doi.org/10.3390/cells11020186

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop