Next Article in Journal
All-in-One Process for Mass Production of Membrane-Type Carbon Aerogel Electrodes for Solid-State Rechargeable Zinc-Air Batteries
Next Article in Special Issue
Potential of Lipid-Based Nanocarriers against Two Major Barriers to Drug Delivery—Skin and Blood–Brain Barrier
Previous Article in Journal
Mechanism of HMBR in Reducing Membrane Fouling under Different SRT: Effect of Sludge Load on Microbial Properties
Previous Article in Special Issue
Ameliorative Effects of Camel Milk and Its Exosomes on Diabetic Nephropathy in Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in Purification, Modification, and Application of Extracellular Vesicles for Novel Clinical Treatments

1
Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
2
Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-0031, Japan
3
Department of Infectious Diseases, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo 181-0004, Japan
*
Author to whom correspondence should be addressed.
Membranes 2022, 12(12), 1244; https://doi.org/10.3390/membranes12121244
Submission received: 20 November 2022 / Revised: 30 November 2022 / Accepted: 6 December 2022 / Published: 8 December 2022
(This article belongs to the Special Issue Drug Delivery Systems Based on Extracellular Vesicles)

Abstract

:
Extracellular vesicles (EV) are membrane vesicles surrounded by a lipid bilayer membrane and include microvesicles, apoptotic bodies, exosomes, and exomeres. Exosome-encapsulated microRNAs (miRNAs) released from cancer cells are involved in the proliferation and metastasis of tumor cells via angiogenesis. On the other hand, mesenchymal stem cell (MSC) therapy, which is being employed in regenerative medicine owing to the ability of MSCs to differentiate into various cells, is due to humoral factors, including messenger RNA (mRNA), miRNAs, proteins, and lipids, which are encapsulated in exosomes derived from transplanted cells. New treatments that advocate cell-free therapy using MSC-derived exosomes will significantly improve clinical practice. Therefore, using highly purified exosomes that perform their original functions is desirable. In this review, we summarized advances in the purification, modification, and application of EVs as novel strategies to treat some diseases.

1. Introduction

Extracellular vesicles (EVs) were discovered in 1946 [1]. Secretory vesicles with a diameter of approximately 100 nm, discovered in 1981 during reticulocyte research, were first named exosomes in 1987 [2]. Subsequently, for a long time, exosomes were thought to be a part of intracellular waste disposal mechanism [3]. However, exosomes contain messenger RNA (mRNA) and microRNA (miRNA) derived from secretory cells, and information exchange takes place between cells when exosomes are transported to other cells [4,5,6]. Since the amount and type of functional molecules, such as RNAs and proteins, in exosomes or membranes vary with diseases, they are suitable for disease diagnosis, prognosis, and identification of therapeutic targets [7,8]. However, since exosome composition does not always match that of secretory-derived cells, the mechanism by which specific molecules are sorted into luminal vesicles in multivesicular bodies remains largely unknown. However, several RNA-binding proteins were found in exosomes [9,10]. Since exosomes are regarded as natural drug delivery systems, they are widely used as a drug discovery technology [11,12]. Furthermore, since exosomes exist in many species, the possibility of information transmission across species has also been suggested, and studies are being conducted to elucidate the mechanisms of various life phenomena and to broadly apply them in the health and medical fields [12,13,14,15,16].
Extracellular vesicle is a general term for membrane vesicles surrounded by a lipid bilayer that is secreted by various tissues and cells [17,18,19,20,21,22,23,24,25,26,27]. These vesicles are classified into exosomes (40–200 nm) that are secreted outside the cell, microvesicles (200–1000 nm) that directly bud from the cell membrane and are secreted outside the cell, and apoptotic vesicles (1000–5000 nm) that are secreted from apoptotic cells [24,28,29,30,31,32,33,34,35]. However, identifying the origin of extracellularly secreted EVs is challenging. Since the classification of EVs is vague, the International Society for EVs (ISEV) has taken the lead to organize EVs [36,37,38,39]. To separate EVs in a pure form from other EVs, such as microvesicles, using centrifugal force is challenging. Categorization into small (ultracentrifugation 100,000× g pellet fraction, particles of size equivalent to exosomes sediment), medium (medium speed centrifugation 20,000× g pellet), and large EVs (low-speed centrifugation 2000× g pellet) has been proposed [40]. Furthermore, through analysis of EVs fractionated according to their size, they were classified into exomeres, without a lipid bilayer, small exosomes, and large exosomes [41,42,43,44,45,46,47,48]. Since exosomes refer to the sum of exosomes released by various cells, only their average values can be analyzed. For example, when exosomes are isolated from peripheral blood to identify those that are diseased and cell derived, the biomarker detection sensitivity is low because healthy cell-derived exosomes are present in majority. Therefore, establishing a method for sorting exosomes using markers specific to organs and cells is desirable. Furthermore, the technology of analyzing exosomes at the level of one particle per cell is still insufficient; however, this technology is hypothesized to lead to the development of highly sensitive diagnostic methods [49,50,51]. In addition, clearly distinguishing exosomes from other EVs is challenging, and high sensitivity can be achieved only if exosomes are highly pure at the time of analysis.

2. Constitution and Characterization of Exosomes

Exosomes are EVs produced from multivesicular endosomes during cell endocytosis and are extracellularly secreted [52,53,54,55,56,57,58,59,60,61,62]. Exosomes are internally formed by budding within endocytic compartments through the fusion of plasma membranes and vesicle-containing endosomes [63,64,65,66]. They are isolated from body fluids such as blood, urine, saliva, and cerebrospinal fluid and contain various biomolecules such as proteins, nucleic acids, and lipids, which are important for intercellular communication [67,68,69,70,71,72,73,74,75,76,77,78,79,80]. Therefore, EVs have attracted attention as biomaterials with drug-delivery capabilities. Owing to their endogenous origin, exosomes are less likely to be immunogenic or cytotoxic compared with that of other artificial delivery agents [59,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109]. In addition, the lipid bilayers of exosomes can protect drugs from rapid blood clearance and reduce unintended drug-induced cytotoxicity. Notably, the exosome is large enough to prevent rapid renal clearance and small enough to avoid being absorbed through the reticuloendothelial system [110]. Small nanoparticles, such as exosomes, tend to accumulate in cancerous tumor sites owing to leaky blood vessels and abnormal lymphatic drainage; thus, exosomes are ideal for drug delivery to treat certain cancers [78,87,93,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129]. Similar to other vesicles, exosomes consist of a lipid bilayer membrane with an aqueous inner compartment and a lipophilic outer layer. This structure allows both hydrophobic and hydrophilic drugs to be loaded into exosomes. Exosomes are of different types; although they contain different amounts of cellular components, certain lipids, proteins, and nucleic acids are common components [8,21,71,72,75,79,85,91,92,102,112,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180]. Exosomes contain large amounts of cholesterol, sphingolipids, phosphoglycerides, ceramides, and saturated fatty acids, which bind with each other to contribute to exosome stability, and a variety of membrane-bound and intracellular proteins [181,182,183,184,185,186,187,188,189,190]. The most common molecules are membrane transporters and fusion proteins, major histocompatibility complexes, heat shock proteins, tetraspanins, endosomal transport sorting complexes required for transport (ESCRTs), and lipid raft-associated proteins [109,134,191,192]. In addition, exosomes are rich in proteins that are specific to the cell types that contain them. For example, exosomes extracted from dendritic cells are rich in a heat shock protein (Hsp73) that may independently exert the anti-tumor effects observed in the whole exosomes [193]. In addition, exosomes contain nucleic acids such as miRNAs, non-coding RNAs, and mRNAs [194,195,196,197,198,199]. Notably, these RNAs are found within the aqueous compartment of exosomes and are bound to the outer membrane of exosomes via the protein Argonaute 2 (Ago2) [200]. Specific miRNAs in exosomes can be targeted using proteins that recognize short RNA motifs, such that RNAs can be selectively packaged into exosomes (Figure 1) [201,202,203,204,205,206,207].
Further, exosomes are released from various cell types, providing a wide range of donor cell options to isolate exosomes. Two important factors in determining donor cells are the biological characteristics of exosomes and amount of exosomes extracted from a particular cell type. For example, exosomes of dendritic cells stimulate stronger anti-tumor immune responses compared with those of exosomes from EG7 tumor cells; exosomes of dendritic cells promote proliferation and differentiation of T cells or may contain more molecular factors that more efficiently interact with T cells compared with those of exosomes extracted from tumor cells [208]. Mesenchymal stem cells (MSCs) produce many exosomes, which are easy to isolate, can be cultured in large quantities, and promotes cell viability [209,210]. They have been identified as a particularly promising cell type. Whether cell culture media or body fluids are used, the purity and quantity of exosomes are extremely important for the development of exosome-based drug delivery agents. Exosomes are typically purified using differential ultracentrifugation and quantified using protein assays [211,212,213,214]. Other methods include filtration, immunoaffinity isolation, and microfluidic analytical techniques to rapidly isolate exosomes for structural and physical analyses; however, whether exosomes purified using these new methods are effective for drug delivery is unclear [215,216,217,218]. Using in vivo or ex vivo techniques, therapeutic agents can be loaded into exosomes. Various ex vivo techniques, such as freeze–thaw cycles, saponin membrane permeabilization, sonication, and extrusion, have been used to load drugs into exosomes [218,219,220,221]; in these techniques, drug behavior and activity were maintained after loading the drugs into exosomes. Additionally, exosomes are stable when stored at −20 °C to −80 °C and exposed to several freeze–thaw cycles [222,223].

3. Molecular Regulation and Biomarkers with Exosome in Diseases

The most advanced research on the functions and applications of exosomes has been in the field of cancer. For example, pancreatic cancer-derived exosomes were shown to make normal cells malignant, and the exosomal proteins, secreted by pancreatic cancer cells, that were responsible for inducing malignancy were identified [178,224,225,226]. Exosome-encapsulated miRNAs released from cancer cells induce angiogenesis within tumors and are involved in cancer proliferation and metastasis [227,228,229,230]. Exosomes secreted from metastatic cancer cells are enriched in specific proteins that promote the formation of a premetastatic niche [178,231,232,233,234]. In addition, miR-155, which is encapsulated in exosomes secreted by breast cancer cells, promotes the formation of beige and brown adipocytes, induces metabolic remodeling by suppressing Pparg expression, and is involved in cancer cachexia [235]. Exosomes are involved in carcinogenesis and malignant transformation [236,237]. In neurodegenerative diseases, abnormally aggregated proteins, which are causative proteins, are released outside the cells by exosomes and spread to surrounding cells [238]. Further, exosomes also deliver the viral genome and proteins to the cells surrounding the infected cells during the process of virus propagation between cells in vivo, which is advantageous for viral survival [239]. In the immune system, they are involved in various immune function controls, such as the exchange of antigen information between immune cells and activation/inactivation of immune cells [240,241]. Exosomes are involved in various diseases, and since the types and amounts of functional molecules encapsulated in exosomes or present in the membrane vary depending on the disease, they are highly likely to be applied in disease detection and prognosis and used as therapeutic targets [242,243,244]. Functional molecules, such as RNAs and proteins, that are encapsulated in exosomes or expressed on membranes are stably retained, and their amounts and types vary depending on the disease. For example, in patients with myalgic encephalomyelitis and chronic fatigue syndrome, which are challenging to diagnose, the amount of EVs in plasma increases, and actin network proteins such as talin-1 and filamin A, which are characteristically encapsulated, may serve as biomarkers [245,246,247]. In addition, since neuron-derived exosomes contain substances such as tau, α-synuclein, and TDP-43 that aggregate within neurons, they contain proteins that cause the onset of Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis and are drawing attention for their association with the onset of each disease [238,248,249,250,251,252,253]. Currently, cerebrospinal fluid collected by lumbar puncture is used for research and diagnosis using brain-derived exosomes [254]. In particular, exosome-encapsulated proteins derived from disease-related cells leaked into body fluids, such as α -synuclein in Parkinson’s disease, function as biomarkers of a disease and its severity [132,238,248,249]. When marker candidates useful for early diagnosis and stratification of diseases are identified, verification based on immunoassay methods such as ELISA is performed. However, analysis of endogenous proteins has not sufficiently progressed compared with that of exosome membrane proteins. This is because of challenges associated with immunoassays in which antibodies react with encapsulated proteins and antigens are enclosed within the membrane of exosomes.
In addition, since some brain-derived exosomes are also detected in peripheral blood, neural cell adhesion molecule (NCAM-1) and L1 cell adhesion molecule (L1CAM) were reported as markers [255,256]. Furthermore, exosomes in urine are new diagnostic markers for kidney, prostate, and bladder diseases, those in cerebrospinal fluid are markers for tumors and neurodegeneration in the brain, and those in amniotic fluid are markers that reflect fetal status [69,257,258,259,260,261]. In addition, since exosomes function as disease mediators, suppression of exosome secretion is attracting attention as a new therapeutic method. For example, in drug repositioning of existing drugs, to control the production and secretion of exosomes, a high-throughput screen of 4580 compounds was performed in prostate cancer cells, and five exosome production inhibitor and six exosome production activator candidate compounds were identified [262]. Furthermore, a study that aimed at cancer-specific regulation identified that miR-26a and its regulatory genes SHC4, PFDN4, and CHORDC1 regulate exosome secretion by prostate cancer cells [263]. Thus, techniques that can specifically manipulate and modify functional molecules encapsulated in exosomes will contribute to exosome function elucidation and effective functional expression.
In addition, the effect of mesenchymal stem cell (MSC) therapy, which is being applied in regenerative medicine owing to its ability to differentiate into various cells, is due to humoral factors, including mRNA, miRNAs, proteins, and lipids, which are encapsulated in exosomes derived from transplanted cells [264,265,266]. Exosomes derived from MSCs suppress tissue fibrosis in liver and kidney diseases and are also effective in treating heart and Alzheimer’s diseases [89,267,268]. In addition, clinical trials were conducted to verify the neuroprotective and anti-fibrosis effects of exosomes secreted by umbilical cord-derived MSCs after cochlear implant surgery and to evaluate the wound healing effect of platelet-derived exosomes [269].

4. Applications of Exosome as Novel Drug Delivery System

With the recognition of the importance of EVs, development of techniques that individually isolate them and analyzing the molecules contained within hold high significance. The transfer of the contents of exosomes to other cells has been analyzed using DNA, RNA, proteins, and intercellular signaling substances. Although the detailed delivery mechanism is of high significance, it has not yet been elucidated. A study reported that EVs specifically adhere to cells, and the development of observational techniques, including live-cell imaging, is necessary to clarify this phenomenon. Clarifying the delivery mechanism of the cargo of EVs will not only lead to true understanding of the types of organisms affected by EVs but also provide clues to the control of substance transport by EVs; the results will also be useful in designing artificial particles that mimic EVs. A device that isolates exosomes in a fluid using microfabrication technology and a method that attaches a tag, such as a barcode, to isolate them are being tested [270,271]. The method of analyzing molecules contained in a single vesicle is challenging, and the extension of conventional omics technology has limitations. An enhanced Raman scattering method using a metal near field and a device that analyzes the surface proteins of individual extracellular microparticles using fluorescent antibodies is under development [272]. In addition, live imaging is a powerful method for understanding the mechanism of EV formation. Super-resolution imaging is essential for observing exosomes with a diameter of ≤200 nm. Its application to living cells was a challenge; however, high-speed super-resolution live imaging technology has been developed [273]. In addition, a technique was developed for the real-time observation of extracellularly released microparticles using a single-cell observation chip [274]. The development of imaging technology is indispensable for the dynamic analysis of structures of several nanometers. This requires the development of microscopes and techniques associated with observing samples, such as sample-fixation techniques and microfluidic devices.
Unlike artificial products such as liposomes, exosomes can function as a natural drug delivery system (DDS), delivering siRNA, miRNA, and low-molecular-weight compounds to target cells. The in vivo dynamics of exosomes is not yet elucidated, and elucidation of the molecular mechanism that controls the dynamics has the potential to contribute to the development of DDSs with high target accuracy. Various cell adhesion molecules and sugar chains are expressed on the surface of exosomes, and based on the expression pattern, the cells whose exosomes exhibit affinity are identified [275,276]. Further, new DDSs are being developed by modifying and applying the properties of exosomes. For example, by encapsulating a STING agonist in exosomes, in which a therapeutic protein such as prostaglandin F2 receptor negative regulator (PTGFRN), is highly expressed in the exosome membrane, ExoSTING was developed to activate the STING pathway in cancer in an antigen-presenting cell-specific manner [277]. Furthermore, clinical trials of exoIL-12, in which IL-12 locally acts on cancer cells through exosomes (with IL-12 expressed on their surface), were conducted (Figure 2) [278]. In addition, bovine milk-derived exosomes are being developed as carriers for oral administration of nucleic acids, peptides, and small molecules, which are challenging to be orally administered [279].
In addition, delivering drugs across certain biological barriers, especially the blood–brain barrier (BBB), is a big challenge in chemotherapy. For example, exosomes released by glioblastomas have been detected in the serum, suggesting that endogenous exosomes cross the BBB [280]. Furthermore, in experiments with mice, exosome preparations loaded with the anti-inflammatory polyphenol curcumin were delivered through the nasal cavity, inducing apoptosis of follicle cells, suggesting that vesicle preparations cross the BBB [281]. In addition, exosomes extracted from brain epithelial cells and loaded with anticancer agents cross the BBB and induce cytotoxicity in zebrafish tumor cells [282]. Taken together, these results suggest that exosomes are particularly effective for drug delivery to the brain.

5. Purification Methods and Drawbacks of Exosome

Although EV-based drug delivery appears to be a promising and effective treatment, several major challenges need to be resolved before they can be safely and efficiently introduced for clinical applications. First, EV isolation and purification procedures should be standardized to eliminate contaminants such as protein aggregates and to improve reproducibility. Second, identifying donor cells that are stable sources of EVs and fully characterizing the EVs extracted from these cells are essential. Finally, developing highly efficient methods for loading drugs into EVs to maximize drug delivery efficacy is necessary. While studies on the identification of biomarkers for diseased cell-derived exosomes and development of diagnostic methods using them are being actively conducted, little progress has been made in the development of methods to particularly isolate diseased cell-derived exosomes. By identifying surface markers specific to exosomes derived from diseased cells, the development of a removal method that does not affect exosomes derived from normal cells is desired. To use exosomes as therapeutic agents and DDS tools, large amounts of high-quality exosomes have to be prepared. As the condition of the producing cells also affects the quality of exosomes, strict culture conditions have to be set. In addition, application of techniques that increase the amount of exosomes produced is recommended. Furthermore, implementing isolation and purification methods using ultracentrifugation on an industrial scale is challenging, and the establishment of other methodologies that can process large amounts of EVs is desired. In addition, establishing evaluation methods and concepts that strictly define the quality of exosomes, such as quantity, purity, particle size, distribution, homogeneity, and potency are necessary.
Thus, various studies are being conducted on EVs, particularly on exosomes; however, a major bottleneck is the lack of standard and efficient techniques for the production and isolation of EVs. When dealing with body fluids, EVs coexist with many proteins and cells with physical and chemical properties similar to that of EVs, making their isolation inherently complex [283,284]. The main separation methods used are those that use differences in EV density, size, and specific surface markers. Techniques based on these principles include ultracentrifugation, precipitation, filtration, size-exclusion chromatography, immunoaffinity, and antigen-antibody reactions [210,211,212,213,214,284,285,286,287,288]. Ultracentrifugation is a separation method that exploits differences in density and size between cells, EVs, and proteins. Separation using an ultracentrifuge is usually used to isolate EVs; however, the drawbacks of this technique are the time taken for collection and high throughput. Cells, apoptotic bodies, and large vesicular fractions of EVs can be separated using standard centrifugation at <20,000× g, whereas centrifugation at <100,000× g should be used to purify exosomes from proteins (Figure 3) [289,290,291]. The major drawback of this method is that it requires a high spin speed and a long operating time of approximately 5 h. In addition, the ultracentrifugation method has a drawback in that the recovery rate of exosomes is low and protein aggregates co-precipitate. Sucrose density gradient centrifugation was used as an additional technique for ultracentrifugation to improve the isolation purity efficiency of exosomes [292]. The sedimentation method was developed without the ultracentrifugation step and has challenges with recovery time and high throughput. The separation efficiencies of these sedimentation methods were compared with those of conventional ultracentrifugation methods, which indicated high separation efficiencies [293,294]. However, factors such as residual precipitating matrices and polymeric additives can affect the biological activity and properties of EVs, including exosomes. In filtration method, membrane filters (pore size of approximately 50 to 450 nm) are used to separate large vesicle components of cells, including EVs, in biological samples [295]. The filtration method uses a membrane to sieve the large vesicle fractions and EVs, followed by the flow of the small vesicle fractions of EVs and exosomes from the proteins using ultracentrifugation. To separate the small vesicle fraction of EVs/exosomes from protein. aggregates and avoid ultracentrifugation, for molecules of 100 kDa, ultrafiltration is generally used. Filtration is generally faster than ultracentrifugation, but exosome yield may decrease due to clogging effects caused by the non-optimization of the operating procedure [296,297]. In addition, size exclusion chromatography (SEC) was used to separate EVs and exosomes from protein aggregates [298,299,300]. Typically, cells and large vesicle fractions of EVs are removed using centrifugation or filtration, followed by small vesicle fractions and exosomes using a size exclusion column. Small substances, such as proteins, are retained on the column for a long time, while large substances, including small vesicle fractions of EVs and exosomes, elute early. Therefore, the separation of small vesicle fractions and exosomes of EVs can be achieved by collecting fractions that elute at specific times. Immunoaffinity separation is a method for separating exosomes from other EVs using specific surface markers [301,302,303,304,305,306]. Further, EVs and exosomes contain cell-of-origin specific markers, and antigen-antibody reactions can prove to be beneficial. A common immunoaffinity-based separation method utilizes antibody-coated magnetic beads to capture EVs and exosomes that contain specific markers, in bodily fluids. Although this method allows the isolation of specific subfractions of EVs or exosomes, it is generally not suitable for isolating EVs or exosomes from large amounts of biological samples. In addition, by using a molecule that binds to phospholipid phosphatidylserine, unlike in conventional methods such as ultracentrifugation, that is specifically expressed on the surface of exosomes, purifying exosomes with 100 times more purity that conventional methods and detecting exosomes with high sensitivity are possible [52]. Moreover, the conventional separation methods require dedicated laboratory equipment or reagents and multi-step work processes, and analyzing EVs and exosomes as routine diagnostic processes in clinical practice poses many challenges. However, microfluidic systems may overcome these shortcomings; this technology enables rapid isolation and analysis of EVs and exosomes from clinical specimens, facilitating diagnosis and treatment [307,308,309,310,311,312]. Further, the microfluidic system not only provides a multipurpose platform for the isolation and analysis of EVs and exosomes, but also contributes to the integration and simplification of multiple processes and risk reduction of cross-contamination. In addition, various methods of separating particles by employing characteristics such as particle size, shape, and electric charge of fine particles have been explored.

6. Application of Exosome in Plant and Food as DDS

Furthermore, exosome studies have mainly focused on animals, such as mammals, but plants also release exosome-like EVs [313,314,315,316]. Using plant-derived exosomes, the growth and toxicity of fungi that are harmful to plants can be suppressed, enabling plants to defend themselves [317]. In addition, the functions of exosomes present in food ingested by humans are also being investigated, and ingested plant-derived exosomes affect the composition of the intestinal microbiota and physiological functions of the host [318,319].
Further, EVs have been shown to activate immunity in various bacteria [320,321,322,323,324]. Vaccines against infectious diseases, including coronavirus disease 2019, and new immunotherapies with cancer-suppressing effects have been developed using new technologies that contribute to the development of modified EVs [325,326,327]. Although the mechanism is unknown, EVs tend to accumulate in cancer cells and are also attracting attention as a tool in DDSs to target specific cells [328]. Further, EVs are beginning to be used as vaccines and antibiotic transporters worldwide; however, since EVs are heterogeneous, the products used for clinical applications must be highly pure and uniform [242,329,330,331,332]. Therefore, modifying and artificially synthesizing EVs are essential. Further, DDSs that use liposomes and other technologies to encapsulate and localize arbitrary proteins, nucleic acids, and other substances are also essential. Thus, studies on the development of a new artificial membrane particle that mimics EVs are essential.

7. Conclusions

Exosomes, which are representative EVs, have a wide range of applications, from the elucidation of disease mechanisms to diagnosis and treatment, mainly in the field of cancer and neurological diseases. However, exosomes secreted from MSCs have therapeutic effects in various diseases, and studies are conducted to develop new therapeutic agents for these diseases. As an increasing number of studies indicate the potential of exosomes as therapeutic agents, the scope of drug discovery using exosomes widens. In the field of regenerative medicine, in particular, new treatments that advocate cell-free therapy using exosomes derived from MSCs, tissue stem cells, and immunocompetent cells is highly likely to be applied in clinical practice. In contrast to exosomes that exhibit benign properties, malignant exosomes are released due to a disease function through cancer cell metastasis and drug resistance. With the new understanding of cancer metastasis through exosomes, blocking exosome secretion and function of cancer cells will lead to drug discovery. Thus, exosome research has progressed at an accelerated pace, and the use of highly purified exosomes that perform their original functions is preferred. This purification technology to obtain exosomes of high purity is hypothesized to become an innovative analysis technology that will significantly change the methodology of exosome studies.

Author Contributions

Writing—review and editing, Y.M.; supervision, R.Y.; funding acquisition, Y.M. All authors have read and agreed to the published version of the manuscript.

Funding

This review was funded by Fukuda Foundation for Medical Technology.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bazzan, E.; Tinè, M.; Casara, A.; Biondini, D.; Semenzato, U.; Cocconcelli, E.; Balestro, E.; Damin, M.; Radu, C.M.; Turato, G.; et al. Critical Review of the Evolution of Extracellular Vesicles’ Knowledge: From 1946 to Today. Int. J. Mol. Sci. 2021, 22, 6417. [Google Scholar] [CrossRef]
  2. Osaki, M.; Okada, F. Exosomes and Their Role in Cancer Progression. Yonago Acta. Med. 2019, 62, 182–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Soleymaninejadian, E. Exosome a Story from Waste to Become a Gold Mine. Jenta. J. Cell. Mol. Biol. 2020, 11, e107622. [Google Scholar] [CrossRef]
  4. Negrete-García, M.C.; de Jesús Ramos-Abundis, J.; Alvarado-Vasquez, N.; Montes-Martínez, E.; Montaño, M.; Ramos, C.; Sommer, B. Exosomal Micro-RNAs as Intercellular Communicators in Idiopathic Pulmonary Fibrosis. Int. J. Mol. Sci. 2022, 23, 11047. [Google Scholar] [CrossRef] [PubMed]
  5. Sun, H.; Sun, R.; Song, X.; Gu, W.; Shao, Y. Mechanism and clinical value of exosomes and exosomal contents in regulating solid tumor radiosensitivity. J. Transl. Med. 2022, 20, 189. [Google Scholar] [CrossRef]
  6. Xu, Y.X.; Pu, S.D.; Li, X.; Yu, Z.W.; Zhang, Y.T.; Tong, X.W.; Shan, Y.Y.; Gao, X.Y. Exosomal ncRNAs: Novel therapeutic target and biomarker for diabetic complications. Pharmacol. Res. 2022; in press. [Google Scholar] [CrossRef]
  7. Ipinmoroti, A.O.; Pandit, R.; Matthews, Q.L. Regenerative mesenchymal stem cell-derived extracellular vesicles: A potential alternative to cell-based therapy in viral infection and disease damage control. WIREs Mech. Dis. 2022, 14, e1574. [Google Scholar] [CrossRef]
  8. Meng, F.; Xue, X.; Yin, Z.; Gao, F.; Wang, X.; Geng, Z. Research Progress of Exosomes in Bone Diseases: Mechanism, Diagnosis and Therapy. Front. Bioeng. Biotechnol. 2022, 10, 866627. [Google Scholar] [CrossRef]
  9. Groot, M.; Lee, H. Sorting Mechanisms for MicroRNAs into Extracellular Vesicles and Their Associated Diseases. Cells 2020, 9, 1044. [Google Scholar] [CrossRef]
  10. Pan, Z.; Zhao, R.; Li, B.; Qi, Y.; Qiu, W.; Guo, Q.; Zhang, S.; Zhao, S.; Xu, H.; Li, M.; et al. EWSR1-induced circNEIL3 promotes glioma progression and exosome-mediated macrophage immunosuppressive polarization via stabilizing IGF2BP3. Mol. Cancer 2022, 21, 16. [Google Scholar] [CrossRef]
  11. Tenchov, R.; Sasso, J.M.; Wang, X.; Liaw, W.S.; Chen, C.A.; Zhou, Q.A. Exosomes─Nature’s Lipid Nanoparticles, a Rising Star in Drug Delivery and Diagnostics. ACS Nano. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  12. Patel, S.; Schmidt, K.F.; Farhoud, M.; Zi, T.; Jang, S.C.; Dooley, K.; Kentala, D.; Dobson, H.; Economides, K.; Williams, D.E. In vivo tracking of [89Zr]Zr-labeled engineered extracellular vesicles by PET reveals organ-specific biodistribution based upon the route of administration. Nucl. Med. Biol. 2022, 112–113, 20–30. [Google Scholar] [CrossRef] [PubMed]
  13. Ogami, K.; Suzuki, H.I. Nuclear RNA Exosome and Pervasive Transcription: Dual Sculptors of Genome Function. Int. J. Mol. Sci. 2021, 22, 13401. [Google Scholar] [CrossRef]
  14. Jiang, X.; You, L.; Zhang, Z.; Cui, X.; Zhong, H.; Sun, X.; Ji, C.; Chi, X. Biological Properties of Milk-Derived Extracellular Vesicles and Their Physiological Functions in Infant. Front. Cell Dev. Biol. 2021, 9, 693534. [Google Scholar] [CrossRef] [PubMed]
  15. Xu, X.H.; Shao, S.L.; Guo, D.; Ge, L.N.; Wang, Z.; Liu, P.; Tao, Y.Y. Roles of microRNAs and exosomes in Helicobacter pylori associated gastric cancer. Mol. Biol. Rep. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  16. Jiang, C.; Zhang, N.; Hu, X.; Wang, H. Tumor-associated exosomes promote lung cancer metastasis through multiple mechanisms. Mol. Cancer 2021, 20, 117. [Google Scholar] [CrossRef]
  17. Aimaletdinov, A.M.; Gomzikova, M.O. Tracking of Extracellular Vesicles’ Biodistribution: New Methods and Approaches. Int. J. Mol. Sci. 2022, 23, 11312. [Google Scholar] [CrossRef] [PubMed]
  18. De Sousa, K.P.; Rossi, I.; Abdullahi, M.; Ramirez, M.I.; Stratton, D.; Inal, J.M. Isolation and characterization of extracellular vesicles and future directions in diagnosis and therapy. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  19. Ng, C.Y.; Kee, L.T.; Al-Masawa, M.E.; Lee, Q.H.; Subramaniam, T.; Kok, D.; Ng, M.H.; Law, J.X. Scalable Production of Extracellular Vesicles and Its Therapeutic Values: A Review. Int. J. Mol. Sci. 2022, 23, 7986. [Google Scholar] [CrossRef] [PubMed]
  20. Arifin, D.R.; Witwer, K.W.; Bulte, J.W.M. Non-Invasive imaging of extracellular vesicles: Quo vaditis in vivo? J. Extracell. Vesicles 2022, 11, e12241. [Google Scholar] [CrossRef]
  21. Matsuzaka, Y.; Yashiro, R. Therapeutic Strategy of Mesenchymal-Stem-Cell-Derived Extracellular Vesicles as Regenerative Medicine. Int. J. Mol. Sci. 2022, 23, 6480. [Google Scholar] [CrossRef] [PubMed]
  22. Vinaiphat, A.; Sze, S.K. Proteomics for comprehensive characterization of extracellular vesicles in neurodegenerative disease. Exp. Neurol. 2022, 355, 114149. [Google Scholar] [CrossRef] [PubMed]
  23. Avalos, P.N.; Forsthoefel, D.J. An Emerging Frontier in Intercellular Communication: Extracellular Vesicles in Regeneration. Front. Cell Dev. Biol. 2022, 10, 849905. [Google Scholar] [CrossRef]
  24. Trisko, J.; Fleck, J.; Kau, S.; Oesterreicher, J.; Holnthoner, W. Lymphatic and Blood Endothelial Extracellular Vesicles: A Story Yet to Be Written. Life 2022, 12, 654. [Google Scholar] [CrossRef] [PubMed]
  25. Hua, Y.; Chang, X.; Fang, L.; Wang, Z. Subgroups of Extracellular Vesicles: Can They Be Defined by “Labels?”. DNA Cell Biol. 2022, 41, 249–256. [Google Scholar] [CrossRef]
  26. Berezin, A.E.; Berezin, A.A. Extracellular Vesicles and Thrombogenicity in Atrial Fibrillation. Int. J. Mol. Sci. 2022, 23, 1774. [Google Scholar] [CrossRef]
  27. Soler-Botija, C.; Monguió-Tortajada, M.; Munizaga-Larroudé, M.; Gálvez-Montón, C.; Bayes-Genis, A.; Roura, S. Mechanisms governing the therapeutic effect of mesenchymal stromal cell-derived extracellular vesicles: A scoping review of preclinical evidence. Biomed. Pharmacother. 2022, 147, 112683. [Google Scholar] [CrossRef] [PubMed]
  28. Liu, C.; Li, Y.; Han, G. Advances of Mesenchymal Stem Cells Released Extracellular Vesicles in Periodontal Bone Remodeling. DNA Cell Biol. 2022, 41, 935–950. [Google Scholar] [CrossRef]
  29. Zhou, M.; Li, Y.J.; Tang, Y.C.; Hao, X.Y.; Xu, W.J.; Xiang, D.X.; Wu, J.Y. Apoptotic bodies for advanced drug delivery and therapy. J. Control. Release 2022, 351, 394–406. [Google Scholar] [CrossRef]
  30. Collado, A.; Gan, L.; Tengbom, J.; Kontidou, E.; Pernow, J.; Zhou, Z. Extracellular vesicles and their non-coding RNA cargos: Emerging players in cardiovascular disease. J. Physiol. 2022; in press. [Google Scholar] [CrossRef]
  31. Ding, X.; Wang, X.; Du, J.; Han, Q.; Zhang, D.; Zhu, H. A systematic review and Meta-analysis of urinary extracellular vesicles proteome in diabetic nephropathy. Front. Endocrinol. 2022, 13, 866252. [Google Scholar] [CrossRef] [PubMed]
  32. Al-Koussa, H.; AlZaim, I.; El-Sabban, M.E. Pathophysiology of Coagulation and Emerging Roles for Extracellular Vesicles in Coagulation Cascades and Disorders. J. Clin. Med. 2022, 11, 4932. [Google Scholar] [CrossRef] [PubMed]
  33. Liu, G.; Yin, X.M. The Role of Extracellular Vesicles in Liver Pathogenesis. Am. J. Pathol. 2022, 192, 1358–1367. [Google Scholar] [CrossRef] [PubMed]
  34. Wei, W.; Pan, Y.; Yang, X.; Chen, Z.; Heng, Y.; Yang, B.; Pu, M.; Zuo, J.; Lai, Z.; Tang, Y.; et al. The Emerging Role of the Interaction of Extracellular Vesicle and Autophagy-Novel Insights into Neurological Disorders. J. Inflamm. Res. 2022, 15, 3395–3407. [Google Scholar] [CrossRef]
  35. Ginini, L.; Billan, S.; Fridman, E.; Gil, Z. Insight into Extracellular Vesicle-Cell Communication: From Cell Recognition to Intracellular Fate. Cells 2022, 11, 1375. [Google Scholar] [CrossRef] [PubMed]
  36. Gul, B.; Syed, F.; Khan, S.; Iqbal, A.; Ahmad, I. Characterization of extracellular vesicles by flow cytometry: Challenges and promises. Micron 2022, 161, 103341. [Google Scholar] [CrossRef] [PubMed]
  37. Awoyemi, T.; Iaccarino, D.A.; Motta-Mejia, C.; Raiss, S.; Kandzija, N.; Zhang, W.; Vatish, M. Neuropilin-1 is uniquely expressed on small syncytiotrophoblast extracellular vesicles but not on medium/large vesicles from preeclampsia and normal placentae. Biochem. Biophys. Res. Commun. 2022, 619, 151–158. [Google Scholar] [CrossRef]
  38. Erdbrügger, U.; Blijdorp, C.J.; Bijnsdorp, I.V.; Borràs, F.E.; Burger, D.; Bussolati, B.; Byrd, J.B.; Clayton, A.; Dear, J.W.; Falcón-Pérez, J.M.; et al. Urinary extracellular vesicles: A position paper by the Urine Task Force of the International Society for Extracellular Vesicles. J. Extracell. Vesicles 2021, 10, e12093. [Google Scholar] [CrossRef]
  39. Börger, V.; Weiss, D.J.; Anderson, J.D.; Borràs, F.E.; Bussolati, B.; Carter, D.R.F.; Dominici, M.; Falcón-Pérez, J.M.; Gimona, M.; Hill, A.F.; et al. International Society for Extracellular Vesicles and International Society for Cell and Gene Therapy statement on extracellular vesicles from mesenchymal stromal cells and other cells: Considerations for potential therapeutic agents to suppress coronavirus disease-19. Cytotherapy 2020, 22, 482–485. [Google Scholar] [CrossRef]
  40. Zhang, H.; Freitas, D.; Kim, H.S.; Fabijanic, K.; Li, Z.; Chen, H.; Mark, M.T.; Molina, H.; Martin, A.B.; Bojmar, L.; et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat. Cell Biol. 2018, 20, 332–343. [Google Scholar] [CrossRef]
  41. Jeppesen, D.K.; Fenix, A.M.; Franklin, J.L.; Higginbotham, J.N.; Zhang, Q.; Zimmerman, L.J.; Liebler, D.C.; Ping, J.; Liu, Q.; Evans, R.; et al. Reassessment of Exosome Composition. Cell 2019, 17, 428–445.e18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Zhang, Q.; Higginbotham, J.N.; Jeppesen, D.K.; Yang, Y.P.; Li, W.; McKinley, E.T.; Graves-Deal, R.; Ping, J.; Britain, C.M.; Dorsett, K.A.; et al. Transfer of Functional Cargo in Exomeres. Cell Rep. 2019, 27, 940–954.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. 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]
  44. Zhang, Q.; Jeppesen, D.K.; Higginbotham, J.N.; Franklin, J.L.; Crowe, J.E., Jr.; Coffey, R.J. Angiotensin-converting Enzyme 2-containing Small Extracellular Vesicles and Exomeres Bind the Severe Acute Respiratory Syndrome Coronavirus 2 Spike Protein. Gastroenterology 2021, 160, 958–961.e3. [Google Scholar] [CrossRef] [PubMed]
  45. Wu, A.Y.; Sung, Y.C.; Chen, Y.J.; Chou, S.T.; Guo, V.; Chien, J.C.; Ko, J.J.; Yang, A.L.; Huang, H.C.; Chuang, J.C.; et al. Multiresolution Imaging Using Bioluminescence Resonance Energy Transfer Identifies Distinct Biodistribution Profiles of Extracellular Vesicles and Exomeres with Redirected Tropism. Adv. Sci. 2020, 7, 2001467. [Google Scholar] [CrossRef]
  46. Anand, S.; Samuel, M.; Mathivanan, S. Exomeres: A New Member of Extracellular Vesicles Family. Subcell. Biochem. 2021, 97, 89–97. [Google Scholar] [CrossRef]
  47. Bojmar, L.; Kim, H.S.; Tobias, G.C.; Pelissier Vatter, F.A.; Lucotti, S.; Gyan, K.E.; Kenific, C.M.; Wan, Z.; Kim, K.A.; Kim, D.; et al. Extracellular vesicle and particle isolation from human and murine cell lines, tissues, and bodily fluids. STAR Protoc. 2020, 2, 100225. [Google Scholar] [CrossRef]
  48. Liangsupree, T.; Multia, E.; Forssén, P.; Fornstedt, T.; Riekkola, M.L. Kinetics and interaction studies of anti-tetraspanin antibodies and ICAM-1 with extracellular vesicle subpopulations using continuous flow quartz crystal microbalance biosensor. Biosens. Bioelectron. 2022, 206, 114151. [Google Scholar] [CrossRef]
  49. An, H.J.; Cho, H.K.; Song, D.H.; Kee, C. Quantitative analysis of exosomes in the aqueous humor of Korean patients with pseudoexfoliation glaucoma. Sci. Rep. 2022, 12, 12875. [Google Scholar] [CrossRef]
  50. Liu, H.; Tian, Y.; Xue, C.; Niu, Q.; Chen, C.; Yan, X. Analysis of extracellular vesicle DNA at the single-vesicle level by nano-flow cytometry. J. Extracell. Vesicles 2022, 11, e12206. [Google Scholar] [CrossRef]
  51. Droste, M.; Tertel, T.; Jeruschke, S.; Dittrich, R.; Kontopoulou, E.; Walkenfort, B.; Börger, V.; Hoyer, P.F.; Büscher, A.K.; Thakur, B.K.; et al. Single Extracellular Vesicle Analysis Performed by Imaging Flow Cytometry and Nanoparticle Tracking Analysis Evaluate the Accuracy of Urinary Extracellular Vesicle Preparation Techniques Differently. Int. J. Mol. Sci. 2021, 22, 12436. [Google Scholar] [CrossRef] [PubMed]
  52. Yoshida, T.; Hanayama, R. TIM4-Affinity Methods Targeting Phosphatidylserine for Isolation or Detection of Extracellular Vesicles. Methods Mol. Biol. 2022, 2466, 23–36. [Google Scholar] [CrossRef] [PubMed]
  53. Ma, S.; Liu, X.; Yin, J.; Hao, L.; Diao, Y.; Zhong, J. Exosomes and autophagy in ocular surface and retinal diseases: New insights into pathophysiology and treatment. Stem Cell Res. Ther. 2022, 13, 174. [Google Scholar] [CrossRef] [PubMed]
  54. Choezom, D.; Gross, J.C. Neutral sphingomyelinase 2 controls exosome secretion by counteracting V-ATPase-mediated endosome acidification. J. Cell Sci. 2022, 135, jcs259324. [Google Scholar] [CrossRef]
  55. Bischoff, J.P.; Schulz, A.; Morrison, H. The role of exosomes in intercellular and inter-organ communication of the peripheral nervous system. FEBS Lett. 2022, 596, 655–664. [Google Scholar] [CrossRef] [PubMed]
  56. Li, C.C.; Hsu, W.F.; Wo, A.M. Exosomes-Potential for Blood-Based Marker in Alzheimer’s Disease. Acta. Neurol. Taiwan 2022, 31, 1–6. [Google Scholar]
  57. Han, C.; Yang, J.; Sun, J.; Qin, G. Extracellular vesicles in cardiovascular disease: Biological functions and therapeutic implications. Pharmacol. Ther. 2022, 233, 108025. [Google Scholar] [CrossRef] [PubMed]
  58. Stotz, H.U.; Brotherton, D.; Inal, J. Communication is key: Extracellular vesicles as mediators of infection and defence during host-microbe interactions in animals and plants. EMS Microbiol. Rev. 2022, 46, fuab044. [Google Scholar] [CrossRef]
  59. Alptekin, A.; Parvin, M.; Chowdhury, H.I.; Rashid, M.H.; Arbab, A.S. Engineered exosomes for studies in tumor immunology. Immunol. Rev. 2022, 312, 76–102. [Google Scholar] [CrossRef] [PubMed]
  60. Heidarzadeh, M.; Sokullu, E.; Saghati, S.; Karimipour, M.; Rahbarghazi, R. Insights into the Critical Role of Exosomes in the Brain; from Neuronal Activity to Therapeutic Effects. Mol. Neurobiol. 2022, 59, 4453–4465. [Google Scholar] [CrossRef]
  61. Liu, Q.W.; He, Y.; Xu, W.W. Molecular functions and therapeutic applications of exosomal noncoding RNAs in cancer. Exp. Mol. Med. 2022, 54, 216–225. [Google Scholar] [CrossRef]
  62. Safari, B.; Aghazadeh, M.; Davaran, S.; Roshangar, L. Exosome-loaded hydrogels: A new cell-free therapeutic approach for skin regeneration. Eur. J. Pharm. Biopharm. 2022, 171, 50–59. [Google Scholar] [CrossRef] [PubMed]
  63. Powell, J.S.; Gandley, R.E.; Lackner, E.; Dolinish, A.; Ouyang, Y.; Powers, R.W.; Morelli, A.E.; Hubel, C.A.; Sadovsky, Y. Small extracellular vesicles from plasma of women with preeclampsia increase myogenic tone and decrease endothelium-dependent relaxation of mouse mesenteric arteries. Pregnancy Hypertens. 2022, 28, 66–73. [Google Scholar] [CrossRef] [PubMed]
  64. Han, Q.F.; Li, W.J.; Hu, K.S.; Gao, J.; Zhai, W.L.; Yang, J.H.; Zhang, S.J. Exosome biogenesis: Machinery, regulation, and therapeutic implications in cancer. Mol. Cancer 2022, 21, 207. [Google Scholar] [CrossRef] [PubMed]
  65. Krause, G.J.; Diaz, A.; Jafari, M.; Khawaja, R.R.; Agullo-Pascual, E.; Santiago-Fernández, O.; Richards, A.L.; Chen, K.H.; Dmitriev, P.; Sun, Y.; et al. Reduced endosomal microautophagy activity in aging associates with enhanced exocyst-mediated protein secretion. Aging Cell 2022, 21, e13713. [Google Scholar] [CrossRef] [PubMed]
  66. Neves, K.B.; Rios, F.J.; Sevilla-Montero, J.; Montezano, A.C.; Touyz, R.M. Exosomes and the cardiovascular system: Role in cardiovascular health and disease. J. Physiol. 2022; in press. [Google Scholar] [CrossRef]
  67. Rashidi, M.; Bijari, S.; Khazaei, A.H.; Shojaei-Ghahrizjani, F.; Rezakhani, L. The role of milk-derived exosomes in the treatment of diseases. Front. Genet. 2022, 13, 1009338. [Google Scholar] [CrossRef] [PubMed]
  68. Carnino, J.M.; Lee, H. Extracellular vesicles in respiratory disease. Adv. Clin. Chem. 2022, 108, 105–127. [Google Scholar] [CrossRef]
  69. Li, S.; Chen, L. Exosomes in Pathogenesis, Diagnosis, and Treatment of Hepatocellular Carcinoma. Front. Oncol. 2022, 12, 793432. [Google Scholar] [CrossRef] [PubMed]
  70. Hosseini, K.; Ranjbar, M.; Pirpour Tazehkand, A.; Asgharian, P.; Montazersaheb, S.; Tarhriz, V.; Ghasemnejad, T. Evaluation of exosomal non-coding RNAs in cancer using high-throughput sequencing. J. Transl. Med. 2022, 20, 30. [Google Scholar] [CrossRef]
  71. Paramanantham, A.; Asfiya, R.; Das, S.; McCully, G.; Srivastava, A. Extracellular Vesicle (EVs) Associated Non-Coding RNAs in Lung Cancer and Therapeutics. Int. J. Mol. Sci. 2022, 23, 13637. [Google Scholar] [CrossRef] [PubMed]
  72. Ebrahimi, N.; Faghihkhorasani, F.; Fakhr, S.S.; Moghaddam, P.R.; Yazdani, E.; Kheradmand, Z.; Rezaei-Tazangi, F.; Adelian, S.; Mobarak, H.; Hamblin, M.R.; et al. Tumor-derived exosomal non-coding RNAs as diagnostic biomarkers in cancer. Cell Mol. Life Sci. 2022, 79, 572. [Google Scholar] [CrossRef]
  73. Zhou, Y.; Xiao, Z.; Zhu, W. The roles of small extracellular vesicles as prognostic biomarkers and treatment approaches in triple-negative breast cancer. Front. Oncol. 2022, 12, 998964. [Google Scholar] [CrossRef] [PubMed]
  74. Sataer, X.; Qifeng, Z.; Yingying, Z.; Chunhua, H.; Bingzhenga, F.; Zhiran, X.; Wanli, L.; Yuwei, Y.; Shuangfeng, C.; Lingling, W.; et al. Exosomal microRNAs as diagnostic biomarkers and therapeutic applications in neurodegenerative diseases. Neurol. Res. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  75. Dhar, R.; Mukherjee, S.; Mukerjee, N.; Mukherjee, D.; Devi, A.; Ashraf, G.M.; Alserihi, R.F.; Tayeb, H.H.; Hashem, A.M.; Alexiou, A.; et al. Interrelation between extracellular vesicles miRNAs with chronic lung diseases. J. Cell Physiol. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  76. Ozkocak, D.C.; Phan, T.K.; Poon, I.K.H. Translating extracellular vesicle packaging into therapeutic applications. Front. Immunol. 2022, 13, 946422. [Google Scholar] [CrossRef] [PubMed]
  77. Rezaie, J.; Akbari, A.; Rahbarghazi, R. Inhibition of extracellular vesicle biogenesis in tumor cells: A possible way to reduce tumorigenesis. Cell Biochem. Funct. 2022, 40, 248–262. [Google Scholar] [CrossRef]
  78. Moon, B.; Chang, S. Exosome as a Delivery Vehicle for Cancer Therapy. Cells 2022, 11, 316. [Google Scholar] [CrossRef]
  79. Yang, L.; Patel, K.D.; Rathnam, C.; Thangam, R.; Hou, Y.; Kang, H.; Lee, K.B. Harnessing the Therapeutic Potential of Extracellular Vesicles for Biomedical Applications Using Multifunctional Magnetic Nanomaterials. Small 2022, 18, e2104783. [Google Scholar] [CrossRef]
  80. Sharma, S.; Sharma, U. Exosomes in cardiovascular diseases: A blessing or a sin for the mankind. Mol. Cell Biochem. 2022, 477, 833–847. [Google Scholar] [CrossRef]
  81. Sun, K.; Zheng, X.; Jin, H.; Yu, F.; Zhao, W. Exosomes as CNS Drug Delivery Tools and Their Applications. Pharmaceutics 2022, 14, 2252. [Google Scholar] [CrossRef] [PubMed]
  82. Xia, Y.; Yang, R.; Hou, Y.; Wang, H.; Li, Y.; Zhu, J.; Fu, C. Application of mesenchymal stem cell-derived exosomes from different sources in intervertebral disc degeneration. Front. Bioeng. Biotechnol. 2022, 10, 1019437. [Google Scholar] [CrossRef] [PubMed]
  83. Zhou, Z.; Wang, R.; Wang, J.; Hao, Y.; Xie, Q.; Wang, L.; Wang, X. Melatonin pretreatment on exosomes: Heterogeneity, therapeutic effects, and usage. Front. Immunol. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  84. Song, J.; Song, B.; Yuan, L.; Yang, G. Multiplexed strategies toward clinical translation of extracellular vesicles. Theranostics 2022, 12, 6740–6761. [Google Scholar] [CrossRef] [PubMed]
  85. Kim, H.; Kim, D.; Kim, W.; Lee, S.; Gwon, Y.; Park, S.; Kim, J. Therapeutic Strategies and Enhanced Production of Stem Cell-Derived Exosomes for Tissue Regeneration. Tissue Eng. Part. B Rev. 2022; in press. [Google Scholar] [CrossRef]
  86. Liu, X.; Wang, C.; Meng, H.; Liao, S.; Zhang, J.; Guan, Y.; Tian, H.; Peng, J. Research Progress on Exosomes in Osteonecrosis of the Femoral Head. Orthop. Surg. 2022, 14, 1951–1957. [Google Scholar] [CrossRef] [PubMed]
  87. Zhao, Y.; Liu, T.; Zhou, M. Immune-Cell-Derived Exosomes for Cancer Therapy. Mol. Pharm. 2022, 19, 3042–3056. [Google Scholar] [CrossRef]
  88. Boyd-Gibbins, N.; Karagiannis, P.; Hwang, D.W.; Kim, S.I. iPSCs in NK Cell Manufacturing and NKEV Development. Front. Immunol. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  89. Ahmed, L.; Al-Massri, K. New Approaches for Enhancement of the Efficacy of Mesenchymal Stem Cell-Derived Exosomes in Cardiovascular Diseases. Tissue Eng. Regen. Med. 2022; in press. [Google Scholar] [CrossRef]
  90. Rezaie, J.; Nejati, V.; Mahmoodi, M.; Ahmadi, M. Mesenchymal stem cells derived extracellular vesicles: A promising nanomedicine for drug delivery system. Biochem. Pharmacol. 2022, 203, 115167. [Google Scholar] [CrossRef]
  91. Song, Q.; Yu, H.; Han, J.; Lv, J.; Lv, Q.; Yang, H. Exosomes in urological diseases—Biological functions and clinical applications. Cancer Lett. 2022, 544, 215809. [Google Scholar] [CrossRef] [PubMed]
  92. Hade, M.D.; Suire, C.N.; Mossell, J.; Suo, Z. Extracellular vesicles: Emerging frontiers in wound healing. Med. Res. Rev. 2022, 42, 2102–2125. [Google Scholar] [CrossRef] [PubMed]
  93. Yong, T.; Wei, Z.; Gan, L.; Yang, X. Extracellular-Vesicle-Based Drug Delivery Systems for Enhanced Antitumor Therapies through Modulating the Cancer-Immunity Cycle. Adv. Mater. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  94. Abbaszadeh, H.; Ghorbani, F.; Abbaspour-Aghdam, S.; Kamrani, A.; Valizadeh, H.; Nadiri, M.; Sadeghi, A.; Shamsasenjan, K.; Jadidi-Niaragh, F.; Roshangar, L.; et al. Chronic obstructive pulmonary disease and asthma: Mesenchymal stem cells and their extracellular vesicles as potential therapeutic tools. Stem Cell Res. Ther. 2022, 13, 262. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, X.; Zhao, X.; Zhong, Y.; Shen, J.; An, W. Biomimetic Exosomes: A New Generation of Drug Delivery System. Front. Bioeng. Biotechnol. 2022, 10, 865682. [Google Scholar] [CrossRef] [PubMed]
  96. Yang, S.; Wang, J.; Wang, S.; Zhou, A.; Zhao, G.; Li, P. Roles of small extracellular vesicles in the development, diagnosis and possible treatment strategies for hepatocellular carcinoma (Review). Int. J. Oncol. 2022, 61, 91. [Google Scholar] [CrossRef]
  97. Moayedfard, Z.; Sani, F.; Alizadeh, A.; Bagheri Lankarani, K.; Zarei, M.; Azarpira, N. The role of the immune system in the pathogenesis of NAFLD and potential therapeutic impacts of mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res. Ther. 2022, 13, 242. [Google Scholar] [CrossRef]
  98. Zhao, J.; An, Q.; Zhu, X.; Yang, B.; Gao, X.; Niu, Y.; Zhang, L.; Xu, K.; Ma, D. Research status and future prospects of extracellular vesicles in primary Sjögren’s syndrome. Stem Cell Res. Ther. 2022, 13, 230. [Google Scholar] [CrossRef]
  99. Zhou, X.; Cao, H.; Guo, J.; Yuan, Y.; Ni, G. Effects of BMSC-Derived EVs on Bone Metabolism. Pharmaceutics 2022, 14, 1012. [Google Scholar] [CrossRef]
  100. Xie, S.; Zhang, Q.; Jiang, L. Current Knowledge on Exosome Biogenesis, Cargo-Sorting Mechanism and Therapeutic Implications. Membranes 2022, 12, 498. [Google Scholar] [CrossRef]
  101. Xiong, Y.; Song, J.; Huang, X.; Pan, Z.; Goldbrunner, R.; Stavrinou, L.; Lin, S.; Hu, W.; Zheng, F.; Stavrinou, P. Exosomes Derived From Mesenchymal Stem Cells: Novel Effects in the Treatment of Ischemic Stroke. Front. Neurosci. 2022, 16, 899887. [Google Scholar] [CrossRef] [PubMed]
  102. Malekian, F.; Shamsian, A.; Kodam, S.P.; Ullah, M. Exosome engineering for efficient and targeted drug delivery: Current status and future perspective. J. Physiol. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  103. Goutas, D.; Pergaris, A.; Goutas, N.; Theocharis, S. Utilizing Exosomal-EPHs/Ephrins as Biomarkers and as a Potential Platform for Targeted Delivery of Therapeutic Exosomes. Int. J. Mol. Sci. 2022, 23, 3551. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, Y.; Dong, B.; Huang, L.; Zhou, J.; Huang, H. Research progress on the role and mechanism of action of exosomes in autoimmune thyroid disease. Int. Rev. Immunol. 2022; in press. [Google Scholar] [CrossRef]
  105. Chen, L.; Wang, L.; Zhu, L.; Xu, Z.; Liu, Y.; Li, Z.; Zhou, J.; Luo, F. Exosomes as Drug Carriers in Anti-Cancer Therapy. Front. Cell Dev. Biol. 2022, 10, 728616. [Google Scholar] [CrossRef] [PubMed]
  106. Li, Q.C.; Li, C.; Zhang, W.; Pi, W.; Han, N. Potential Effects of Exosomes and their MicroRNA Carrier on Osteoporosis. Curr. Pharm. Des. 2022, 28, 899–909. [Google Scholar] [CrossRef]
  107. Jiang, L.; Chen, W.; Ye, J.; Wang, Y. Potential Role of Exosomes in Ischemic Stroke Treatment. Biomolecules 2022, 12, 115. [Google Scholar] [CrossRef]
  108. Yang, J.; Zhang, L. The roles and therapeutic approaches of MSC-derived exosomes in colorectal cancer. Clin. Transl. Oncol. 2022, 24, 959–967. [Google Scholar] [CrossRef]
  109. He, J.; Ren, W.; Wang, W.; Han, W.; Jiang, L.; Zhang, D.; Guo, M. Exosomal targeting and its potential clinical application. Drug Deliv. Transl. Res. 2022, 12, 2385–2402. [Google Scholar] [CrossRef]
  110. Ren, J.; He, W.; Zheng, L.; Duan, H. From structures to functions: Insights into exosomes as promising drug delivery vehicles. Biomater. Sci. 2016, 4, 910–921. [Google Scholar] [CrossRef]
  111. Bie, N.; Yong, T.; Wei, Z.; Gan, L.; Yang, X. Extracellular vesicles for improved tumor accumulation and penetration. Adv. Drug Deliv. Rev. 2022, 188, 114450. [Google Scholar] [CrossRef] [PubMed]
  112. Matsuzaka, Y.; Yashiro, R. Molecular Docking and Intracellular Translocation of Extracellular Vesicles for Efficient Drug Delivery. Int. J. Mol. Sci. 2022, 23, 12971. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, Y.; Li, J.; Gao, W.; Xie, N. Exosomes as Anticancer Drug Delivery Vehicles: Prospects and Challenges. Front. Biosci. (Landmark Ed) 2022, 27, 293. [Google Scholar] [CrossRef]
  114. Yang, F.; Wang, M.; Guan, X. Exosomes and mimics as novel delivery platform for cancer therapy. Front. Pharmacol. 2022, 13, 1001417. [Google Scholar] [CrossRef]
  115. Dalmizrak, A.; Dalmizrak, O. Mesenchymal stem cell-derived exosomes as new tools for delivery of miRNAs in the treatment of cancer. Front. Bioeng. Biotechnol. 2022, 10, 956563. [Google Scholar] [CrossRef] [PubMed]
  116. Muhammad, S.A.; Mohammed, J.S.; Rabiu, S. Exosomes as Delivery Systems for Targeted Tumour Therapy: A Systematic Review and Meta-analysis of In vitro Studies. Pharm. Nanotechnol. 2022; in press. [Google Scholar] [CrossRef]
  117. Rezaie, J.; Etemadi, T.; Feghhi, M. The distinct roles of exosomes in innate immune responses and therapeutic applications in cancer. Eur. J. Pharmacol. 2022, 933, 175292. [Google Scholar] [CrossRef]
  118. Rezaie, J.; Feghhi, M.; Etemadi, T. A review on exosomes application in clinical trials: Perspective, questions, and challenges. Cell Commun. Signal. 2022, 20, 145. [Google Scholar] [CrossRef] [PubMed]
  119. Mansourabadi, A.H.; Aghamajidi, A.; Faraji, F.; Taghizadeh, S.; Mohamed Khosroshahi, L.; Bahramkiya, M.; Azimi, M. Mesenchymal stem cells- derived exosomes inhibit the expression of Aquaporin-5 and EGFR in HCT-116 human colorectal carcinoma cell line. BMC Mol. Cell Biol. 2022, 23, 40. [Google Scholar] [CrossRef] [PubMed]
  120. Li, Y.; Meng, L.; Li, B.; Li, Y.; Shen, T.; Zhao, B. The Exosome Journey: From Biogenesis to Regulation and Function in Cancers. J. Oncol. 2022; in press. [Google Scholar] [CrossRef]
  121. Liu, H.; Huang, Y.; Huang, M.; Huang, Z.; Wang, Q.; Qing, L.; Li, L.; Xu, S.; Jia, B. Current Status, Opportunities, and Challenges of Exosomes in Oral Cancer Diagnosis and Treatment. Int. J. Nanomed. 2022, 17, 2679–2705. [Google Scholar] [CrossRef] [PubMed]
  122. Cai, Y.; Zhang, L.; Zhang, Y.; Lu, R. Plant-Derived Exosomes as a Drug-Delivery Approach for the Treatment of Inflammatory Bowel Disease and Colitis-Associated Cancer. Pharmaceutics 2022, 14, 822. [Google Scholar] [CrossRef] [PubMed]
  123. Ansari, M.A.; Thiruvengadam, M.; Venkidasamy, B.; Alomary, M.N.; Salawi, A.; Chung, I.M.; Shariati, M.A.; Rebezov, M. Exosome-based nanomedicine for cancer treatment by targeting inflammatory pathways: Current status and future perspectives. Semin. Cancer Biol. 2022, 86, 678–696. [Google Scholar] [CrossRef]
  124. Vahabi, A.; Rezaie, J.; Hassanpour, M.; Panahi, Y.; Nemati, M.; Rasmi, Y.; Nemati, M. Tumor Cells-derived exosomal CircRNAs: Novel cancer drivers, molecular mechanisms, and clinical opportunities. Biochem. Pharmacol. 2022, 200, 115038. [Google Scholar] [CrossRef]
  125. Jiang, J.; Li, J.; Zhou, X.; Zhao, X.; Huang, B.; Qin, Y. Exosomes Regulate the Epithelial-Mesenchymal Transition in Cancer. Front. Oncol. 2022, 12, 864980. [Google Scholar] [CrossRef]
  126. Xu, Y.; Feng, K.; Zhao, H.; Di, L.; Wang, L.; Wang, R. Tumor-derived extracellular vesicles as messengers of natural products in cancer treatment. Theranostics 2022, 12, 1683–1714. [Google Scholar] [CrossRef] [PubMed]
  127. Srivastava, A.; Rathore, S.; Munshi, A.; Ramesh, R. Organically derived exosomes as carriers of anticancer drugs and imaging agents for cancer treatment. Semin. Cancer Biol. 2022, 86, 80–100. [Google Scholar] [CrossRef]
  128. Ferreira, D.; Moreira, J.N.; Rodrigues, L.R. New advances in exosome-based targeted drug delivery systems. Crit. Rev. Oncol. Hematol. 2022, 172, 103628. [Google Scholar] [CrossRef]
  129. St-Denis-Bissonnette, F.; Khoury, R.; Mediratta, K.; El-Sahli, S.; Wang, L.; Lavoie, J.R. Applications of Extracellular Vesicles in Triple-Negative Breast Cancer. Cancers 2022, 14, 451. [Google Scholar] [CrossRef]
  130. Liu, Z.; Zhang, H.; Liu, S.; Hou, Y.; Chi, G. The Dual Role of Astrocyte-Derived Exosomes and Their Contents in the Process of Alzheimer’s Disease. J. Alzheimer’s Dis. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  131. Delcorte, O.; Degosserie, J.; Pierreux, C.E. Role of Extracellular Vesicles in Thyroid Physiology and Diseases: Implications for Diagnosis and Treatment. Biomedicines 2022, 10, 2585. [Google Scholar] [CrossRef] [PubMed]
  132. Nila, I.S.; Sumsuzzman, D.M.; Khan, Z.A.; Jung, J.H.; Kazema, A.S.; Kim, S.J.; Hong, Y. Identification of exosomal biomarkers and its optimal isolation and detection method for the diagnosis of Parkinson’s disease: A systematic review and meta-analysis. Ageing Res. Rev. 2022, 82, 101764. [Google Scholar] [CrossRef]
  133. Zhou, H.; Wan, H.; Feng, Y.; Zhu, L.; Mi, Y. The diagnostic role and mechanistic functions of exosomal lncRNAs in prostate cancer. Clin. Transl. Oncol. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  134. Wei, F.; Li, Y. The emerging roles of exosome-derived noncoding RNAs in the tumor immune microenvironment and their future applications. Biomed. Pharmacother. 2022, 156, 113863. [Google Scholar] [CrossRef] [PubMed]
  135. Li, M.; Cai, H.; Deng, R.; Cheng, J.; Shi, Y. Effects of exosomes on tumor immunomodulation and their potential clinical applications (Review). Int. J. Oncol. 2022, 61, 147. [Google Scholar] [CrossRef] [PubMed]
  136. Fang, J.; Zhang, Y.; Chen, D.; Zheng, Y.; Jiang, J. Exosomes and Exosomal Cargos: A Promising World for Ventricular Remodeling Following Myocardial Infarction. Int. J. Nanomedicine 2022, 17, 4699–4719. [Google Scholar] [CrossRef]
  137. Hullin-Matsuda, F.; Colosetti, P.; Rabia, M.; Luquain-Costaz, C.; Delton, I. Exosomal lipids from membrane organization to biomarkers: Focus on an endolysosomal-specific lipid. Biochimie, 2022; in press. [Google Scholar] [CrossRef]
  138. Xiang, H.; Zhang, C.; Xiong, J. Emerging role of extracellular vesicles in kidney diseases. Front. Pharmacol. 2022, 13, 985030. [Google Scholar] [CrossRef]
  139. Qian, K.; Fu, W.; Li, T.; Zhao, J.; Lei, C.; Hu, S. The roles of small extracellular vesicles in cancer and immune regulation and translational potential in cancer therapy. J. Exp. Clin. Cancer Res. 2022, 41, 286. [Google Scholar] [CrossRef]
  140. Sadu, L.; Krishnan, R.H.; Akshaya, R.L.; Das, U.R.; Satishkumar, S.; Selvamurugan, N. Exosomes in bone remodeling and breast cancer bone metastasis. Prog. Biophys. Mol. Biol. 2022, 175, 120–130. [Google Scholar] [CrossRef]
  141. Yi, X.; Chen, J.; Huang, D.; Feng, S.; Yang, T.; Li, Z.; Wang, X.; Zhao, M.; Wu, J.; Zhong, T. Current perspectives on clinical use of exosomes as novel biomarkers for cancer diagnosis. Front. Oncol. 2022, 12, 966981. [Google Scholar] [CrossRef]
  142. Wang, X.; Tian, L.; Lu, J.; Ng, I.O. Exosomes and cancer—Diagnostic and prognostic biomarkers and therapeutic vehicle. Oncogenesis 2022, 11, 54. [Google Scholar] [CrossRef] [PubMed]
  143. Ahmadi, M.; Hassanpour, M.; Rezaie, J. Engineered extracellular vesicles: A novel platform for cancer combination therapy and cancer immunotherapy. Life Sci. 2022, 308, 120935. [Google Scholar] [CrossRef] [PubMed]
  144. Karami Fath, M.; Azami, J.; Jaafari, N.; Akbari Oryani, M.; Jafari, N.; Karim Poor, A.; Azargoonjahromi, A.; Nabi-Afjadi, M.; Payandeh, Z.; Zalpoor, H.; et al. Exosome application in treatment and diagnosis of B-cell disorders: Leukemias, multiple sclerosis, and arthritis rheumatoid. Cell Mol. Biol. Lett. 2022, 27, 74. [Google Scholar] [CrossRef]
  145. Pan, S.; Chen, Y.; Yan, J.; Li, F.; Chen, X.; Xu, X.; Xing, H. The emerging roles and mechanisms of exosomal non-coding RNAs in the mutual regulation between adipose tissue and other related tissues in obesity and metabolic diseases. Front. Endocrinol. 2022, 13, 975334. [Google Scholar] [CrossRef]
  146. Ormazabal, V.; Nair, S.; Carrión, F.; Mcintyre, H.D.; Salomon, C. The link between gestational diabetes and cardiovascular diseases: Potential role of extracellular vesicles. Cardiovasc. Diabetol. 2022, 21, 174. [Google Scholar] [CrossRef] [PubMed]
  147. Bano, A.; Vats, R.; Yadav, P.; Bhardwaj, R. Exosomics in oral cancer diagnosis, prognosis, and therapeutics—An emergent and imperative non-invasive natural nanoparticle-based approach. Crit. Rev. Oncol. Hematol. 2022, 178, 103799. [Google Scholar] [CrossRef] [PubMed]
  148. Hosseinikhah, S.M.; Gheybi, F.; Moosavian, S.A.; Shahbazi, M.A.; Jaafari, M.R.; Sillanpää, M.; Kesharwani, P.; Alavizadeh, S.H.; Sahebkar, A. Role of exosomes in tumour growth, chemoresistance and immunity: State-of-the-art. J. Drug Target, 2022; in press. [Google Scholar] [CrossRef]
  149. Khan, F.H.; Reza, M.J.; Shao, Y.F.; Perwez, A.; Zahra, H.; Dowlati, A.; Abbas, A. Role of exosomes in lung cancer: A comprehensive insight from immunomodulation to theragnostic applications. Biochim. Biophys. Acta. Rev. Cancer 2022, 1877, 188776. [Google Scholar] [CrossRef]
  150. Zelli, V.; Compagnoni, C.; Capelli, R.; Corrente, A.; Di Vito Nolfi, M.; Zazzeroni, F.; Alesse, E.; Tessitore, A. Role of exosomal microRNAs in cancer therapy and drug resistance mechanisms: Focus on hepatocellular carcinoma. Front. Oncol. 2022, 12, 940056. [Google Scholar] [CrossRef] [PubMed]
  151. Li, J.; Zhang, Y.; Luo, B. Effects of Exosomal Viral Components on the Tumor Microenvironment. Cancers 2022, 14, 3552. [Google Scholar] [CrossRef]
  152. Liu, C.; Wang, Y.; Li, L.; He, D.; Chi, J.; Li, Q.; Wu, Y.; Zhao, Y.; Zhang, S.; Wang, L.; et al. Engineered extracellular vesicles and their mimetics for cancer immunotherapy. J. Control. Release 2022, 349, 679–698. [Google Scholar] [CrossRef] [PubMed]
  153. Gulati, R.; Nandi, D.; Sarkar, K.; Venkataraman, P.; Ramkumar, K.M.; Ranjan, P.; Janardhanan, R. Exosomes as Theranostic Targets: Implications for the Clinical Prognosis of Aggressive Cancers. Front. Mol. Biosci. 2022, 9, 890768. [Google Scholar] [CrossRef] [PubMed]
  154. Paskeh, M.D.A.; Entezari, M.; Mirzaei, S.; Zabolian, A.; Saleki, H.; Naghdi, M.J.; Sabet, S.; Khoshbakht, M.A.; Hashemi, M.; Hushmandi, K.; et al. Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. J. Hematol. Oncol. 2022, 15, 83. [Google Scholar] [CrossRef] [PubMed]
  155. Fan, Y.; Chen, Z.; Zhang, M. Role of exosomes in the pathogenesis, diagnosis, and treatment of central nervous system diseases. J. Transl. Med. 2022, 20, 291. [Google Scholar] [CrossRef]
  156. Di Bella, M.A. Overview and Update on Extracellular Vesicles: Considerations on Exosomes and Their Application in Modern Medicine. Biology 2022, 11, 804. [Google Scholar] [CrossRef] [PubMed]
  157. Matsuzaka, Y.; Yashiro, R. Extracellular Vesicles as Novel Drug-Delivery Systems through Intracellular Communications. Membranes 2022, 12, 550. [Google Scholar] [CrossRef]
  158. Guo, Z.Y.; Tang, Y.; Cheng, Y.C. Exosomes as Targeted Delivery Drug System: Advances in Exosome Loading, Surface Functionalization and Potential for Clinical Application. Curr. Drug Deliv. 2022; in press. [Google Scholar] [CrossRef]
  159. Wang, H.; You, Y.; Zhu, X. The Role of Exosomes in the Progression and Therapeutic Resistance of Hematological Malignancies. Front. Oncol. 2022, 12, 887518. [Google Scholar] [CrossRef]
  160. Gurunathan, S.; Kang, M.H.; Song, H.; Kim, N.H.; Kim, J.H. The role of extracellular vesicles in animal reproduction and diseases. J. Anim. Sci. Biotechnol. 2022, 13, 62. [Google Scholar] [CrossRef]
  161. Lee, C.; Han, J.; Jung, Y. Pathological Contribution of Extracellular Vesicles and Their MicroRNAs to Progression of Chronic Liver Disease. Biology 2022, 1, 637. [Google Scholar] [CrossRef] [PubMed]
  162. Asemani, Y.; Najafi, S.; Ezzatifar, F.; Zolbanin, N.M.; Jafari, R. Recent highlights in the immunomodulatory aspects of Treg cell-derived extracellular vesicles: Special emphasis on autoimmune diseases and transplantation. Cell Biosci. 2022, 12, 67. [Google Scholar] [CrossRef] [PubMed]
  163. Liu, Q.; Zhang, X.; Zhang, J. Exosome-Based Nanoplatforms: The Emerging Tools for Breast Cancer Therapy. Front. Oncol. 2022, 12, 898605. [Google Scholar] [CrossRef] [PubMed]
  164. Fang, Y.; Dai, X. Emerging Roles of Extracellular Non-Coding RNAs in Vascular Diseases. J. Cardiovasc. Transl. Res. 2022, 15, 492–499. [Google Scholar] [CrossRef]
  165. Bazzoni, R.; Tanasi, I.; Turazzi, N.; Krampera, M. Update on the Role and Utility of Extracellular Vesicles in Hematological Malignancies. Stem Cells 2022, 40, 619–629. [Google Scholar] [CrossRef]
  166. Liu, K.; Gao, X.; Kang, B.; Liu, Y.; Wang, D.; Wang, Y. The Role of Tumor Stem Cell Exosomes in Cancer Invasion and Metastasis. Front. Oncol. 2022, 12, 836548. [Google Scholar] [CrossRef]
  167. Kumari, M.; Anji, A. Small but Mighty-Exosomes, Novel Intercellular Messengers in Neurodegeneration. Biology 2022, 11, 413. [Google Scholar] [CrossRef]
  168. Al Halawani, A.; Mithieux, S.M.; Yeo, G.C.; Hosseini-Beheshti, E.; Weiss, A.S. Extracellular Vesicles: Interplay with the Extracellular Matrix and Modulated Cell Responses. Int. J. Mol. Sci. 2022, 23, 3389. [Google Scholar] [CrossRef]
  169. Xu, K.; Jin, Y.; Li, Y.; Huang, Y.; Zhao, R. Recent Progress of Exosome Isolation and Peptide Recognition-Guided Strategies for Exosome Research. Front. Chem. 2022, 10, 844124. [Google Scholar] [CrossRef]
  170. Zhao, K.; Li, X.; Shi, Y.; Lu, Y.; Qiu, P.; Deng, Z.; Yao, W.; Wang, J. Exosomes in the tumor microenvironment of cholangiocarcinoma: Current status and future perspectives. J. Transl. Med. 2022, 20, 117. [Google Scholar] [CrossRef]
  171. Zhang, X.; Xu, D.; Song, Y.; He, R.; Wang, T. Research Progress in the Application of Exosomes in Immunotherapy. Front. Immunol. 2022, 13, 731516. [Google Scholar] [CrossRef] [PubMed]
  172. Keshtkar, S.; Kaviani, M.; Soleimanian, S.; Azarpira, N.; Asvar, Z.; Pakbaz, S. Stem Cell-Derived Exosome as Potential Therapeutics for Microbial Diseases. Front. Microbiol. 2022, 12, 786111. [Google Scholar] [CrossRef] [PubMed]
  173. Kowalczyk, A.; Wrzecińska, M.; Czerniawska-Piątkowska, E.; Kupczyński, R. Exosomes—Spectacular role in reproduction. Biomed. Pharmacother. 2022, 148, 112752. [Google Scholar] [CrossRef]
  174. Xue, D.; Han, J.; Liang, Z.; Jia, L.; Liu, Y.; Tuo, H.; Peng, Y. Current Perspectives on the Unique Roles of Exosomes in Drug Resistance of Hepatocellular Carcinoma. J. Hepatocell. Carcinoma 2022, 9, 99–112. [Google Scholar] [CrossRef]
  175. Dehkordi, N.R.; Dehkordi, N.R.; Farjoo, M.H. Therapeutic properties of stem cell-derived exosomes in ischemic heart disease. Eur. J. Pharmacol. 2022, 920, 174839. [Google Scholar] [CrossRef] [PubMed]
  176. Pan, Y.; Tan, W.F.; Yang, M.Q.; Li, J.Y.; Geller, D.A. The therapeutic potential of exosomes derived from different cell sources in liver diseases. Am. J. Physiol. Gastrointest. Liver Physiol. 2022, 322, G397–G404. [Google Scholar] [CrossRef]
  177. Whittle, K.; Kao, S.; Clarke, S.; Grau, G.E.R.; Hosseini-Beheshti, E. Exploring the role of extracellular vesicles and their protein cargo in lung cancer metastasis: A review. Crit. Rev. Oncol. Hematol. 2022, 171, 103603. [Google Scholar] [CrossRef]
  178. Zhang, H.; Xing, J.; Dai, Z.; Wang, D.; Tang, D. Exosomes: The key of sophisticated cell-cell communication and targeted metastasis in pancreatic cancer. Cell Commun. Signal. 2022, 20, 9. [Google Scholar] [CrossRef]
  179. Zhou, Z.W.; Zheng, W.; Xiang, Z.; Ye, C.S.; Yin, Q.Q.; Wang, S.H.; Xu, C.A.; Wu, W.H.; Hui, T.C.; Wu, Q.Q.; et al. Clinical implications of exosome-derived noncoding RNAs in liver. Lab. Investig. 2022, 102, 464–473. [Google Scholar] [CrossRef]
  180. Brown, P.A. Differential and targeted vesiculation: Pathologic cellular responses to elevated arterial pressure. Mol. Cell Biochem. 2022, 477, 1023–1040. [Google Scholar] [CrossRef]
  181. Gholami Farashah, M.S.; Javadi, M.; Mohammadi, A.; Soleimani Rad, J.; Shakouri, S.K.; Roshangar, L. Bone marrow mesenchymal stem cell’s exosomes as key nanoparticles in osteogenesis and bone regeneration: Specific capacity based on cell type. Mol. Biol. Rep. 2022; in press. [Google Scholar] [CrossRef]
  182. Taniguchi, M.; Nagaya, S.; Yuyama, K.; Kotani, A.; Igarashi, Y.; Okazaki, T. Ceramide Metabolism Regulated by Sphingomyelin Synthase 2 Is Associated with Acquisition of Chemoresistance via Exosomes in Human Leukemia Cells. Int. J. Mol. Sci. 2022, 23, 10648. [Google Scholar] [CrossRef] [PubMed]
  183. Schumacher, F.; Carpinteiro, A.; Edwards, M.J.; Wilson, G.C.; Keitsch, S.; Soddemann, M.; Wilker, B.; Kleuser, B.; Becker, K.A.; Müller, C.P.; et al. Stress induces major depressive disorder by a neutral sphingomyelinase 2-mediated accumulation of ceramide-enriched exosomes in the blood plasma. J. Mol. Med. 2022, 100, 1493–1508. [Google Scholar] [CrossRef] [PubMed]
  184. Arya, S.B.; Chen, S.; Jordan-Javed, F.; Parent, C.A. Ceramide-rich microdomains facilitate nuclear envelope budding for non-conventional exosome formation. Nat. Cell Biol. 2022, 24, 1019–1028. [Google Scholar] [CrossRef] [PubMed]
  185. Habibi, J.; DeMarco, V.G.; Hulse, J.L.; Hayden, M.R.; Whaley-Connell, A.; Hill, M.A.; Sowers, J.R.; Jia, G. Inhibition of sphingomyelinase attenuates diet—Induced increases in aortic stiffness. J. Mol. Cell Cardiol. 2022, 167, 32–39. [Google Scholar] [CrossRef]
  186. Melero-Fernandez de Mera, R.M.; Villaseñor, A.; Rojo, D.; Carrión-Navarro, J.; Gradillas, A.; Ayuso-Sacido, A.; Barbas, C. Ceramide Composition in Exosomes for Characterization of Glioblastoma Stem-Like Cell Phenotypes. Front. Oncol. 2022, 11, 788100. [Google Scholar] [CrossRef] [PubMed]
  187. Nakao, Y.; Fukushima, M.; Mauer, A.S.; Liao, C.Y.; Ferris, A.; Dasgupta, D.; Heppelmann, C.J.; Vanderboom, P.M.; Saraswat, M.; Pandey, A.; et al. A Comparative Proteomic Analysis of Extracellular Vesicles Associated With Lipotoxicity. Front. Cell Dev. Biol. 2021, 9, 735001. [Google Scholar] [CrossRef] [PubMed]
  188. Abesekara, M.S.; Chau, Y. Recent advances in surface modification of micro- and nano-scale biomaterials with biological membranes and biomolecules. Front. Bioeng. Biotechnol. 2022, 10, 972790. [Google Scholar] [CrossRef]
  189. Canning, P.; Alwan, A.; Khalil, F.; Zhang, Y.; Opara, E.C. Perspectives and Challenges on the Potential Use of Exosomes in Bioartificial Pancreas Engineering. Ann. Biomed. Eng. 2022, 50, 1177–1186. [Google Scholar] [CrossRef]
  190. Sundaram, T.S.; Giromini, C.; Rebucci, R.; Pistl, J.; Bhide, M.; Baldi, A. Role of omega-3 polyunsaturated fatty acids, citrus pectin, and milk-derived exosomes on intestinal barrier integrity and immunity in animals. J. Anim. Sci. Biotechnol. 2022, 13, 40. [Google Scholar] [CrossRef]
  191. Fang, Z.; Ding, Y.; Xue, Z.; Li, P.; Li, J.; Li, F. Roles of exosomes as drug delivery systems in cancer immunotherapy: A mini-review. Discov. Oncol. 2022, 13, 74. [Google Scholar] [CrossRef] [PubMed]
  192. Eguchi, T.; Sheta, M.; Fujii, M.; Calderwood, S.K. Cancer extracellular vesicles, tumoroid models, and tumor microenvironment. Semin. Cancer Biol. 2022, 86, 112–126. [Google Scholar] [CrossRef] [PubMed]
  193. Liu, Y.; Gu, Y.; Cao, X. The exosomes in tumor immunity. Oncoimmunology 2015, 4, e1027472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Szeliski, K.; Drewa, T.; Pokrywczyńska, M. Small extracellular vesicles as a multicomponent biomarker platform in urinary tract carcinomas. Front. Mol. Biosci. 2022, 9, 916666. [Google Scholar] [CrossRef]
  195. Shang, X.; Fang, Y.; Xin, W.; You, H. The Application of Extracellular Vesicles Mediated miRNAs in Osteoarthritis: Current Knowledge and Perspective. J. Inflamm. Res. 2022, 15, 2583–2599. [Google Scholar] [CrossRef] [PubMed]
  196. Xu, Z.; Chen, Y.; Ma, L.; Chen, Y.; Liu, J.; Guo, Y.; Yu, T.; Zhang, L.; Zhu, L.; Shu, Y. Role of exosomal non-coding RNAs from tumor cells and tumor-associated macrophages in the tumor microenvironment. Mol. Ther. 2022, 30, 3133–3154. [Google Scholar] [CrossRef]
  197. Huang, Z.; Keramat, S.; Izadirad, M.; Chen, Z.S.; Soukhtanloo, M. The Potential Role of Exosomes in the Treatment of Brain Tumors, Recent Updates and Advances. Front. Oncol. 2022, 12, 869929. [Google Scholar] [CrossRef]
  198. Wang, W.; Hao, L.P.; Song, H.; Chu, X.Y.; Wang, R. The Potential Roles of Exosomal Non-Coding RNAs in Hepatocellular Carcinoma. Front. Oncol. 2022, 12, 790916. [Google Scholar] [CrossRef]
  199. Javadi, M.; Rad, J.S.; Farashah, M.S.G.; Roshangar, L. An Insight on the Role of Altered Function and Expression of Exosomes and MicroRNAs in Female Reproductive Diseases. Reprod. Sci. 2022, 29, 1395–1407. [Google Scholar] [CrossRef]
  200. Weaver, A.M.; Patton, J.G. Argonautes in Extracellular Vesicles: Artifact or Selected Cargo? Cancer Res. 2020, 80, 379–381. [Google Scholar] [CrossRef]
  201. O’Grady, T.; Njock, M.S.; Lion, M.; Bruyr, J.; Mariavelle, E.; Galvan, B.; Boeckx, A.; Struman, I.; Dequiedt, F. Sorting and packaging of RNA into extracellular vesicles shape intracellular transcript levels. BMC Biol. 2022, 20, 72. [Google Scholar] [CrossRef]
  202. Zhou, X.; Wang, L.; Zou, W.; Chen, X.; Roizman, B.; Zhou, G.G. hnRNPA2B1 Associated with Recruitment of RNA into Exosomes Plays a Key Role in Herpes Simplex Virus 1 Release from Infected Cells. J. Virol. 2020, 94, e00367-20. [Google Scholar] [CrossRef]
  203. Villarroya-Beltri, C.; Gutiérrez-Vázquez, C.; Sánchez-Cabo, F.; Pérez-Hernández, D.; Vázquez, J.; Martin-Cofreces, N.; Martinez-Herrera, D.J.; Pascual-Montano, A.; Mittelbrunn, M.; Sánchez-Madrid, F. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat. Commun. 2013, 4, 2980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Robinson, H.; Ruelcke, J.E.; Lewis, A.; Bond, C.S.; Fox, A.H.; Bharti, V.; Wani, S.; Cloonan, N.; Lai, A.; Margolin, D.; et al. Caveolin-1-driven membrane remodelling regulates hnRNPK-mediated exosomal microRNA sorting in cancer. Clin. Transl. Med. 2021, 11, e381. [Google Scholar] [CrossRef]
  205. Wozniak, A.L.; Adams, A.; King, K.E.; Dunn, W.; Christenson, L.K.; Hung, W.T.; Weinman, S.A. The RNA binding protein FMR1 controls selective exosomal miRNA cargo loading during inflammation. J. Cell Biol. 2020, 219, e201912074. [Google Scholar] [CrossRef] [PubMed]
  206. Wu, B.; Su, S.; Patil, D.P.; Liu, H.; Gan, J.; Jaffrey, S.R.; Ma, J. Molecular basis for the specific and multivariant recognitions of RNA substrates by human hnRNP A2/B1. Nat. Commun. 2018, 9, 420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Santangelo, L.; Giurato, G.; Cicchini, C.; Montaldo, C.; Mancone, C.; Tarallo, R.; Battistelli, C.; Alonzi, T.; Weisz, A.; Tripodi, M. The RNA-Binding Protein SYNCRIP Is a Component of the Hepatocyte Exosomal Machinery Controlling MicroRNA Sorting. Cell Rep. 2016, 17, 799–808. [Google Scholar] [CrossRef] [Green Version]
  208. Hao, S.; Bai, O.; Yuan, J.; Qureshi, M.; Xiang, J. Dendritic cell-derived exosomes stimulate stronger CD8+ CTL responses and antitumor immunity than tumor cell-derived exosomes. Cell Mol. Immunol. 2006, 3, 205–211. [Google Scholar] [PubMed]
  209. Staubach, S.; Bauer, F.N.; Tertel, T.; Börger, V.; Stambouli, O.; Salzig, D.; Giebel, B. Scaled preparation of extracellular vesicles from conditioned media. Adv. Drug Deliv. Rev. 2021, 177, 113940. [Google Scholar] [CrossRef] [PubMed]
  210. Bogatcheva, N.V.; Coleman, M.E. Conditioned Medium of Mesenchymal Stromal Cells: A New Class of Therapeutics. Biochemistry 2019, 84, 1375–1389. [Google Scholar] [CrossRef] [PubMed]
  211. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef]
  212. Xu, W.M.; Li, A.; Chen, J.J.; Sun, E.J. Research Development on Exosome Separation Technology. J. Membr. Biol. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  213. 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]
  214. Martins, T.S.; Vaz, M.; Henriques, A.G. A review on comparative studies addressing exosome isolation methods from body fluids. Anal. Bioanal. Chem. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  215. Sidhom, K.; Obi, P.O.; Saleem, A. A Review of Exosomal Isolation Methods: Is Size Exclusion Chromatography the Best Option? Int. J. Mol. Sci. 2020, 21, 6466. [Google Scholar] [CrossRef] [PubMed]
  216. Wang, J.; Ma, P.; Kim, D.H.; Liu, B.F.; Demirci, U. Towards Microfluidic-Based Exosome Isolation and Detection for Tumor Therapy. Nano Today 2021, 37, 101066. [Google Scholar] [CrossRef] [PubMed]
  217. Bari, S.M.I.; Hossain, F.B.; Nestorova, G.G. Advances in Biosensors Technology for Detection and Characterization of Extracellular Vesicles. Sensors 2021, 21, 7645. [Google Scholar] [CrossRef]
  218. Gebeyehu, A.; Kommineni, N.; Meckes, D.G., Jr.; Sachdeva, M.S. Role of Exosomes for Delivery of Chemotherapeutic Drugs. Crit. Rev. Ther. Drug Carr. Syst. 2021, 38, 53–97. [Google Scholar] [CrossRef] [PubMed]
  219. Xi, X.M.; Xia, S.J.; Lu, R. Drug loading techniques for exosome-based drug delivery systems. Pharmazie 2021, 76, 61–67. [Google Scholar] [CrossRef]
  220. Armstrong, J.P.; Holme, M.N.; Stevens, M.M. Re-Engineering Extracellular Vesicles as Smart Nanoscale Therapeutics. ACS Nano 2017, 11, 69–83. [Google Scholar] [CrossRef] [Green Version]
  221. Choi, H.; Yim, H.; Park, C.; Ahn, S.H.; Ahn, Y.; Lee, A.; Yang, H.; Choi, C. Targeted Delivery of Exosomes Armed with Anti-Cancer Therapeutics. Membranes 2022, 12, 85. [Google Scholar] [CrossRef]
  222. Yuan, F.; Li, Y.M.; Wang, Z. Preserving extracellular vesicles for biomedical applications: Consideration of storage stability before and after isolation. Drug Deliv. 2021, 28, 1501–1509. [Google Scholar] [CrossRef] [PubMed]
  223. Gelibter, S.; Marostica, G.; Mandelli, A.; Siciliani, S.; Podini, P.; Finardi, A.; Furlan, R. The impact of storage on extracellular vesicles: A systematic study. J. Extracell. Vesicles 2022, 11, e12162. [Google Scholar] [CrossRef] [PubMed]
  224. Jiang, Z.; Wang, H.; Mou, Y.; Li, L.; Jin, W. Functions and clinical applications of exosomes in pancreatic cancer. Mol. Biol. Rep. 2022, 49, 11037–11048. [Google Scholar] [CrossRef] [PubMed]
  225. Chu, X.; Yang, Y.; Tian, X. Crosstalk between Pancreatic Cancer Cells and Cancer-Associated Fibroblasts in the Tumor Microenvironment Mediated by Exosomal MicroRNAs. Int. J. Mol. Sci. 2022, 23, 9512. [Google Scholar] [CrossRef]
  226. Pan, Y.; Tang, H.; Li, Q.; Chen, G.; Li, D. Exosomes and their roles in the chemoresistance of pancreatic cancer. Cancer Med. 2022; in press. [Google Scholar] [CrossRef]
  227. Xu, W.X.; Wang, D.D.; Zhao, Z.Q.; Zhang, H.D.; Yang, S.J.; Zhang, Q.; Li, L.; Zhang, J. Exosomal microRNAs shuttling between tumor cells and macrophages: Cellular interactions and novel therapeutic strategies. Cancer Cell Int. 2022, 22, 190. [Google Scholar] [CrossRef]
  228. Tang, J.; He, J.; Feng, C.; Tu, C. Exosomal MiRNAs in Osteosarcoma: Biogenesis and Biological Functions. Front. Pharmacol. 2022, 13, 902049. [Google Scholar] [CrossRef]
  229. Kumar, V.B.S.; Anjali, K. Tumour generated exosomal miRNAs: A major player in tumour angiogenesis. Biochim. Biophys Acta. Mol. Basis Dis. 2022, 1868, 166383. [Google Scholar] [CrossRef]
  230. Li, C.; Zhou, T.; Chen, J.; Li, R.; Chen, H.; Luo, S.; Chen, D.; Cai, C.; Li, W. The role of Exosomal miRNAs in cancer. J. Transl. Med. 2022, 20, 6. [Google Scholar] [CrossRef]
  231. Chen, X.; Feng, J.; Chen, W.; Shao, S.; Chen, L.; Wan, H. Small extracellular vesicles: From promoting pre-metastatic niche formation to therapeutic strategies in breast cancer. Cell Commun. Signal. 2022, 20, 141. [Google Scholar] [CrossRef] [PubMed]
  232. Baldasici, O.; Pileczki, V.; Cruceriu, D.; Gavrilas, L.I.; Tudoran, O.; Balacescu, L.; Vlase, L.; Balacescu, O. Breast Cancer-Delivered Exosomal miRNA as Liquid Biopsy Biomarkers for Metastasis Prediction: A Focus on Translational Research with Clinical Applicability. Int. J. Mol. Sci. 2022, 23, 9371. [Google Scholar] [CrossRef] [PubMed]
  233. Zhang, W.; Jiang, Z.; Tang, D. The value of exosome-derived noncoding RNAs in colorectal cancer proliferation, metastasis, and clinical applications. Clin. Transl. Oncol. 2022, 24, 2305–2318. [Google Scholar] [CrossRef] [PubMed]
  234. Wang, S.E. Extracellular vesicles in cancer therapy. Semin. Cancer Biol. 2022, 86, 296–309. [Google Scholar] [CrossRef]
  235. Wu, Q.; Sun, S.; Li, Z.; Yang, Q.; Li, B.; Zhu, S.; Wang, L.; Wu, J.; Yuan, J.; Wang, C.; et al. Breast cancer-released exosomes trigger cancer-associated cachexia to promote tumor progression. Adipocyte 2019, 8, 31–45. [Google Scholar] [CrossRef]
  236. Kalita-de Croft, P.; Sharma, S.; Sobrevia, L.; Salomon, C. Extracellular vesicle interactions with the external and internal exposome in mediating carcinogenesis. Mol. Asp. Med. 2022, 87, 101039. [Google Scholar] [CrossRef]
  237. Pandian, S.R.K.; Vijayakumar, K.K.; Kunjiappan, S.; Babkiewicz, E.; Maszczyk, P. Emerging role of exosomes in hematological malignancies. Clin. Exp. Med. 2022; in press. [Google Scholar] [CrossRef]
  238. Mavroeidi, P.; Vetsi, M.; Dionysopoulou, D.; Xilouri, M. Exosomes in Alpha-Synucleinopathies: Propagators of Pathology or Potential Candidates for Nanotherapeutics? Biomolecules 2022, 12, 957. [Google Scholar] [CrossRef]
  239. Sun, X.; Zhang, S. Exosomes from WSSV-infected shrimp contain viral components that mediate virus infection. J. Gen. Virol. 2022; in press. [Google Scholar] [CrossRef]
  240. Huda, M.N.; Nurunnabi, M. Potential Application of Exosomes in Vaccine Development and Delivery. Pharm. Res. 2022, 39, 2635–2671. [Google Scholar] [CrossRef]
  241. Matsuzaka, Y.; Yashiro, R. Regulation of Extracellular Vesicle-Mediated Immune Responses against Antigen-Specific Presentation. Vaccines 2022, 10, 1691. [Google Scholar] [CrossRef]
  242. McGowan, R.; Sally, Á.; McCabe, A.; Moran, B.M.; Finn, K. Circulating Nucleic Acids as Novel Biomarkers for Pancreatic Ductal Adenocarcinoma. Cancers 2022, 14, 2027. [Google Scholar] [CrossRef] [PubMed]
  243. Preethi, K.A.; Selvakumar, S.C.; Ross, K.; Jayaraman, S.; Tusubira, D.; Sekar, D. Liquid biopsy: Exosomal microRNAs as novel diagnostic and prognostic biomarkers in cancer. Mol. Cancer 2022, 21, 54. [Google Scholar] [CrossRef] [PubMed]
  244. Peng, J.; Liang, Q.; Xu, Z.; Cai, Y.; Peng, B.; Li, J.; Zhang, W.; Kang, F.; Hong, Q.; Yan, Y.; et al. Current Understanding of Exosomal MicroRNAs in Glioma Immune Regulation and Therapeutic Responses. Front. Immunol. 2022, 12, 813747. [Google Scholar] [CrossRef] [PubMed]
  245. Bonilla, H.; Hampton, D.; Marques de Menezes, E.G.; Deng, X.; Montoya, J.G.; Anderson, J.; Norris, P.J. Comparative Analysis of Extracellular Vesicles in Patients with Severe and Mild Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Front. Immunol. 2022, 13, 841910. [Google Scholar] [CrossRef] [PubMed]
  246. Giloteaux, L.; O’Neal, A.; Castro-Marrero, J.; Levine, S.M.; Hanson, M.R. Cytokine profiling of extracellular vesicles isolated from plasma in myalgic encephalomyelitis/chronic fatigue syndrome: A pilot study. J. Transl. Med. 2020, 18, 387. [Google Scholar] [CrossRef]
  247. Eguchi, A.; Fukuda, S.; Kuratsune, H.; Nojima, J.; Nakatomi, Y.; Watanabe, Y.; Feldstein, A.E. Identification of actin network proteins, talin-1 and filamin-A, in circulating extracellular vesicles as blood biomarkers for human myalgic encephalomyelitis/chronic fatigue syndrome. Brain. Behav. Immun. 2020, 84, 106–114. [Google Scholar] [CrossRef]
  248. Valencia, J.; Ferreira, M.; Merino-Torres, J.F.; Marcilla, A.; Soriano, J.M. The Potential Roles of Extracellular Vesicles as Biomarkers for Parkinson’s Disease: A Systematic Review. Int. J. Mol. Sci. 2022, 23, 11508. [Google Scholar] [CrossRef]
  249. Sepúlveda, D.; Cisternas-Olmedo, M.; Arcos, J.; Nassif, M.; Vidal, R.L. Contribution of Autophagy-Lysosomal Pathway in the Exosomal Secretion of Alpha-Synuclein and Its Impact in the Progression of Parkinson’s Disease. Front. Mol. Neurosci. 2022, 15, 805087. [Google Scholar] [CrossRef]
  250. Li, K.L.; Huang, H.Y.; Ren, H.; Yang, X.L. Role of exosomes in the pathogenesis of inflammation in Parkinson’s disease. Neural. Regen. Res. 2022, 17, 1898–1906. [Google Scholar] [CrossRef]
  251. Ouerdane, Y.; Hassaballah, M.Y.; Nagah, A.; Ibrahim, T.M.; Mohamed, H.A.H.; El-Baz, A.; Attia, M.S. Exosomes in Parkinson: Revisiting Their Pathologic Role and Potential Applications. Pharmaceuticals 2022, 15, 76. [Google Scholar] [CrossRef] [PubMed]
  252. Zhao, Y.; Liu, B.; Wang, J.; Xu, L.; Yu, S.; Fu, J.; Yan, X.; Su, J. Aβ and Tau Regulate Microglia Metabolism via Exosomes in Alzheimer’s Disease. Biomedicines 2022, 10, 1800. [Google Scholar] [CrossRef]
  253. Chen, P.C.; Wu, D.; Hu, C.J.; Chen, H.Y.; Hsieh, Y.C.; Huang, C.C. Exosomal TAR DNA-binding protein-43 and neurofilaments in plasma of amyotrophic lateral sclerosis patients: A longitudinal follow-up study. J. Neurol. Sci. 2020, 418, 117070. [Google Scholar] [CrossRef] [PubMed]
  254. Younas, N.; Fernandez Flores, L.C.; Hopfner, F.; Höglinger, G.U.; Zerr, I. A new paradigm for diagnosis of neurodegenerative diseases: Peripheral exosomes of brain origin. Transl. Neurodegener. 2022, 11, 28. [Google Scholar] [CrossRef]
  255. Mustapic, M.; Eitan, E.; Werner, J.K., Jr.; Berkowitz, S.T.; Lazaropoulos, M.P.; Tran, J.; Goetzl, E.J.; Kapogiannis, D. Plasma Extracellular Vesicles Enriched for Neuronal Origin: A Potential Window into Brain Pathologic Processes. Front. Neurosci. 2017, 11, 278. [Google Scholar] [CrossRef] [Green Version]
  256. Goetzl, E.J.; Elahi, F.M.; Mustapic, M.; Kapogiannis, D.; Pryhoda, M.; Gilmore, A.; Gorgens, K.A.; Davidson, B.; Granholm, A.C.; Ledreux, A. Altered levels of plasma neuron-derived exosomes and their cargo proteins characterize acute and chronic mild traumatic brain injury. FASEB J. 2019, 33, 5082–5088. [Google Scholar] [CrossRef] [PubMed]
  257. Shi, M.; Kovac, A.; Korff, A.; Cook, T.J.; Ginghina, C.; Bullock, K.M.; Yang, L.; Stewart, T.; Zheng, D.; Aro, P.; et al. CNS tau efflux via exosomes is likely increased in Parkinson’s disease but not in Alzheimer’s disease. Alzheimers. Dement. 2016, 12, 1125–1131. [Google Scholar] [CrossRef] [Green Version]
  258. Blijdorp, C.J.; Hartjes, T.A.; Wei, K.Y.; van Heugten, M.H.; Bovée, D.M.; Budde, R.P.J.; van de Wetering, J.; Hoenderop, J.G.J.; van Royen, M.E.; Zietse, R.; et al. Nephron mass determines the excretion rate of urinary extracellular vesicles. J. Extracell. Vesicles 2022, 11, e12181. [Google Scholar] [CrossRef]
  259. Tomiyama, E.; Fujita, K.; Nonomura, N. Urinary Extracellular Vesicles: Ultracentrifugation Method. Methods Mol. Biol. 2021, 2292, 173–181. [Google Scholar] [CrossRef]
  260. Yu, D.; Li, Y.; Wang, M.; Gu, J.; Xu, W.; Cai, H.; Fang, X.; Zhang, X. Exosomes as a new frontier of cancer liquid biopsy. Mol. Cancer 2022, 21, 56. [Google Scholar] [CrossRef]
  261. Ebert, B.; Rai, A.J. Isolation and Characterization of Amniotic Fluid-Derived Extracellular Vesicles for Biomarker Discovery. Methods Mol. Biol. 2019, 1885, 287–294. [Google Scholar] [CrossRef] [PubMed]
  262. Datta, A.; Kim, H.; McGee, L.; Johnson, A.E.; Talwar, S.; Marugan, J.; Southall, N.; Hu, X.; Lal, M.; Mondal, D.; et al. High-throughput screening identified selective inhibitors of exosome biogenesis and secretion: A drug repurposing strategy for advanced cancer. Sci. Rep. 2018, 8, 8161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Urabe, F.; Kosaka, N.; Sawa, Y.; Yamamoto, Y.; Ito, K.; Yamamoto, T.; Kimura, T.; Egawa, S.; Ochiya, T. miR-26a regulates extracellular vesicle secretion from prostate cancer cells via targeting SHC4, PFDN4, and CHORDC1. Sci. Adv. 2020, 6, eaay3051. [Google Scholar] [CrossRef] [PubMed]
  264. Asgarpour, K.; Shojaei, Z.; Amiri, F.; Ai, J.; Mahjoubin-Tehran, M.; Ghasemi, F.; ArefNezhad, R.; Hamblin, M.R.; Mirzaei, H. Exosomal microRNAs derived from mesenchymal stem cells: Cell-to-cell messages. Cell Commun. Signal. 2020, 18, 149. [Google Scholar] [CrossRef] [PubMed]
  265. Li, T.; Gu, J.; Yang, O.; Wang, J.; Wang, Y.; Kong, J. Bone Marrow Mesenchymal Stem Cell-Derived Exosomal miRNA-29c Decreases Cardiac Ischemia/Reperfusion Injury Through Inhibition of Excessive Autophagy via the PTEN/Akt/mTOR Signaling Pathway. Circ. J. 2020, 84, 1304–1311. [Google Scholar] [CrossRef]
  266. Nikfarjam, S.; Rezaie, J.; Zolbanin, N.M.; Jafari, R. Mesenchymal stem cell derived-exosomes: A modern approach in translational medicine. J. Transl. Med. 2020, 18, 449. [Google Scholar] [CrossRef] [PubMed]
  267. Tian, S.; Zhou, X.; Zhang, M.; Cui, L.; Li, B.; Liu, Y.; Su, R.; Sun, K.; Hu, Y.; Yang, F.; et al. Mesenchymal stem cell-derived exosomes protect against liver fibrosis via delivering miR-148a to target KLF6/STAT3 pathway in macrophages. Stem Cell Res. Ther. 2022, 13, 330. [Google Scholar] [CrossRef]
  268. Chen, Y.A.; Lu, C.H.; Ke, C.C.; Chiu, S.J.; Jeng, F.S.; Chang, C.W.; Yang, B.H.; Liu, R.S. Mesenchymal Stem Cell-Derived Exosomes Ameliorate Alzheimer’s Disease Pathology and Improve Cognitive Deficits. Biomedicines 2021, 9, 594. [Google Scholar] [CrossRef]
  269. Lee, B.C.; Kang, I.; Yu, K.R. Therapeutic Features and Updated Clinical Trials of Mesenchymal Stem Cell (MSC)-Derived Exosomes. J. Clin. Med. 2021, 10, 711. [Google Scholar] [CrossRef]
  270. Le, M.N.; Fan, Z.H. Exosome isolation using nanostructures and microfluidic devices. Biomed. Mater. 2021, 16, 022005. [Google Scholar] [CrossRef]
  271. Wu, D.; Yan, J.; Shen, X.; Sun, Y.; Thulin, M.; Cai, Y.; Wik, L.; Shen, Q.; Oelrich, J.; Qian, X.; et al. Profiling surface proteins on individual exosomes using a proximity barcoding assay. Nat. Commun. 2019, 10, 3854. [Google Scholar] [CrossRef]
  272. Koster, H.J.; Rojalin, T.; Powell, A.; Pham, D.; Mizenko, R.R.; Birkeland, A.C.; Carney, R.P. Surface enhanced Raman scattering of extracellular vesicles for cancer diagnostics despite isolation dependent lipoprotein contamination. Nanoscale 2021, 13, 14760–14776. [Google Scholar] [CrossRef] [PubMed]
  273. Nizamudeen, Z.; Markus, R.; Lodge, R.; Parmenter, C.; Platt, M.; Chakrabarti, L.; Sottile, V. Rapid and accurate analysis of stem cell-derived extracellular vesicles with super resolution microscopy and live imaging. Biochim. Biophys. Acta. Mol. Cell Res. 2018, 1865, 1891–1900. [Google Scholar] [CrossRef] [PubMed]
  274. Riazanski, V.; Mauleon, G.; Lucas, K.; Walker, S.; Zimnicka, A.M.; McGrath, J.L.; Nelson, D.J. Real time imaging of single extracellular vesicle pH regulation in a microfluidic cross-flow filtration platform. Commun. Biol. 2022, 5, 13. [Google Scholar] [CrossRef] [PubMed]
  275. Martins, Á.M.; Ramos, C.C.; Freitas, D.; Reis, C.A. Glycosylation of Cancer Extracellular Vesicles: Capture Strategies, Functional Roles and Potential Clinical Applications. Cells 2021, 10, 109. [Google Scholar] [CrossRef]
  276. Willms, E.; Cabañas, C.; Mäger, I.; Wood, M.J.A.; Vader, P. Extracellular Vesicle Heterogeneity: Subpopulations, Isolation Techniques, and Diverse Functions in Cancer Progression. Front. Immunol. 2018, 9, 738. [Google Scholar] [CrossRef] [Green Version]
  277. Jang, S.C.; Economides, K.D.; Moniz, R.J.; Sia, C.L.; Lewis, N.; McCoy, C.; Zi, T.; Zhang, K.; Harrison, R.A.; Lim, J.; et al. ExoSTING, an extracellular vesicle loaded with STING agonists, promotes tumor immune surveillance. Commun. Biol. 2021, 4, 497. [Google Scholar] [CrossRef]
  278. Lewis, N.D.; Sia, C.L.; Kirwin, K.; Haupt, S.; Mahimkar, G.; Zi, T.; Xu, K.; Dooley, K.; Jang, S.C.; Choi, B.; et al. Exosome Surface Display of IL12 Results in Tumor-Retained Pharmacology with Superior Potency and Limited Systemic Exposure Compared with Recombinant IL12. Mol. Cancer Ther. 2021, 20, 523–534. [Google Scholar] [CrossRef]
  279. Munagala, R.; Aqil, F.; Jeyabalan, J.; Gupta, R.C. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2016, 371, 48–61. [Google Scholar] [CrossRef] [Green Version]
  280. Benecke, L.; Coray, M.; Umbricht, S.; Chiang, D.; Figueiró, F.; Muller, L. Exosomes: Small EVs with Large Immunomodulatory Effect in Glioblastoma. Int. J. Mol. Sci. 2021, 22, 3600. [Google Scholar] [CrossRef]
  281. Zhuang, X.; Xiang, X.; Grizzle, W.; Sun, D.; Zhang, S.; Axtell, R.C.; Ju, S.; Mu, J.; Zhang, L.; Steinman, L.; et al. Treatment of brain inflammatory diseases by delivering exosome encapsulated anti-inflammatory drugs from the nasal region to the brain. Mol. Ther. 2011, 19, 1769–1779. [Google Scholar] [CrossRef] [PubMed]
  282. 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, 2003–20014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  283. Huang, D.; Chen, J.; Hu, D.; Xie, F.; Yang, T.; Li, Z.; Wang, X.; Xiao, Y.; Zhong, J.; Jiang, Y.; et al. Advances in Biological Function and Clinical Application of Small Extracellular Vesicle Membrane Proteins. Front. Oncol. 2021, 11, 675940. [Google Scholar] [CrossRef]
  284. Ter-Ovanesyan, D.; Norman, M.; Lazarovits, R.; Trieu, W.; Lee, J.H.; Church, G.M.; Walt, D.R. Framework for rapid comparison of extracellular vesicle isolation methods. eLife 2021, 10, e70725. [Google Scholar] [CrossRef] [PubMed]
  285. Jia, Y.; Yu, L.; Ma, T.; Xu, W.; Qian, H.; Sun, Y.; Shi, H. Small extracellular vesicles isolation and separation: Current techniques, pending questions and clinical applications. Theranostics 2022, 12, 6548–6575. [Google Scholar] [CrossRef]
  286. Tzaridis, T.; Bachurski, D.; Liu, S.; Surmann, K.; Babatz, F.; Gesell Salazar, M.; Völker, U.; Hallek, M.; Herrlinger, U.; Vorberg, I.; et al. Extracellular Vesicle Separation Techniques Impact Results from Human Blood Samples: Considerations for Diagnostic Applications. Int. J. Mol. Sci. 2021, 22, 9211. [Google Scholar] [CrossRef] [PubMed]
  287. Liangsupree, T.; Multia, E.; Riekkola, M.L. Modern isolation and separation techniques for extracellular vesicles. J. Chromatogr. A 2021, 1636, 461773. [Google Scholar] [CrossRef]
  288. Zhang, H.; Zhang, Q.; Deng, Y.; Chen, M.; Yang, C. Improving Isolation of Extracellular Vesicles by Utilizing Nanomaterials. Membranes 2021, 12, 55. [Google Scholar] [CrossRef]
  289. Wang, F.; Cerione, R.A.; Antonyak, M.A. Isolation and characterization of extracellular vesicles produced by cell lines. STAR Protoc. 2021, 2, 100295. [Google Scholar] [CrossRef]
  290. Neyroud, A.S.; Chiechio, R.M.; Moulin, G.; Ducarre, S.; Heichette, C.; Dupont, A.; Budzynski, M.; Even-Hernandez, P.; Faro, M.J.L.; Yefimova, M.; et al. Diversity of Extracellular Vesicles in Human Follicular Fluid: Morphological Analysis and Quantification. Int. J. Mol. Sci. 2022, 23, 11676. [Google Scholar] [CrossRef]
  291. Huang, Y.; Wang, S.; Cai, Q.; Jin, H. Effective methods for isolation and purification of extracellular vesicles from plants. J. Integr. Plant Biol. 2021, 63, 2020–2030. [Google Scholar] [CrossRef]
  292. Chen, B.Y.; Sung, C.W.; Chen, C.; Cheng, C.M.; Lin, D.P.; Huang, C.T.; Hsu, M.Y. Advances in exosomes technology. Clin. Chim. Acta. 2019, 493, 14–19. [Google Scholar] [CrossRef] [PubMed]
  293. Hu, H.T.; Nishimura, T.; Suetsugu, S. Ultracentrifugal separation, characterization, and functional study of extracellular vesicles derived from serum-free cell culture. STAR Protoc. 2021, 2, 100625. [Google Scholar] [CrossRef] [PubMed]
  294. Campos-Silva, C.; Cáceres-Martell, Y.; Sánchez-Herrero, E.; Sandúa, A.; Beneitez-Martínez, A.; González, Á.; Provencio, M.; Romero, A.; Jara-Acevedo, R.; Yáñez-Mó, M.; et al. A simple immunoassay for extracellular vesicle liquid biopsy in microliters of non-processed plasma. J. Nanobiotechnol. 2022, 20, 72. [Google Scholar] [CrossRef]
  295. Gurunathan, S.; Kang, M.H.; Jeyaraj, M.; Qasim, M.; Kim, J.H. Review of the Isolation, Characterization, Biological Function, and Multifarious Therapeutic Approaches of Exosomes. Cells 2019, 8, 307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  296. Yang, D.; Zhang, W.; Zhang, H.; Zhang, F.; Chen, L.; Ma, L.; Larcher, L.M.; Chen, S.; Liu, N.; Zhao, Q.; et al. Progress, opportunity, and perspective on exosome isolation—Efforts for efficient exosome-based theranostics. Theranostics 2020, 10, 3684–3707. [Google Scholar] [CrossRef] [PubMed]
  297. Chen, J.; Li, P.; Zhang, T.; Xu, Z.; Huang, X.; Wang, R.; Du, L. Review on Strategies and Technologies for Exosome Isolation and Purification. Front. Bioeng. Biotechnol. 2022, 9, 811971. [Google Scholar] [CrossRef]
  298. Kaddour, H.; Tranquille, M.; Okeoma, C.M. The Past, the Present, and the Future of the Size Exclusion Chromatography in Extracellular Vesicles Separation. Viruses 2021, 13, 2272. [Google Scholar] [CrossRef] [PubMed]
  299. Liu, D.S.K.; Upton, F.M.; Rees, E.; Limb, C.; Jiao, L.R.; Krell, J.; Frampton, A.E. Size-Exclusion Chromatography as a Technique for the Investigation of Novel Extracellular Vesicles in Cancer. Cancers 2020, 12, 3156. [Google Scholar] [CrossRef] [PubMed]
  300. Monguió-Tortajada, M.; Gálvez-Montón, C.; Bayes-Genis, A.; Roura, S.; Borràs, F.E. Extracellular vesicle isolation methods: Rising impact of size-exclusion chromatography. Cell Mol. Life Sci. 2019, 76, 2369–2382. [Google Scholar] [CrossRef] [PubMed]
  301. Logozzi, M.; Di Raimo, R.; Mizzoni, D.; Fais, S. Immunocapture-based ELISA to characterize and quantify exosomes in both cell culture supernatants and body fluids. Methods Enzymol. 2020, 645, 155–180. [Google Scholar] [CrossRef] [PubMed]
  302. Isaksson, G.L.; Nielsen, M.B.; Hinrichs, G.R.; Krogstrup, N.V.; Zachar, R.; Stubmark, H.; Svenningsen, P.; Madsen, K.; Bistrup, C.; Jespersen, B.; et al. Proteinuria is accompanied by intratubular complement activation and apical membrane deposition of C3dg and C5b-9 in kidney transplant recipients. Am. J. Physiol. Renal. Physiol. 2022, 322, F150–F163. [Google Scholar] [CrossRef]
  303. Yousif, G.; Qadri, S.; Parray, A.; Akhthar, N.; Shuaib, A.; Haik, Y. Exosomes Derived Neuronal Markers: Immunoaffinity Isolation and Characterization. Neuromol. Med. 2022, 24, 339–351. [Google Scholar] [CrossRef]
  304. Wang, Y.T.; Cai, M.D.; Sun, L.L.; Hua, R.N. A Rapid and Facile Separation-Detection Integrated Strategy for Exosome Profiling Based on Boronic Acid-Directed Coupling Immunoaffinity. Anal. Chem. 2021, 93, 16059–16067. [Google Scholar] [CrossRef] [PubMed]
  305. Yasui, T.; Paisrisarn, P.; Yanagida, T.; Konakade, Y.; Nakamura, Y.; Nagashima, K.; Musa, M.; Thiodorus, I.A.; Takahashi, H.; Naganawa, T.; et al. Molecular profiling of extracellular vesicles via charge-based capture using oxide nanowire microfluidics. Biosens. Bioelectron. 2021, 194, 113589. [Google Scholar] [CrossRef] [PubMed]
  306. Zhu, J.; Zhang, J.; Ji, X.; Tan, Z.; Lubman, D.M. Column-based Technology for CD9-HPLC Immunoaffinity Isolation of Serum Extracellular Vesicles. J. Proteome Res. 2021, 20, 4901–4911. [Google Scholar] [CrossRef]
  307. Paulmurugan, R.; Liu, Y.; Sukumar, U.K.; Kanada, M.; Massoud, T.F. BRET Sensors for Imaging Membrane Integrity of Microfluidically Generated Extracellular Vesicles. Methods Mol. Biol. 2022, 2525, 227–238. [Google Scholar] [CrossRef]
  308. Mousavi, S.M.; Amin Mahdian, S.M.; Ebrahimi, M.S.; Taghizadieh, M.; Vosough, M.; Sadri Nahand, J.; Hosseindoost, S.; Vousooghi, N.; Javar, H.A.; Larijani, B.; et al. Microfluidics for detection of exosomes and microRNAs in cancer: State of the art. Mol. Ther. Nucleic Acids 2022, 28, 758–791. [Google Scholar] [CrossRef]
  309. Abreu, C.M.; Costa-Silva, B.; Reis, R.L.; Kundu, S.C.; Caballero, D. Microfluidic platforms for extracellular vesicle isolation, analysis and therapy in cancer. Lab. Chip. 2022, 22, 1093–1125. [Google Scholar] [CrossRef]
  310. Diaz-Armas, G.G.; Cervantes-Gonzalez, A.P.; Martinez-Duarte, R.; Perez-Gonzalez, V.H. Electrically driven microfluidic platforms for exosome manipulation and characterization. Electrophoresis 2022, 43, 327–339. [Google Scholar] [CrossRef]
  311. Shirejini, S.Z.; Inci, F. The Yin and Yang of exosome isolation methods: Conventional practice, microfluidics, and commercial kits. Biotechnol. Adv. 2022, 54, 107814. [Google Scholar] [CrossRef]
  312. Gwak, H.; Park, S.; Kim, J.; Lee, J.D.; Kim, I.S.; Kim, S.I.; Hyun, K.A.; Jung, H.I. Microfluidic chip for rapid and selective isolation of tumor-derived extracellular vesicles for early diagnosis and metastatic risk evaluation of breast cancer. Biosens. Bioelectron. 2021, 192, 113495. [Google Scholar] [CrossRef]
  313. Fang, Z.; Liu, K. Plant-derived extracellular vesicles as oral drug delivery carriers. J. Control. Release 2022, 350, 389–400. [Google Scholar] [CrossRef] [PubMed]
  314. Ali, N.B.; Abdull Razis, A.F.; Ooi, J.; Chan, K.W.; Ismail, N.; Foo, J.B. Theragnostic Applications of Mammal and Plant-Derived Extracellular Vesicles: Latest Findings, Current Technologies, and Prospects. Molecules 2022, 27, 3941. [Google Scholar] [CrossRef] [PubMed]
  315. Muñoz, E.L.; Fuentes, F.B.; Felmer, R.N.; Yeste, M.; Arias, M.E. Extracellular vesicles in mammalian reproduction: A review. Zygote 2022, 30, 440–463. [Google Scholar] [CrossRef] [PubMed]
  316. Tesfaye, D.; Menjivar, N.; Gebremedhn, S. Current knowledge and the future potential of extracellular vesicles in mammalian reproduction. Reprod. Fertil. Dev. 2021, 34, 174–189. [Google Scholar] [CrossRef]
  317. Zhou, Q.; Ma, K.; Hu, H.; Xing, X.; Huang, X.; Gao, H. Extracellular vesicles: Their functions in plant-pathogen interactions. Mol. Plant Pathol. 2022, 23, 760–771. [Google Scholar] [CrossRef]
  318. Díez-Sainz, E.; Milagro, F.I.; Riezu-Boj, J.I.; Lorente-Cebrián, S. Effects of gut microbiota-derived extracellular vesicles on obesity and diabetes and their potential modulation through diet. J. Physiol. Biochem. 2022, 78, 485–499. [Google Scholar] [CrossRef] [PubMed]
  319. Teng, Y.; Ren, Y.; Sayed, M.; Hu, X.; Lei, C.; Kumar, A.; Hutchins, E.; Mu, J.; Deng, Z.; Luo, C.; et al. Plant-Derived Exosomal MicroRNAs Shape the Gut Microbiota. Cell Host Microbe. 2018, 24, 637–652.e8. [Google Scholar] [CrossRef] [Green Version]
  320. Suri, K.; D’Souza, A.; Huang, D.; Bhavsar, A.; Amiji, M. Bacterial extracellular vesicle applications in cancer immunotherapy. Bioact. Mater. 2022, 22, 551–566. [Google Scholar] [CrossRef]
  321. Mishra, S.; Amatya, S.B.; Salmi, S.; Koivukangas, V.; Karihtala, P.; Reunanen, J. Microbiota and Extracellular Vesicles in Anti-PD-1/PD-L1 Therapy. Cancers 2022, 4, 5121. [Google Scholar] [CrossRef] [PubMed]
  322. Liu, H.; Zhang, H.; Han, Y.; Hu, Y.; Geng, Z.; Su, J. Bacterial extracellular vesicles-based therapeutic strategies for bone and soft tissue tumors therapy. Theranostics 2022, 12, 6576–6594. [Google Scholar] [CrossRef] [PubMed]
  323. Xie, J.; Li, Q.; Haesebrouck, F.; Van Hoecke, L.; Vandenbroucke, R.E. The tremendous biomedical potential of bacterial extracellular vesicles. Trends Biotechnol. 2022, 40, 1173–1194. [Google Scholar] [CrossRef]
  324. Janda, M.; Robatzek, S. Extracellular vesicles from phytobacteria: Properties, functions and uses. Biotechnol. Adv. 2022, 58, 107934. [Google Scholar] [CrossRef] [PubMed]
  325. Mustajab, T.; Kwamboka, M.S.; Choi, D.A.; Kang, D.W.; Kim, J.; Han, K.R.; Han, Y.; Lee, S.; Song, D.; Chwae, Y.J. Update on Extracellular Vesicle-Based Vaccines and Therapeutics to Combat COVID-19. Int. J. Mol. Sci. 2022, 23, 11247. [Google Scholar] [CrossRef]
  326. Goubran, H.; Seghatchian, J.; Sabry, W.; Ragab, G.; Burnouf, T. Platelet and extracellular vesicles in COVID-19 infection and its vaccines. Transfus. Apher. Sci. 2022, 61, 103459. [Google Scholar] [CrossRef] [PubMed]
  327. Yoo, K.H.; Thapa, N.; Kim, B.J.; Lee, J.O.; Jang, Y.N.; Chwae, Y.J.; Kim, J. Possibility of exosome-based coronavirus disease 2019 vaccine (Review). Mol. Med. Rep. 2022, 25, 26. [Google Scholar] [CrossRef]
  328. Burgos-Ravanal, R.; Campos, A.; Díaz-Vesga, M.C.; González, M.F.; León, D.; Lobos-González, L.; Leyton, L.; Kogan, M.J.; Quest, A.F.G. Extracellular Vesicles as Mediators of Cancer Disease and as Nanosystems in Theranostic Applications. Cancers 2021, 13, 3324. [Google Scholar] [CrossRef] [PubMed]
  329. Negahdaripour, M.; Vakili, B.; Nezafat, N. Exosome-based vaccines and their position in next generation vaccines. Int. Immunopharmacol. 2022, 113, 109265. [Google Scholar] [CrossRef]
  330. Dyball, L.E.; Smales, C.M. Exosomes: Biogenesis, targeting, characterization and their potential as "Plug & Play" vaccine platforms. Biotechnol. J. 2022, 17, e2100646. [Google Scholar] [CrossRef]
  331. Parveen, S.; Subramanian, K. Emerging Roles of Extracellular Vesicles in Pneumococcal Infections: Immunomodulators to Potential Novel Vaccine Candidates. Front. Cell Infect. Microbiol. 2022, 12, 836070. [Google Scholar] [CrossRef] [PubMed]
  332. Jahan, S.; Mukherjee, S.; Ali, S.; Bhardwaj, U.; Choudhary, R.K.; Balakrishnan, S.; Naseem, A.; Mir, S.A.; Banawas, S.; Alaidarous, M.; et al. Pioneer Role of Extracellular Vesicles as Modulators of Cancer Initiation in Progression, Drug Therapy, and Vaccine Prospects. Cells 2022, 11, 490. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The exosome formation and structure with components. The exosome is constituted by bi-layer membranes, on which some molecules, such as cermide, lipid, prorein, and saccharide, are located. Some proteins, HSPs, Ago2, and ESCART, and miRNAs are encapsulated during formation of exosome.
Figure 1. The exosome formation and structure with components. The exosome is constituted by bi-layer membranes, on which some molecules, such as cermide, lipid, prorein, and saccharide, are located. Some proteins, HSPs, Ago2, and ESCART, and miRNAs are encapsulated during formation of exosome.
Membranes 12 01244 g001
Figure 2. exoIL-12 is improved exosome expressed IL-12 on surface of exosome. ExoSTING in improved exosome encapsulating a STING agonist in exosomes, in which a therapeutic protein such as prostaglandin F2 receptor negative regulator (PTGFRN), is highly expressed in the exosome membrane.
Figure 2. exoIL-12 is improved exosome expressed IL-12 on surface of exosome. ExoSTING in improved exosome encapsulating a STING agonist in exosomes, in which a therapeutic protein such as prostaglandin F2 receptor negative regulator (PTGFRN), is highly expressed in the exosome membrane.
Membranes 12 01244 g002
Figure 3. The exosome purification by ultracentrifugation and immunoprecipitation using antibody. Contents in solution from supernatant of culture cells and blood samples are isolated by sequential centrifugation with different centrifugation speeds. In addition, some antibodies for some antigens on surface membrane of exosome can bind and isolated via association between exosome and antibody.
Figure 3. The exosome purification by ultracentrifugation and immunoprecipitation using antibody. Contents in solution from supernatant of culture cells and blood samples are isolated by sequential centrifugation with different centrifugation speeds. In addition, some antibodies for some antigens on surface membrane of exosome can bind and isolated via association between exosome and antibody.
Membranes 12 01244 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Matsuzaka, Y.; Yashiro, R. Advances in Purification, Modification, and Application of Extracellular Vesicles for Novel Clinical Treatments. Membranes 2022, 12, 1244. https://doi.org/10.3390/membranes12121244

AMA Style

Matsuzaka Y, Yashiro R. Advances in Purification, Modification, and Application of Extracellular Vesicles for Novel Clinical Treatments. Membranes. 2022; 12(12):1244. https://doi.org/10.3390/membranes12121244

Chicago/Turabian Style

Matsuzaka, Yasunari, and Ryu Yashiro. 2022. "Advances in Purification, Modification, and Application of Extracellular Vesicles for Novel Clinical Treatments" Membranes 12, no. 12: 1244. https://doi.org/10.3390/membranes12121244

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