Abstract
Early diagnosis and timely management are critical for determining disease outcomes and prognoses. To date, certain methods for developing disease-specific biomarkers have been reported; however, strategies for musculoskeletal disease-specific biomarker development have rarely been studied. Recent studies have highlighted the potential application of extracellular vesicles (EVs) as disease-specific biomarkers. EVs encapsulate proteins, lipids, messenger RNAs, and microRNAs derived from their cellular origin; these constituents remain stable within the EVs and can traverse the blood–brain barrier. Because of these distinctive characteristics, EVs have been actively investigated as diagnostic tools for various conditions, including cancer, inflammatory diseases, and musculoskeletal disorders. Although EVs have many advantages for biomarker development, they have not yet been fully researched in the context of musculoskeletal pathologies. The current review aimed to highlight the potential of EVs in the development of disease-specific biomarkers, summarize the processes of EV biomarkers, and discuss current limitations and future perspectives of EVs as biomarkers.
Keywords: Extracellular vesicle, biomarker, development, disease, musculoskeletal system, diagnostic tool
Introduction
Most musculoskeletal diseases, such as osteoarthritis and avascular necrosis, are clinically diagnosed using imaging modalities, including radiography, computed tomography, and magnetic resonance imaging. An imaging referral and the diagnosis are generally based on the patient’s clinical symptoms, such as pain and abnormal gait, reported during their visit to the hospital. These clinical manifestations indicate injury/damage to the underlying structure (e.g., bone, cartilage), and at this stage, the injured structure is unlikely to heal or regenerate itself. In most cases, the damaged tissues or structures will need to be managed with interventional procedures. Thus, the diagnosis of most musculoskeletal diseases has traditionally been based on patients’ symptoms, leading to delayed diagnosis and treatment, resulting in the inability to recover/regenerate the damaged tissue or structure. Therefore, early identification and management of musculoskeletal pathologies, before tissue damage, is of utmost importance.
Extracellular vesicles (EVs) have attracted considerable attention for the development of disease-specific biomarkers. EVs are 40 to 200 nm spherical vesicles encapsulated in phospholipid bilayer membranes.1,2 They are found in almost all body fluids, including cerebrospinal fluid, saliva, breast milk, and blood. EVs are secreted from all cell types, especially cells under specific disease conditions, and contain several bioactive molecules, such as functional proteins, lipids, and nucleic acids (messenger RNA (mRNA) and micro-RNA (miRNA)).1,2 On the basis of these characteristics, several studies focusing on various cancers, inflammatory diseases, and cardiovascular diseases have reported the development of disease-specific biomarkers for early diagnosis (Figure 1). 1 However, reports on EV biomarkers for musculoskeletal disorders are limited. Moreover, it is crucial to diagnose certain musculoskeletal diseases early, before disease progression, because the treatment method depends on the diagnosis time. When the disease is detected early during the cellular damage phase before the occurrence of any structural damage, the damaged tissue can be repaired and restored using regenerative medicine modalities (e.g., stem cells); however, once the disease has caused structural damage, the tissue or structure will need to be replaced using salvage methods.
Figure 1.
Extracellular vesicles have shown potential as biomarkers for both preclinical and clinical applications. Extracellular vesicles can be isolated from biofluids such as those in the respiratory and urinary systems, blood, and that in the spinal cord. They can also be isolated from cells. These extracellular vesicles can be exploited for biomarker and therapeutic purposes.
Studies on EVs in musculoskeletal disorders have been reported from various perspectives, indicating potential clinical applications. This review aims to highlight recent advancements and the clinical potential of EV-based biomarkers in musculoskeletal diseases. The authors previously identified several miRNAs that are highly associated with avascular necrosis of the femoral head and may serve as disease-specific biomarkers. 1 Identification of EV biomarkers is significant for early diagnosis of diseases and for a comprehensive understanding of the disease itself (e.g., disease occurrence, progression, or regression). Therefore, in the current review, we aim to describe the detailed procedure for developing disease-specific biomarkers using EVs, understand the current limitations of the technique, and provide future perspectives. Given the era and emphasis on cutting-edge techniques such as EV biomarkers, the current review may serve as a cornerstone for next-generation research focused on the development of EV-based disease-specific biomarkers.
This review is guided by the Scale for the Assessment of narrative review articles. 3 Our objective is to synthesize the latest research on the role of EVs as biomarkers in musculoskeletal diseases, with a particular focus on their diagnostic and prognostic potential. Through this review, we aim to provide a cohesive understanding of the clinical utility and technological requirements needed to advance EVs as reliable biomarkers in musculoskeletal diseases.
Considerations for EV biomarker discovery
Detection of EV biomarkers in biological fluids
Genetic materials and proteins in EVs are more stable than RNA or proteins in biological fluids. However, before describing suitable EV isolation methods for the diagnosis of musculoskeletal diseases, it is important to consider the body fluids from which EVs can be sourced. Because EVs are secreted from cells, they can be isolated from cells and most biological fluids, such as saliva, urine, serum, plasma, and breast milk. Among bodily fluids lacking cells, saliva and urine samples are convenient sources because they can be obtained noninvasively. Urinary EVs are used to treat kidney diseases and urinary tumors.4,5 Therefore, the selection of a suitable biological fluid to target a specific disease is crucial for demonstrating its potential as an EV biomarker source. In the case of musculoskeletal diseases, the extraction of cells and identification of EVs remain challenging. Therefore, changes in the blood environment can be predicted from serum and plasma, which constitute bodily fluids that circulate throughout the body and are suitable for identifying biomarkers related to a disease. 2 miRNAs and proteins in serum represent potential biomarkers for detecting various diseases, including muscle or bone diseases. 6 Primary cell culture of diseased myoblasts or osteocytes and acquisition of the conditioned culture medium allow for the detection of biomarkers. However, the biopsy of each patient sample remains a challenging, time-consuming, and inefficient diagnostic method. Therefore, body fluids, such as blood, are suitable for diagnosing musculoskeletal diseases. Furthermore, whole blood can be separated into plasma and serum for diagnosis.
EV isolation (serum-derived EVs)
Obtaining a sufficient quantity of biological fluids for disease diagnosis remains challenging. Therefore, an efficient and simple separation method needs to be used to isolate sufficient EVs for diagnostic purposes, even from samples with small amounts. Various techniques are available for EV isolation. Classical methods involve differential and gradient-density centrifugation. EVs can be easily separated step-by-step from cellular debris and microparticles using ultracentrifugation. 7 However, this method requires expensive equipment, and EVs isolated at high speed and high pressure have low yield and functional problems owing to deformation. 8 Therefore, filters with a pore size of approximately 0.1 to 0.22 μm are used to separate EVs, which are nanoparticles ranging in size between 40 and 50 nm. Size exclusion chromatography is a size-based separation method that uses porous phases. It is suitable for separating EVs from small samples, such as plasma, and for monitoring internal miRNAs. 9 Tangential flow filtration in the form of cross-flow filtration can be used to separate EVs, but this method is suitable for the separation of EVs from large samples such as culture media. 10 Using commercially available reagents based on polymer-based precipitation reactions may be the easiest and most convenient method to isolate serum-derived EVs. However, several limitations need to be overcome to isolate EVs with clinical applicability. 11 Moreover, it is difficult to separate pure EVs using this method because proteins with reduced solubility in the reagents (owing to polyethylene glycol) tend to precipitate and mix with the EVs. 12 Recently, to overcome the purity issue, a non-polyethylene glycol-based precipitation technique has been developed by Invent Biotechnologies. 13 Meanwhile, another method was developed to separate EVs from relatively small volume samples of body fluids, such as serum or plasma, by combining two nanofilters using a lab-scale centrifugal microfluidic system. 14 Overall, various strategies for isolating EVs of sufficient quantity and high purity from biofluids are still being developed, and the methods will be optimized by characterization of the isolated EVs.
EV characterization
According to the Minimum Information for Extracellular Vesicle Research 2023 guidelines published by the International Society for Extracellular Vesicles, researchers can define the following: nomenclature, sample collection, separation, and EV characterization. 15 Quantification of the EV particle number, concentration, and size distribution can be performed using nanoparticle tracking analysis (NTA) and dynamic light scattering (DLS). NTA and DLS can confirm the size distribution by observing the fluctuations caused by the Brownian motion of the nanoparticles through scattered light measurements. DLS can measure particles ranging from 1 nm to 6 μm but is less accurate for particle suspensions of different sizes (polydisperse suspensions). NTA can measure particles <1 μm at a wavelength of 30 nm. Visual verification is achieved by capturing light scattering-based measurements with a camera, and the size distribution, average size, and concentration of particles can be calculated.16,17 The external morphology of EVs can be visualized using transmission electron microscopy (TEM) and cryo-TEM. TEM and transmission scanning electron microscopy are used to detect and characterize EVs. TEM analysis requires the fixation and staining of EVs; however, these processes can be avoided in cryo-TEM. Therefore, cryo-TEM imaging allows for the characterization of EVs under conditions closest to their native state by directly applying the sample to an EM grid and visualizing it. 18 The internal or surface proteins of EVs can be analyzed using western blotting, enzyme-linked immunosorbent assays, and flow cytometry. 19 These methods generally use the tetraspanins CD9, CD63, and CD81, flotillin 1 and 2, which are proteins that make up the surface membrane of EVs, and ALIX, TSG101, clatherin, and ubiquitin, which are proteins involved in EV biogenesis. 20 Additionally, heat shock protein (HSP)70 and HSP90 are used as protein biomarkers. EV markers may appear different depending on their source (disease type) and isolation method. 21
miRNAs in EVs for biomarker discovery
Numerous reports have shown that the miRNA profiles of EVs isolated from the body fluids of patients differ from those of healthy individuals. Through small RNA analysis, a list of miRNAs that are candidate indicators specific to musculoskeletal diseases can be obtained. miRNAs are small non-coding RNA molecules that are primarily involved in regulating gene expression. 22 Therefore, they can be used as diagnostic markers for cancer and diseases to identify gene regulation targeted by miRNAs and analyze related mechanisms. The feasibility of miRNA expression analysis in body fluids, including blood, has increased, and this method is helpful to characterize disease states. miRNAs are being studied for the early diagnosis of cancers, including breast, lung, and colorectal cancers, and are being identified in various other diseases such as cardiovascular diseases, sepsis, nervous system disorders, diabetes mellitus, and rheumatoid arthritis.23 –25
Identification and target prediction of miRNAs have emerged as novel strategies in the field of diagnostic biomarkers for diseases. Recent advances in next-generation sequencing technology have provided improved predictions because of the improved accuracy and reliability obtained by experimentally validating miRNAs in EVs. The workflow of standardized miRNA analysis consists of (i) downloading datasets using miRNA sequencing and microarray Gene Expression Omnibus databases, (ii) searching existing literature and compiling existing information using PubMed, (iii) use of a short lead aligner, (iv) identification and characterization of miRNAs, (v) target mRNA prediction, and (vi) functional and downstream analyses. 26 Various tools with high sensitivity and specificity have been developed for miRNA validation based on quantitative reverse-transcriptase polymerase chain reaction and TaqMan miRNA assays that incorporate target-specific stem-loop reverse transcription.27,28
Proteins in EVs for biomarker discovery
EV proteins can be analyzed to identify biomarkers for different types of musculoskeletal diseases. Proteins specific to the early stages of disease development must be identified among the numerous proteins and verified for reproducibility and correlation with the disease. Liquid chromatography (LC)-mass spectrometry (MS)-based proteomics analysis is the most widely used technique for identifying new disease biomarkers in the discovery phase. 29 This technology is suitable for the discovery phase because it can precisely identify specific and abundant proteins in a limited group of patient samples. 30 The workflow to identify biomarker candidates using mass spectrometry involves the following steps: (i) protein digestion; (ii) sample desalting; (iii) data acquisition using LC-MS/MS equipment; (iv) peptide identification; (v) quality control; (vi) statistical analyses; (vii) data analysis and validation. Predicting or diagnosing a disease using a single marker remains challenging. 30 Therefore, multiple protein diagnostic analyses are essential to ensure the accuracy of protein biomarkers associated with musculoskeletal diseases.
EVs as musculoskeletal disease biomarkers
Recent studies have explored the role of EVs in musculoskeletal diseases, revealing specific proteins, miRNAs, and other molecular components in EVs that are associated with disease processes such as inflammation and tissue degeneration. For instance, in osteoarthritis, 31 EVs have been shown to carry miRNAs and inflammatory markers that reflect cartilage degradation stages, potentially providing early diagnostic cues before radiographic evidence is apparent. Similarly, in rheumatoid arthritis, 32 EVs containing immunomodulatory molecules have been correlated with disease activity, suggesting their use in monitoring treatment responses and inflammation levels in rheumatoid arthritis patients. In femoral head avascular necrosis, 1 EVs have been identified to carry markers linked to joint and spine inflammation, which could help to track disease progression at early stages where imaging may not yet show structural changes. Moreover, new approaches to identify disease-specific biomarkers using less invasive or different approaches have been developed. A diagnostic approach to develop stage-specific biomarkers using EVs from joint fluid from osteoarthritis patients has been studied. 33 In addition, marker development with less invasive specimens such as saliva and hair has been attempted and has shown potential. 34 These disease-specific EV profiles indicate promising applications of EVs as diagnostic and prognostic biomarkers in musculoskeletal diseases, enhancing the precision of current diagnostic approaches. On the basis of the relevant research that exists, the diagnostic efficacy and accuracy for musculoskeletal diseases using EVs has been promising at the experimental level, but mass production for diagnosis using EVs is challenging because miRNA-based techniques need to be improved to approach the level of over-the-counter products. 35
Clinical applications in musculoskeletal disease
We have expanded our discussion on the application of EVs in musculoskeletal diseases, integrating recent studies that highlight their utility in rheumatoid arthritis and ankylosing spondylitis in addition to osteoarthritis and avascular necrosis. Specific proteins, miRNAs, and inflammatory markers found in EVs have shown correlations with disease progression and severity in these conditions. For example, in rheumatoid arthritis, 32 EVs containing immunomodulatory components correlate with disease activity, providing potential biomarkers for monitoring inflammation and the treatment response. Similarly, in ankylosing spondylitis, 36 specific EV-associated proteins linked to inflammation offer insights into early-stage disease progression, even before significant radiographic changes occur. These additions provide a comprehensive overview of EV biomarkers across a broader range of musculoskeletal diseases, aligning the manuscript more closely with the title’s emphasis on musculoskeletal applications.
Limitations of EV-based biomarker development
In the field of biomarker development, EVs have emerged as promising candidates owing to their role in intercellular communication and their ability to transport a variety of bioactive molecules, including proteins, lipids, and nucleic acids. 37 EVs are involved in numerous physiological and pathological processes, making them valuable for disease diagnosis, prognosis, and therapy. 38 However, despite their potential, several limitations and technical challenges hinder the effective utilization of EVs in clinical applications.
Standardization of isolation and characterization protocols
Current isolation techniques, such as ultracentrifugation,39 –41 tangential flow filtration,42 –44 size-exclusion chromatography,44,45 and immunoaffinity capture40,46 often result in contamination of samples with non-EV particles, such as protein aggregates, lipoproteins, and other extracellular components.47,48 Such contaminants compromise the specificity and accuracy of downstream analyses. Furthermore, the variability in these methods results in differences in the yield, purity, and composition of isolated EVs, making it difficult to compare results across studies.49,50 This inconsistency can hamper the reproducibility and reliability of EV-based biomarkers.
Heterogeneity and variability of EVs
EVs are heterogeneous in nature and comprise various subpopulations that differ in size, origin, and cargo.51,52 The content and properties of EVs are influenced by various environmental factors, including stress conditions, disease states, and treatment regimens.53,54 This variability needs to be carefully considered and controlled in biomarker studies.55,56 The heterogeneity poses a significant challenge in identifying disease-specific biomarkers because the specific subpopulation responsible for biomarker activity needs to be accurately identified and isolated. The presence of various EV types in biological samples complicates data analysis and interpretation. Biological variability, such as differences in EV content owing to patient heterogeneity (e.g., age, sex, and disease state) and technical variability, including differences in sample handling and processing, significantly affects EV analyses.53,57 This variability complicates the identification of reliable and reproducible biomarkers. Standardized preanalytical and analytical protocols are required to minimize these variations.
Limited understanding of the biogenesis and functions of EVs
Although significant progress has been made in the study of EVs, the mechanisms underlying the biogenesis, release, and uptake of EVs are not fully understood. The limited understanding of these processes hinders the effective manipulation of EVs for biomarker discovery and therapeutic purposes. A deeper understanding of EV biology is essential for the advancement of EV-based biomarker development.58 –60
Challenges in cargo analysis
Analysis of EV cargos, including proteins, lipids, and nucleic acids, requires advanced and often complex technologies such as MS and next-generation sequencing.61 –63 The sensitivity and specificity of analyses are crucial for identifying potential biomarkers but are limited by current technological capabilities. Therefore, improved methods for high-throughput and high-sensitivity analyses are needed.64,65
Regulatory and clinical implementation hurdles
Translating EV-based biomarkers into clinical practice involves the navigation of complex regulatory pathways. Ensuring the clinical efficacy, safety, and reproducibility of EV-based diagnostics and therapeutics is challenging and requires extensive clinical validation.66,67 Regulatory guidelines specific to EV-based products are lacking, which can delay their clinical application. 68 Additionally, the translation of EV-based biomarkers from the laboratory to the clinic involves scaling up the isolation and analysis processes to handle large sample volumes. Scalability requires the development of automated and high-throughput technologies that can efficiently and reproducibly process clinical samples.
Storage and stability issues
The stability of EVs during storage and the effect of storage conditions on EV integrity and bioactivity have not yet been determined, rendering this a key area for further research.69 –71 This uncertainty can affect the reliability of EV-based biomarkers when stored for extended periods or transported between locations. The development of standardized storage protocols is essential to maintaining the integrity of EVs.
Future perspectives on developing EV-based biomarkers
The use of EVs as biomarkers for specific diseases holds great promise owing to several emerging trends and advancements in the field (Figure 1). The following points highlight key areas of focus for future research and development.
Advanced isolation and characterization techniques
The development of standardized and efficient isolation techniques is crucial for advancing EV research. Innovations in microfluidics, nanotechnology, and immunoaffinity capture are expected to enhance EV purity and yield.72 –74 Improved characterization methods, including high-resolution imaging and single-vesicle analysis, will allow for a more detailed understanding of EV heterogeneity and its specific roles in diseases.75,76 For example, recent studies have shown the necessity of combining multiple approaches to accurately diagnose or predict diseases such as breast cancer, highlighting the improved accuracy of multi-omic biomarker signatures over single-omic biomarkers. 77
Integration with omics technologies
The integration of multi-omics approaches, such as genomics, proteomics, metabolomics, and lipidomics, will provide comprehensive insights into the molecular cargo of EVs.78 –80 High-throughput technologies and advanced bioinformatics will enable the identification of novel biomarkers and the elucidation of complex disease mechanisms. This holistic approach will facilitate the discovery of robust and clinically relevant EV-based biomarker signatures. In the context of neurodegenerative diseases such as Alzheimer’s disease, multi-omic approaches may help uncover the dual roles of EVs in disease progression and therapy. 81
Clinical validation and standardization
Rigorous clinical validation of EV-based biomarkers is essential before their adoption in clinical practice.82,83 Large-scale studies and longitudinal cohorts are required to confirm the diagnostic and prognostic value of EV biomarkers. Standardization of protocols for EV isolation, characterization, and analysis will ensure the reproducibility and comparability of results across different laboratories and studies.84 –86
Single-vesicle analysis
Advances in single-vesicle analysis techniques such as super-resolution microscopy and flow cytometry will enable researchers to study the heterogeneity of EV populations in greater detail. This will help to identify specific EV subpopulations associated with particular disease states, thereby improving the specificity and sensitivity of EV-based biomarkers.75,87,88
Functional studies and mechanistic insights
Understanding the functional role of EVs in disease pathogenesis is critical for their application as biomarkers and therapeutics. Future studies should focus on elucidating the mechanisms by which EVs mediate intercellular communication, modulate immune responses, and influence disease progression. Functional studies using advanced in vitro and in vivo models will provide valuable insights into the biological relevance of EVs.89 –91
Ethical considerations
Addressing regulatory and ethical challenges is essential for the successful clinical implementation of EV-based diagnostics and therapeutics. Clear regulatory pathways and guidelines must be established to ensure the safety, efficacy, and quality of EV-based products. Ethical considerations, including patient consent and data privacy, must be thoroughly addressed in clinical studies.15,92
Personalized medicine and precision health
EV-based biomarkers have the potential to revolutionize personalized medicine by enabling the development of tailored diagnostic and therapeutic strategies. The ability of EVs to reflect dynamic changes in disease states and treatment responses will facilitate the creation of precision health approaches that improve patient outcomes and reduce adverse effects.93,94 Beyond their use as biomarkers, EVs are being explored as therapeutic agents because of their ability to deliver bioactive molecules to target cells. Engineering EVs for enhanced targeting and therapeutic efficacy is a promising area of research.95 –97 The development of EV-based therapeutics will require overcoming challenges related to large-scale production, stability, and delivery.
Conclusion
The future of EV-based biomarker research is promising, with numerous opportunities for technological and clinical advancement. By encapsulating unique bioactive molecules reflective of disease-specific processes, EVs can supplement existing diagnostic tools for conditions such as osteoarthritis, rheumatoid arthritis, and ankylosing spondylitis, offering insights into disease status and therapeutic responses. Future research should focus on standardizing EV isolation and analysis protocols to ensure reproducibility and exploring EV biomarkers in larger, disease-specific cohorts to validate their clinical applicability in musculoskeletal diagnosis and management.
Search strategy
To ensure a comprehensive review of the existing literature, we performed a structured search strategy across databases including PubMed, Scopus, and Web of Science. The key search terms used included “extracellular vesicles,” “EVs,” “biomarkers,” “musculoskeletal diseases,” “osteoarthritis,” “rheumatoid arthritis,” and “ankylosing spondylitis.” This approach allowed us to capture a thorough range of recent studies, offering a broad view of EV applications in the musculoskeletal field.
Footnotes
Author contributions: Sung SE, Seo MS, Park WT, Park S, and Lee GW developed the idea; Sung SE and Seo MS wrote the manuscript; Park WT, Lim YJ, and Park S helped to write and revise the manuscript; and Lee GW revised and finalized the manuscript. All authors significantly contributed to the document and have reviewed the final manuscript.
The authors declare that there are no conflicts of interest.
Funding: This study was supported by the 2023 Yeungnam University Research Grant.
ORCID iD: Gun Woo Lee https://orcid.org/0000-0002-8441-0802
References
- 1.Sung SE, Lim JH, Kang KK, et al. Proteomic profiling of extracellular vesicles derived from human serum for the discovery of biomarkers in avascular necrosis. Clin Proteomics 2024; 21: 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bottani M, Banfi G, Lombardi G. The clinical potential of circulating miRNAs as biomarkers: present and future applications for diagnosis and prognosis of age-associated bone diseases. Biomolecules 2020; 10: 589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Baethge C, Goldbeck-Wood S, Mertens S. SANRA-a scale for the quality assessment of narrative review articles. Res Integr Peer Rev 2019; 4: 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Li X, Yang L. Urinary exosomes: emerging therapy delivery tools and biomarkers for urinary system diseases. Biomed Pharmacother 2022; 150: 113055. [DOI] [PubMed] [Google Scholar]
- 5.Liu Q, Li S. Exosomal circRNAs: novel biomarkers and therapeutic targets for urinary tumors. Cancer Lett 2024; 588: 216759. [DOI] [PubMed] [Google Scholar]
- 6.Chen X, Ba Y, Ma L, et al. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res 2008; 18: 997–1006. [DOI] [PubMed] [Google Scholar]
- 7.Witwer KW, Buzas EI, Bemis LT, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles 2013; 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Tauro BJ, Greening DW, Mathias RA, et al. Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes. Methods 2012; 56: 293–304. [DOI] [PubMed] [Google Scholar]
- 9.Van Eijndhoven MA, Zijlstra JM, Groenewegen NJ, et al. Plasma vesicle miRNAs for therapy response monitoring in Hodgkin lymphoma patients. JCI Insight 2016; 1: e89631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.McNamara RP, Caro-Vegas CP, Costantini LM, et al. Large-scale, cross-flow based isolation of highly pure and endocytosis-competent extracellular vesicles. J Extracell Vesicles 2018; 7: 1541396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Yakubovich EI, Polischouk AG, Evtushenko VI. Principles and problems of exosome isolation from biological fluids. Biochem (Mosc) Suppl Ser A Membr Cell Biol 2022; 16: 115–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kim J, Lee H, Park K, et al. Rapid and efficient isolation of exosomes by clustering and scattering. J Clin Med 2020; 9: 650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Omrani M, Beyrampour-Basmenj H, Jahanban-Esfahlan R, et al. Global trend in exosome isolation and application: an update concept in management of diseases. Mol Cell Biochem 2024; 479: 679–691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Woo HK, Sunkara V, Park J, et al. Exodisc for rapid, size-selective, and efficient isolation and analysis of nanoscale extracellular vesicles from biological samples. ACS Nano 2017; 11: 1360–1370. [DOI] [PubMed] [Google Scholar]
- 15.Welsh JA, Goberdhan DCI, O'Driscoll L; MISEV Consortium et al. Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J Extracell Vesicles 2024; 13: e12404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Szatanek R, Baj-Krzyworzeka M, Zimoch J, et al. The methods of choice for extracellular vesicles (EVs) characterization. Int J Mol Sci 2017; 18: 1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bachurski D, Schuldner M, Nguyen PH, et al. Extracellular vesicle measurements with nanoparticle tracking analysis – An accuracy and repeatability comparison between NanoSight NS300 and ZetaView. J Extracell Vesicles 2019; 8: 1596016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cizmar P, Yuana Y. Detection and characterization of extracellular vesicles by transmission and cryo-transmission electron microscopy. Methods Mol Biol 2017; 1660: 221–232. [DOI] [PubMed] [Google Scholar]
- 19.Ansari FJ, Tafti HA, Amanzadeh A, et al. Comparison of the efficiency of ultrafiltration, precipitation, and ultracentrifugation methods for exosome isolation. Biochem Biophys Rep 2024; 38: 101668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.DeCastro J, Littig J, Chou PP, et al. The microfluidic toolbox for analyzing exosome biomarkers of aging. Molecules 2021; 26: 535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Deng F, Miller J. A review on protein markers of exosome from different bio-resources and the antibodies used for characterization. J Histotechnol 2019; 42: 226–239. [DOI] [PubMed] [Google Scholar]
- 22.Wang J, Chen J, Sen S. MicroRNA as biomarkers and diagnostics. J Cell Physiol 2016; 231: 25–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ma J, Wang P, Huang L, et al. Bioinformatic analysis reveals an exosomal miRNA-mRNA network in colorectal cancer. BMC Med Genomics 2021; 14: 60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Manna I, Quattrone A, De Benedittis S, et al. Exosomal miRNA as peripheral biomarkers in Parkinson's disease and progressive supranuclear palsy: a pilot study. Parkinsonism Relat Disord 2021; 93: 77–84. [DOI] [PubMed] [Google Scholar]
- 25.Condrat CE, Thompson DC, Barbu MG, et al. miRNAs as biomarkers in disease: latest findings regarding their role in diagnosis and prognosis. Cells 2020; 9: 276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Chen L, Heikkinen L, Wang C, et al. Trends in the development of miRNA bioinformatics tools. Brief Bioinform 2019; 20: 1836–1852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Tong L, Xue H, Xiong L, et al. Improved RT-PCR assay to quantitate the pri-, pre-, and mature microRNAs with higher efficiency and accuracy. Mol Biotechnol 2015; 57: 939–946. [DOI] [PubMed] [Google Scholar]
- 28.Forero DA, Gonzalez-Giraldo Y, Castro-Vega LJ, et al. qPCR-based methods for expression analysis of miRNAs. Biotechniques 2019; 67: 192–199. [DOI] [PubMed] [Google Scholar]
- 29.Cao Z, Yu LR. Mass spectrometry-based proteomics for biomarker discovery. Methods Mol Biol 2022; 2486: 3–17. [DOI] [PubMed] [Google Scholar]
- 30.Nakayasu ES, Gritsenko M, Piehowski PD, et al. Tutorial: best practices and considerations for mass-spectrometry-based protein biomarker discovery and validation. Nat Protoc 2021; 16: 3737–3760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Syed NH, Misbah I, Azlan M, et al. Exosomes in osteoarthritis: a review on their isolation techniques and therapeutic potential. Indian J Orthop 2024; 58: 866–875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Wu H, Chen Q, Wang S, et al. Serum exosomes lncRNAs: TCONS_I2_00013502 and ENST00000363624 are new diagnostic markers for rheumatoid arthritis. Front Immunol 2024; 15: 1419683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Moravek M, Matejova J, Spakova T. Soluble and EV-associated diagnostic and prognostic biomarkers in knee osteoarthritis pathology and detection. Life (Basel) 2023; 13: 342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Rampam S, Carnino JM, Xiao B, et al. Extracellular vesicles: an emerging clinical opportunity in musculoskeletal disease. Tissue Eng Part B Rev 2024; 30: 359–370. [DOI] [PubMed] [Google Scholar]
- 35.Crossland RE, Albiero A, Sanjurjo-Rodriguez C, et al. MicroRNA profiling of low concentration extracellular vesicle RNA utilizing NanoString nCounter technology. J Extracell Biol 2023; 2: e72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Zhang L, Qu L, Zhang Y, et al. Differential expression of circular RNAs in plasma exosomes from patients with ankylosing spondylitis. Cell Biol Int 2022; 46: 649–659. [DOI] [PubMed] [Google Scholar]
- 37.Johnstone RM, Adam M, Hammond JR, et al. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem 1987; 262: 9412–9420. [PubMed] [Google Scholar]
- 38.Chiabotto G, Gai C, Deregibus MC, et al. Salivary extracellular vesicle-associated exRNA as cancer biomarker. Cancers (Basel) 2019; 11: 891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Coughlan C, Bruce KD, Burgy O, et al. Exosome isolation by ultracentrifugation and precipitation and techniques for downstream analyses. Curr Protoc Cell Biol 2020; 88: e110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Greening DW, Xu R, Ji H, et al. A protocol for exosome isolation and characterization: evaluation of ultracentrifugation, density-gradient separation, and immunoaffinity capture methods. Methods Mol Biol 2015; 1295: 179–209. [DOI] [PubMed] [Google Scholar]
- 41.Helwa I, Cai J, Drewry MD, et al. A comparative study of serum exosome isolation using differential ultracentrifugation and three commercial reagents. PLoS One 2017; 12: e0170628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Haraszti RA, Miller R, Stoppato M, et al. Exosomes produced from 3D cultures of MSCs by tangential flow filtration show higher yield and improved activity. Mol Ther 2018; 26: 2838–2847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kim K, Park J, Jung JH, et al. Cyclic tangential flow filtration system for isolation of extracellular vesicles. APL Bioeng 2021; 5: 016103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ko M, Kim HJ, Park J, et al. Isolation of bovine milk exosome using electrophoretic oscillation assisted tangential flow filtration with antifouling of micro-ultrafiltration membrane filters. ACS Appl Mater Interfaces 2023; 15: 26069–26080. [DOI] [PubMed] [Google Scholar]
- 45.Sidhom K, Obi PO, Saleem A. A review of exosomal isolation methods: is size exclusion chromatography the best option? Int J Mol Sci 2020; 21: 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Jeppesen DK, Fenix AM, Franklin JL, et al. Reassessment of exosome composition. Cell 2019; 177: 428–445.e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Yuana Y, Levels J, Grootemaat A, et al. Co-isolation of extracellular vesicles and high-density lipoproteins using density gradient ultracentrifugation. J Extracell Vesicles 2014; 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zhang X, Takeuchi T, Takeda A, et al. Comparison of serum and plasma as a source of blood extracellular vesicles: increased levels of platelet-derived particles in serum extracellular vesicle fractions alter content profiles from plasma extracellular vesicle fractions. PLoS One 2022; 17: e0270634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Konoshenko MY, Lekchnov EA, Vlassov AV, et al. Isolation of extracellular vesicles: general methodologies and latest trends. Biomed Res Int 2018; 2018: 8545347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Jia Y, Yu L, Ma T, et al. Small extracellular vesicles isolation and separation: current techniques, pending questions and clinical applications. Theranostics 2022; 12: 6548–6575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol 2021; 16: 748–759. [DOI] [PubMed] [Google Scholar]
- 52.Malkin EZ, Bratman SV. Bioactive DNA from extracellular vesicles and particles. Cell Death Dis 2020; 11: 584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Noren Hooten N, Byappanahalli AM, Vannoy M, et al. Influences of age, race, and sex on extracellular vesicle characteristics. Theranostics 2022; 12: 4459–4476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Yin Y, Chen H, Wang Y, et al. Roles of extracellular vesicles in the aging microenvironment and age-related diseases. J Extracell Vesicles 2021; 10: e12154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Barile L, Vassalli G. Exosomes: therapy delivery tools and biomarkers of diseases. Pharmacol Ther 2017; 174: 63–78. [DOI] [PubMed] [Google Scholar]
- 56.Pletcher MJ, Pignone M. Evaluating the clinical utility of a biomarker: a review of methods for estimating health impact. Circulation 2011; 123: 1116–1124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zhang Y, Liang F, Zhang D, et al. Metabolites as extracellular vesicle cargo in health, cancer, pleural effusion, and cardiovascular diseases: an emerging field of study to diagnostic and therapeutic purposes. Biomed Pharmacother 2023; 157: 114046. [DOI] [PubMed] [Google Scholar]
- 58.Colombo M, Raposo G, Thery C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol 2014; 30: 255–289. [DOI] [PubMed] [Google Scholar]
- 59.Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science 2020; 367: eaau6977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Dixson AC, Dawson TR, Di Vizio D, et al. Context-specific regulation of extracellular vesicle biogenesis and cargo selection. Nat Rev Mol Cell Biol 2023; 24: 454–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Ludwig N, Whiteside TL, Reichert TE. Challenges in exosome isolation and analysis in health and disease. Int J Mol Sci 2019; 20: 4684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Jablonska J, Pietrowska M, Ludwig S, et al. Challenges in the isolation and proteomic analysis of cancer exosomes-implications for translational research. Proteomes 2019; 7: 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kumar SR, Kimchi ET, Manjunath Y, et al. RNA cargos in extracellular vesicles derived from blood serum in pancreas associated conditions. Sci Rep 2020; 10: 2800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Romano E, Netti PA, Torino E. A high throughput approach based on dynamic high pressure for the encapsulation of active compounds in exosomes for precision medicine. Int J Mol Sci 2021; 22: 9896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Gao J, Li A, Hu J, et al. Recent developments in isolating methods for exosomes. Front Bioeng Biotechnol 2022; 10: 1100892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Xu Y, Wan W, Zeng H, et al. Exosomes and their derivatives as biomarkers and therapeutic delivery agents for cardiovascular diseases: situations and challenges. J Transl Int Med 2023; 11: 341–354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Chen X, Luo Q. Potential clinical applications of exosomes in the diagnosis, treatment, and prognosis of cardiovascular diseases: a narrative review. Ann Transl Med 2022; 10: 372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Kavanagh JJ, Yeung KY, Savaraj N, et al. Phase I clinical evaluation of oral and intravenous 4-demethoxydaunorubicin. Eur J Cancer Clin Oncol 1985; 21: 1187–1189. [DOI] [PubMed] [Google Scholar]
- 69.Gorgens A, Corso G, Hagey DW, et al. Identification of storage conditions stabilizing extracellular vesicles preparations. J Extracell Vesicles 2022; 11: e12238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yuan F, Li YM, Wang Z. Preserving extracellular vesicles for biomedical applications: consideration of storage stability before and after isolation. Drug Deliv 2021; 28: 1501–1509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Qin B, Zhang Q, Hu XM, et al. How does temperature play a role in the storage of extracellular vesicles? J Cell Physiol 2020; 235: 7663–7680. [DOI] [PubMed] [Google Scholar]
- 72.Meggiolaro A, Moccia V, Brun P, et al. Microfluidic strategies for extracellular vesicle isolation: towards clinical applications. Biosensors (Basel) 2022; 13: 50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Yang J, Gao X, Xing X, et al. An isolation system to collect high quality and purity extracellular vesicles from serum. Int J Nanomedicine 2021; 16: 6681–6692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Chen J, Zheng M, Xiao Q, et al. Recent advances in microfluidic-based extracellular vesicle analysis. Micromachines (Basel) 2024; 15: 630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Zhang J, Wu J, Wang G, et al. Extracellular vesicles: techniques and biomedical applications related to single vesicle analysis. ACS Nano 2023; 17: 17668–17698. [DOI] [PubMed] [Google Scholar]
- 76.McIlvenna LC, Parker HJ, Seabright AP, et al. Single vesicle analysis reveals the release of tetraspanin positive extracellular vesicles into circulation with high intensity intermittent exercise. J Physiol 2023; 601: 5093–5106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Lane RE, Korbie D, Khanna KK, et al. Defining the relationship between cellular and extracellular vesicle (EV) content in breast cancer via an integrative multi-omic analysis. Proteomics 2024; 24: e2300089. [DOI] [PubMed] [Google Scholar]
- 78.Ge Y, Ye T, Fu S, et al. Research progress of extracellular vesicles as biomarkers in immunotherapy for non-small cell lung cancer. Front Immunol 2023; 14: 1114041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Li J, Fu Y, Liu Q, et al. Multiomics-based study of amniotic fluid small extracellular vesicles identified Moesin as a biomarker for antenatal hydronephrosis. Clin Transl Med 2023; 13: e1360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Chen F, Xu Y, Shi K, et al. Multi-omics study reveals associations among neurotransmitter, extracellular vesicle-derived microRNA and psychiatric comorbidities during heroin and methamphetamine withdrawal. Biomed Pharmacother 2022; 155: 113685. [DOI] [PubMed] [Google Scholar]
- 81.Cohn W, Melnik M, Huang C, et al. Multi-omics analysis of microglial extracellular vesicles from human Alzheimer's disease brain tissue reveals disease-associated signatures. Front Pharmacol 2021; 12: 766082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Yekula A, Muralidharan K, Kang KM, et al. From laboratory to clinic: translation of extracellular vesicle based cancer biomarkers. Methods 2020; 177: 58–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Cheng CA. Before translating extracellular vesicles into personalized diagnostics and therapeutics: what we could do. Mol Pharm 2024; 21: 2625–2636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Gualerzi A, Picciolini S, Bedoni M, et al. Extracellular vesicles as biomarkers for Parkinson's disease: how far from clinical translation? Int J Mol Sci 2024; 25: 1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Li Y, Sui S, Goel A. Extracellular vesicles associated microRNAs: their biology and clinical significance as biomarkers in gastrointestinal cancers. Semin Cancer Biol 2024; 99: 5–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Zhang Z, Liu X, Yang X, et al. Identification of faecal extracellular vesicles as novel biomarkers for the non-invasive diagnosis and prognosis of colorectal cancer. J Extracell Vesicles 2023; 12: e12300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bordanaba-Florit G, Royo F, Kruglik SG, et al. Using single-vesicle technologies to unravel the heterogeneity of extracellular vesicles. Nat Protoc 2021; 16: 3163–3185. [DOI] [PubMed] [Google Scholar]
- 88.Zhang Z, Yu K, You Y, et al. Comprehensive characterization of human brain-derived extracellular vesicles using multiple isolation methods: implications for diagnostic and therapeutic applications. J Extracell Vesicles 2023; 12: e12358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Brenner AW, Su GH, Momen-Heravi F. Isolation of extracellular vesicles for cancer diagnosis and functional studies. Methods Mol Biol 2019; 1882: 229–237. [DOI] [PubMed] [Google Scholar]
- 90.Xiao Y, Peng X, Peng Y, et al. Macrophage-derived extracellular vesicles regulate follicular activation and improve ovarian function in old mice by modulating local environment. Clin Transl Med 2022; 12: e1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Zhao S, Mi Y, Zheng B, et al. Highly-metastatic colorectal cancer cell released miR-181a-5p-rich extracellular vesicles promote liver metastasis by activating hepatic stellate cells and remodelling the tumour microenvironment. J Extracell Vesicles 2022; 11: e12186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Vaiaki EM, Falasca M. Comparative analysis of the minimal information for studies of extracellular vesicles guidelines: advancements and implications for extracellular vesicle research. Semin Cancer Biol 2024; 101: 12–24. [DOI] [PubMed] [Google Scholar]
- 93.Li W, Liu JB, Hou LK, et al. Liquid biopsy in lung cancer: significance in diagnostics, prediction, and treatment monitoring. Mol Cancer 2022; 21: 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Freitas AJA, Causin RL, Varuzza MB, et al. Liquid biopsy as a tool for the diagnosis, treatment, and monitoring of breast cancer. Int J Mol Sci 2022; 23: 9952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kučuk N, Primožič M, Knez Ž, et al. Exosomes engineering and their roles as therapy delivery tools, therapeutic targets, and biomarkers. Int J Mol Sci 2021; 22: 9543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Malekian F, Shamsian A, Kodam SP, et al. Exosome engineering for efficient and targeted drug delivery: current status and future perspective. J Physiol 2023; 601: 4853–4872. [DOI] [PubMed] [Google Scholar]
- 97.Huda MN, Nafiujjaman M, Deaguero IG, et al. Potential use of exosomes as diagnostic biomarkers and in targeted drug delivery: progress in clinical and preclinical applications. ACS Biomater Sci Eng 2021; 7: 2106–2149. [DOI] [PMC free article] [PubMed] [Google Scholar]