Characterization Technique for Exosomes: Comparison
Please note this is a comparison between Version 3 by Lindsay Dong and Version 2 by shijia wu.

Exosomes distributed by extracellular vesicles carry various information highly consistent with cells, becoming a new type of biomarker for tumor screening. However, although conventional characterization technologies can quantify size and morphology for exosomes, they are limited in related fields such as function tracing, protein quantification at unit point, and microstructural information. In this paper, firstly, different exosome characterization methods are systematically reviewed, such as dynamic light scattering, nanoparticle tracking analysis, flow cytometry, electron microscope, and emerging super-resolution imaging technologies. Then, advances in applications are described one by one. Last but not least, we compare the features of different technologies for exosomes and propose that super-resolution imaging technology can not only take into account the advantages of conventional characterization techniques but also provide accurate, real-time, and super-resolution quantitative analysis for exosomes. It provides a fine guide for exosome-related biomedical research, as well as application in liquid biopsy and analysis techniques.

  • exosome
  • tumor diagnosis
  • optical analysis technology
  • super-resolution microscope

1. Introduction

Exosomes, a novel tumor biomarker after circulating tumor cells and circulating tumor DNA [1,2,3][1][2][3], can be used in tumor diagnosis [4]. Biosignaling molecules of exosomes are exchanged by endocytosis, and thereby tumor cell activities such as growth, metastasis, drug resistance, and immune evasion are regulated [5,6][5][6]. Therefore, exosomes have important value in the early diagnosis of tumors, the monitoring of treatment progress, and prognosis as shown in Figure 1 [4,7,8,9][4][7][8][9].
Figure 1. Exosome information transfer process and application [4].
Exosomes are membranous extracellular vesicles (EV) secreted by cells with a particle size of 30 to 150 nm [10]. The study found that the function of exosomes depends on the cell type from which they are derived, while it maintains the same genetic material as the donor cells [11]. Different analyses become one of the tumor-specific research targets during exosome activity. For example, exosomes derived from tumor cells carry many types of proteins, such as surface proteins, inclusions, enzymes, etc. Among them, the surface proteins such as CD9, CD63, and CD81 and the inclusion factors such as HSP70 and Alix are representative proteins for the isolation and identification of exosomes [12,13,14][12][13][14]. The differences in proteins can reflect information exchange between tumor cells and basal cells and between tumor cells and tumor cells, which regulates immune response, migration, differentiation, and other basic cellular functions [15,16,17,18][15][16][17][18]. For example, studies have found that exosomes derived from different cells are different in size, morphology, and composition [19]. The exosomes derived from tumor cells are large and contain more lipids and outer membrane proteins, which promote tumor cell growth, invasion, and metastasis [20]. The surface proteins of cancer exosomes are often different in different stages, which indicates that these proteins are closely related to the process of cancer. Similarly, the surface proteins of cancer exosomes from different sources are also different, which can be used for the early diagnosis of cancer [21,22][21][22]. Research has proven that ADAM10, metalloprotease, CD9, Annexin-1, and HSP70 are enriched in exosomes isolated from the pleural effusion or serum of breast cancer patients [23]. However, the exosomes derived from immune cells are smaller and include a variety of immune molecules, such as cytokines, antigens, antibodies, etc., which could regulate immune responses and antitumor effects [24]. More importantly, quantitative analysis is necessary to identify exosomes accurately. In addition, some studies believe that the morphology, quantity, and concentration of exosomes are different in the process of secretion [25], so real-time tracking and quantification can objectively analyze the delicate mechanism to achieve optimal anti-tumor effect. In conclusion, the morphological characterization and function of exosomes are the basis for exploring the fine physiological information and biochemical mechanisms in cellular biology [26,27][26][27]. Exosomes are emerging biomarkers of tumor liquid biopsy, and therefore it is particularly important to explore their biological information such as function tracing, protein quantification at unit point, and microstructural changes [28,29][28][29]. At present, much attention has been paid to their basic characteristics such as concentration, diameter, morphology, and particle tracking in high-throughput samples [30,31,32][30][31][32]. In this review, we propose that the characterization techniques for obtaining this information are exosome detection techniques. Conventional characterization methods can achieve the basic characterization of exosomes, yet they are subject to certain limitations. DLS can only obtain the size distribution of exosomes but cannot detect concentration [33]. Although flow cytometry (FCM) realizes multi-parameter detection, the analysis results are based on high-throughput samples and cannot obtain the morphological characteristics of a single EV [34]. EM can visually observe the morphological characteristics of a single EV, but it is not suitable for living cells, and the direction is limited in biological research [35]. Therefore, there is an urgent need for an exosome detection technique that can preserve the fluorescence specificity and achieve high-throughput sample single-molecule detection. In recent years, researchers have paid more attention to fluorescence microscopy. The fluorescence microscope has the advantage of live cell imaging and specific labeling; however, its resolution ranges from 200 nm to 500 nm, which cannot be applied to detecting exosomes. Therefore, how to improve the imaging resolution of optical microscopy and apply it to the imaging observation of subcellular structures such as exosomes has become a research hotspot in recent years. With the development of fluorescence microscopy, super-resolution imaging technology breaks through the diffraction limitation, which is conducive to the field of exosomes [26,36][26][36]. Compared with others, it has the advantages of high resolution, specific detection, and live-cell imaging to realize the qualitative and quantitative analysis of proteins of living or fixed cells and track the mechanism of exosomes [36]. In order to better guide the research on exosomes, we systematically discussed and compared characterization techniques for exosomes. This review system elaborates on the principles and applications of conventional characterization technologies for exosomes such as dynamic light scattering (DLS), nanoparticle tracking analysis (NTA), flow cytometry (FCM), electron microscopy (EM), and scanning electron microscopy (SEM), as well as super-resolution technologies represented by stochastic optical reconstruction microscopy (STORM), photoactivated localization microscopy (PALM), stimulated emission depletion (STED), and structured illumination microscopy (SIM). More importantly, this paper discusses the medical problems and characterization techniques of exosomes from morphological characterization to the functional expression of exosomes, from complex samples to single exosomes, and from multiple protein quantification to a single protein of a single exosome. The mechanism and information communication of exosomes were systematically settled from the perspective of technology development, which provides more systematic and comprehensive guidance in the field of basic research.

2. Conventional Characterization Technologies

Due to the unique biological function of exosomes, an increasing amount of basic research is being concentrated on it [37,38,39,40,41][37][38][39][40][41]. Characterization technologies play important roles in the study of exosomes [30]. Generally speaking, various approaches for analysis are categorized into two primary types: biochemical analysis and physical analysis. Biochemical analysis mainly determines the source and composition of exosomes, including Western blot and enzyme-linked immunosorbent assay (ELISA), in which the specific binding of antibody antigens decides the effect qualitatively or quantitatively [42]. However, the disadvantage is that the morphological characteristics and concentration of exosomes cannot be obtained. We introduce electron technologies such as tunable resistive pulse sensing (TRPS) and electron microscope (EM) in the first section. Then, we compare optical analysis technologies including dynamic light scattering (DLS), nanoparticle tracking analysis (NTA), and flow cytometry (FCM). Last but not least, we discuss the main parameters of different technologies, providing technical guidance for the fundamental research on exosome characterization. 

2.1. Tunable Resistive Pulse Sensing

Tunable Resistive Pulse Sensing (TRPS) is based on Coulter’s principle. The suspension was mixed in the electrolyte, which could go through the nanopore chip with a specific aperture. The resistance between the two electrodes inside and outside changes instantaneously at the moment of passing through the nanopore, the result of which is a pulse signal as shown in Figure 2a. The intensity and frequency of the signal are related to the size and number of exosomes. The expression of exosomes was counted by the pulse signal. In 2014, Maas proposed a method to characterize the concentration and size of EVs by the TRPS based on the qNano system [43]. In 2017, researchers pointed out that TRPS has promise in the quantitative and dimensional analysis of single-particle EV [44]. In 2018, Durak-Kozica analyzed EVs from endothelial cells for a short time and found that the diameter of EVs was 121.84 ± 0.08 and 115.82 ± 0.96 nm from microvascular and big vessels, respectively [45]. This technology enables the efficient quantification of size and number, which cannot be specifically analyzed for exosomes due to the principle of potential pulses.

 

References

  1. Wan, J.C.M.; Massie, C.; Garcia-Corbacho, J.; Mouliere, F.; Brenton, J.D.; Caldas, C.; Pacey, S.; Baird, R.; Rosenfeld, N. Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat. Rev. Cancer 2017, 17, 223–238.
  2. Alix-Panabières, C.; Pantel, K. Circulating tumor cells: Liquid biopsy of cancer. Clin. Chem. 2013, 59, 110–118.
  3. Guo, L.C.; He, B. Extracellular vesicles and their diagnostic and prognostic potential in cancer. Transl. Cancer Res. 2017, 6, 599–612.
  4. Huang, T.; Deng, C.X. Current Progresses of Exosomes as Cancer Diagnostic and Prognostic Biomarkers. Int. J. Biol. Sci. 2019, 15, 1–11.
  5. Im, E.J.; Lee, C.H.; Moon, P.G.; Rangaswamy, G.G.; Lee, B.; Lee, J.M.; Lee, J.C.; Jee, J.G.; Bae, J.S.; Kwon, T.K.; et al. Sulfisoxazole inhibits the secretion of small extracellular vesicles by targeting the endothelin receptor A. Nat. Commun. 2019, 10, 1387.
  6. Junker, K.; Heinzelmann, J.; Beckham, C.; Ochiya, T.; Jenster, G. Extracellular Vesicles and Their Role in Urologic Malignancies. Eur. Urol. 2016, 70, 323–331.
  7. Akers, J.C.; Ramakrishnan, V.; Kim, R.; Phillips, S.; Kaimal, V.; Mao, Y.; Hua, W.; Yang, I.; Fu, C.C.; Nolan, J.; et al. miRNA contents of cerebrospinal fluid extracellular vesicles in glioblastoma patients. J. Neuro-Oncol. 2015, 123, 205–216.
  8. Minciacchi, V.R.; Freeman, M.R.; Di Vizio, D. Extracellular vesicles in cancer: Exosomes, microvesicles and the emerging role of large oncosomes. Semin. Cell Dev. Biol. 2015, 40, 41–51.
  9. Ciardiello, C.; Cavallini, L.; Spinelli, C.; Yang, J.; Reis-Sobreiro, M.; de Candia, P.; Minciacchi, V.R.; Di Vizio, D. Focus on Extracellular Vesicles: New Frontiers of Cell-to-Cell Communication in Cancer. Int. J. Mol. Sci. 2016, 17, 175.
  10. Liu, J.; Ren, L.; Li, S.; Li, W.; Zheng, X.; Yang, Y.; Fu, W.; Yi, J.; Wang, J.; Du, G. The biology, function, and applications of exosomes in cancer. Acta Pharm. Sin. B 2021, 11, 2783–2797.
  11. Duijvesz, D.; Luider, T.; Bangma, C.H.; Jenster, G. Exosomes as biomarker treasure chests for prostate cancer. Eur. Urol. 2011, 59, 823–831.
  12. Kimiz-Gebologlu, I.; Oncel, S.S. Exosomes: Large-scale production, isolation, drug loading efficiency, and biodistribution and uptake. J. Control. Release 2022, 347, 533–543.
  13. Andreu, Z.; Yáñez-Mó, M. Tetraspanins in extracellular vesicle formation and function. Front. Immunol. 2014, 5, 442.
  14. Berditchevski, F.; Odintsova, E. Tetraspanins as regulators of protein trafficking. Traffic 2007, 8, 89–96.
  15. 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.
  16. Boucheix, C.; Rubinstein, E. Tetraspanins. Cell. Mol. Life Sci. 2001, 58, 1189–1205.
  17. He, C.; Zheng, S.; Luo, Y.; Wang, B. Exosome Theranostics: Biology and Translational Medicine. Theranostics 2018, 8, 237–255.
  18. He, M.; Crow, J.; Roth, M.; Zeng, Y.; Godwin, A.K. Integrated immunoisolation and protein analysis of circulating exosomes using microfluidic technology. Lab Chip 2014, 14, 3773–3780.
  19. Nikishin, I.; Dulimov, R.; Skryabin, G.; Galetsky, S.; Tchevkina, E.; Bagrov, D. ScanEV—A neural network-based tool for the automated detection of extracellular vesicles in TEM images. Micron 2021, 145, 103044.
  20. Kahlert, C.; Kalluri, R. Exosomes in tumor microenvironment influence cancer progression and metastasis. J. Mol. Med. 2013, 91, 431–437.
  21. Wang, X.; Huang, J.; Chen, W.; Li, G.; Li, Z.; Lei, J. The updated role of exosomal proteins in the diagnosis, prognosis, and treatment of cancer. Exp. Mol. Med. 2022, 54, 1390–1400.
  22. 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.
  23. Risha, Y.; Minic, Z.; Ghobadloo, S.M.; Berezovski, M.V. The proteomic analysis of breast cell line exosomes reveals disease patterns and potential biomarkers. Sci. Rep. 2020, 10, 13572.
  24. Haraszti, R.A.; Didiot, M.C.; Sapp, E.; Leszyk, J.; Shaffer, S.A.; Rockwell, H.E.; Gao, F.; Narain, N.R.; DiFiglia, M.; Kiebish, M.A.; et al. High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources. J. Extracell. Vesicles 2016, 5, 32570.
  25. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. Bioscience 2015, 65, 783–797.
  26. Yang, B.; Chen, Y.; Shi, J. Exosome Biochemistry and Advanced Nanotechnology for Next-Generation Theranostic Platforms. Adv. Mater. 2019, 31, e1802896.
  27. 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.
  28. Hsu, M.T.; Wang, Y.K.; Tseng, Y.J. Exosomal Proteins and Lipids as Potential Biomarkers for Lung Cancer Diagnosis, Prognosis, and Treatment. Cancers 2022, 14, 732.
  29. Lee, Y.R.; Kim, G.; Tak, W.Y.; Jang, S.Y.; Kweon, Y.O.; Park, J.G.; Lee, H.W.; Han, Y.S.; Chun, J.M.; Park, S.Y.; et al. Circulating exosomal noncoding RNAs as prognostic biomarkers in human hepatocellular carcinoma. Int. J. Cancer 2019, 144, 1444–1452.
  30. Zhu, L.; Sun, H.T.; Wang, S.; Huang, S.L.; Zheng, Y.; Wang, C.Q.; Hu, B.Y.; Qin, W.; Zou, T.T.; Fu, Y.; et al. Isolation and characterization of exosomes for cancer research. J. Hematol. Oncol. 2020, 13, 152.
  31. Liu, Y.; Zhou, Z.; Wang, F.; Kewes, G.; Wen, S.; Burger, S.; Ebrahimi Wakiani, M.; Xi, P.; Yang, J.; Yang, X.; et al. Axial localization and tracking of self-interference nanoparticles by lateral point spread functions. Nat. Commun. 2021, 12, 2019.
  32. Wu, H.J.; Huang, C.L.; Wang, L.W.; Li, Q.H.; Li, Y.J.; Zhang, L.H.; Zhu, D.W. Folate-targeted co-delivery polymersomes for efficient photo-chemo-antiangiogenic therapy against breast cancer and in vivo evaluation via OCTA_NIRF dual-modal imaging-3. Chin. Chem. Lett. 2022, 33, 5035–5041.
  33. Filipe, V.; Hawe, A.; Jiskoot, W. Critical evaluation of Nanoparticle Tracking Analysis (NTA) by NanoSight for the measurement of nanoparticles and protein aggregates. Pharm. Res. 2010, 27, 796–810.
  34. Libregts, S.; Arkesteijn, G.J.A.; Németh, A.; Nolte-'t Hoen, E.N.M.; Wauben, M.H.M. Flow cytometric analysis of extracellular vesicle subsets in plasma: Impact of swarm by particles of non-interest. J. Thromb. Haemost. 2018, 16, 1423–1436.
  35. Arraud, N.; Linares, R.; Tan, S.; Gounou, C.; Pasquet, J.M.; Mornet, S.; Brisson, A.R. Extracellular vesicles from blood plasma: Determination of their morphology, size, phenotype and concentration. J. Thromb. Haemost. 2014, 12, 614–627.
  36. Lai, J.J.; Chau, Z.L.; Chen, S.Y.; Hill, J.J.; Korpany, K.V.; Liang, N.W.; Lin, L.H.; Lin, Y.H.; Liu, J.K.; Liu, Y.C.; et al. Exosome Processing and Characterization Approaches for Research and Technology Development. Adv. Sci. 2022, 9, e2103222.
  37. Mashouri, L.; Yousefi, H.; Aref, A.R.; Ahadi, A.M.; Molaei, F.; Alahari, S.K. Exosomes: Composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol. Cancer 2019, 18, 75.
  38. Dai, J.; Su, Y.; Zhong, S.; Cong, L.; Liu, B.; Yang, J.; Tao, Y.; He, Z.; Chen, C.; Jiang, Y. Exosomes: Key players in cancer and potential therapeutic strategy. Signal Transduct. Target. Ther. 2020, 5, 145.
  39. Jiang, C.; Zhang, N.; Hu, X.; Wang, H. Tumor-associated exosomes promote lung cancer metastasis through multiple mechanisms. Mol. Cancer 2021, 20, 117.
  40. Zhao, S.; Mi, Y.; Guan, B.; Zheng, B.; Wei, P.; Gu, Y.; Zhang, Z.; Cai, S.; Xu, Y.; Li, X.; et al. Tumor-derived exosomal miR-934 induces macrophage M2 polarization to promote liver metastasis of colorectal cancer. J. Hematol. Oncol. 2020, 13, 156.
  41. Zhang, L.; Yu, D. Exosomes in cancer development, metastasis, and immunity. Biochim. Biophys. (BBA) Acta Rev. Cancer 2019, 1871, 455–468.
  42. Hartjes, T.A.; Mytnyk, S.; Jenster, G.W.; van Steijn, V.; van Royen, M.E. Extracellular Vesicle Quantification and Characterization: Common Methods and Emerging Approaches. Bioengineering 2019, 6, 7.
  43. Maas, S.L.; De Vrij, J.; Broekman, M.L. Quantification and size-profiling of extracellular vesicles using tunable resistive pulse sensing. J. Vis. Exp. 2014, 92, e51623.
  44. Maas, S.L.; Broekman, M.L.; de Vrij, J. Tunable Resistive Pulse Sensing for the Characterization of Extracellular Vesicles. Methods Mol. Biol. 2017, 1545, 21–33.
  45. Durak-Kozica, M.; Baster, Z.; Kubat, K.; Stępień, E. 3D visualization of extracellular vesicle uptake by endothelial cells. Cell. Mol. Biol. Lett. 2018, 23, 57.
  46. Vogel, R.; Pal, A.K.; Jambhrunkar, S.; Patel, P.; Thakur, S.S.; Reátegui, E.; Parekh, H.S.; Saá, P.; Stassinopoulos, A.; Broom, M.F. High-Resolution Single Particle Zeta Potential Characterisation of Biological Nanoparticles using Tunable Resistive Pulse Sensing. Sci. Rep. 2017, 7, 17479.
  47. Malenica, M.; Vukomanović, M.; Kurtjak, M.; Masciotti, V.; Dal Zilio, S.; Greco, S.; Lazzarino, M.; Krušić, V.; Perčić, M.; Jelovica Badovinac, I.; et al. Perspectives of Microscopy Methods for Morphology Characterisation of Extracellular Vesicles from Human Biofluids. Biomedicines 2021, 9, 603.
  48. McNamara, R.P.; Zhou, Y.; Eason, A.B.; Landis, J.T.; Chambers, M.G.; Willcox, S.; Peterson, T.A.; Schouest, B.; Maness, N.J.; MacLean, A.G.; et al. Imaging of surface microdomains on individual extracellular vesicles in 3-D. J. Extracell. Vesicles 2022, 11, e12191.
  49. Fakhredini, F.; Mansouri, E.; Mard, S.A.; Valizadeh Gorji, A.; Rashno, M.; Orazizadeh, M. Effects of Exosomes Derived from Kidney Tubular Cells on Diabetic Nephropathy in Rats. Cell J. 2022, 24, 28–35.
  50. Conde-Vancells, J.; Rodriguez-Suarez, E.; Embade, N.; Gil, D.; Matthiesen, R.; Valle, M.; Elortza, F.; Lu, S.C.; Mato, J.M.; Falcon-Perez, J.M. Characterization and comprehensive proteome profiling of exosomes secreted by hepatocytes. J. Proteome Res. 2008, 7, 5157–5166.
  51. Jeong, D.; Kim, M.J.; Park, Y.; Chung, J.; Kweon, H.S.; Kang, N.G.; Hwang, S.J.; Youn, S.H.; Hwang, B.K.; Kim, D. Visualizing extracellular vesicle biogenesis in gram-positive bacteria using super-resolution microscopy. BMC Biol. 2022, 20, 270.
  52. Zabeo, D.; Cvjetkovic, A.; Lässer, C.; Schorb, M.; Lötvall, J.; Höög, J.L. Exosomes purified from a single cell type have diverse morphology. J. Extracell. Vesicles 2017, 6, 1329476.
  53. Gardiner, C.; Di Vizio, D.; Sahoo, S.; Théry, C.; Witwer, K.W.; Wauben, M.; Hill, A.F. Techniques used for the isolation and characterization of extracellular vesicles: Results of a worldwide survey. J. Extracell. Vesicles 2016, 5, 32945.
  54. Issman, L.; Brenner, B.; Talmon, Y.; Aharon, A. Cryogenic transmission electron microscopy nanostructural study of shed microparticles. PLoS ONE 2013, 8, e83680.
  55. Jung, M.K.; Mun, J.Y. Sample Preparation and Imaging of Exosomes by Transmission Electron Microscopy. J. Vis. Exp. 2018, 131, e56482.
  56. Božič, D.; Hočevar, M.; Kisovec, M.; Pajnič, M.; Pađen, L.; Jeran, M.; Bedina Zavec, A.; Podobnik, M.; Kogej, K.; Iglič, A.; et al. Stability of Erythrocyte-Derived Nanovesicles Assessed by Light Scattering and Electron Microscopy. Int. J. Mol. Sci. 2021, 22, 12772.
  57. Sokolova, V.; Ludwig, A.K.; Hornung, S.; Rotan, O.; Horn, P.A.; Epple, M.; Giebel, B. Characterisation of exosomes derived from human cells by nanoparticle tracking analysis and scanning electron microscopy. Colloids Surf. B Biointerfaces 2011, 87, 146–150.
  58. Cizmar, P.; Yuana, Y. Detection and Characterization of Extracellular Vesicles by Transmission and Cryo-Transmission Electron Microscopy. Methods Mol. Biol. 2017, 1660, 221–232.
  59. Liu, Z.; Xue, H.; Chen, Q.; Yang, G. A method for extraction of exosomes from breast tumour cells and characterisation by transmission electron microscopy. J. Microsc. 2023, 292, 117–122.
  60. Park, Y.H.; Shin, H.W.; Jung, A.R.; Kwon, O.S.; Choi, Y.J.; Park, J.; Lee, J.Y. Prostate-specific extracellular vesicles as a novel biomarker in human prostate cancer. Sci. Rep. 2016, 6, 30386.
  61. Kurtjak, M.; Kereïche, S.; Klepac, D.; Križan, H.; Perčić, M.; Krušić Alić, V.; Lavrin, T.; Lenassi, M.; Wechtersbach, K.; Kojc, N.; et al. Unveiling the Native Morphology of Extracellular Vesicles from Human Cerebrospinal Fluid by Atomic Force and Cryogenic Electron Microscopy. Biomedicines 2022, 10, 1251.
  62. Stetefeld, J.; McKenna, S.A.; Patel, T.R. Dynamic light scattering: A practical guide and applications in biomedical sciences. Biophys. Rev. 2016, 8, 409–427.
  63. Palmieri, V.; Lucchetti, D.; Gatto, I.; Maiorana, A.; Marcantoni, M.; Maulucci, G.; Papi, M.; Pola, R.; De Spirito, M.; Sgambato, A. Dynamic light scattering for the characterization and counting of extracellular vesicles: A powerful noninvasive tool. J. Nanopart. Res. 2014, 16, 2583.
  64. Perpetuo, L.; Ferreira, R.; Thongboonkerd, V.; Guedes, S.; Amado, F.; Vitorino, R. Urinary exosomes: Diagnostic impact with a bioinformatic approach. Adv. Clin. Chem. 2022, 111, 69–99.
  65. Gercel-Taylor, C.; Atay, S.; Tullis, R.H.; Kesimer, M.; Taylor, D.D. Nanoparticle analysis of circulating cell-derived vesicles in ovarian cancer patients. Anal. Biochem. 2012, 428, 44–53.
  66. Tajik, T.; Baghaei, K.; Moghadam, V.E.; Farrokhi, N.; Salami, S.A. Extracellular vesicles of cannabis with high CBD content induce anticancer signaling in human hepatocellular carcinoma. Biomed. Pharmacother. 2022, 152, 113209.
  67. Hassan, P.A.; Rana, S.; Verma, G. Making sense of Brownian motion: Colloid characterization by dynamic light scattering. Langmuir 2015, 31, 3–12.
  68. Lawrie, A.S.; Albanyan, A.; Cardigan, R.A.; Mackie, I.J.; Harrison, P. Microparticle sizing by dynamic light scattering in fresh-frozen plasma. Vox Sang. 2009, 96, 206–212.
  69. Szatanek, R.; Baj-Krzyworzeka, M.; Zimoch, J.; Lekka, M.; Siedlar, M.; Baran, J. The Methods of Choice for Extracellular Vesicles (EVs) Characterization. Int. J. Mol. Sci. 2017, 18, 1153.
  70. Cho, S.; Yi, J.; Kwon, Y.; Kang, H.; Han, C.; Park, J. Multifluorescence Single Extracellular Vesicle Analysis by Time-Sequential Illumination and Tracking. ACS Nano 2021, 15, 11753–11761.
  71. Vestad, B.; Llorente, A.; Neurauter, A.; Phuyal, S.; Kierulf, B.; Kierulf, P.; Skotland, T.; Sandvig, K.; Haug, K.B.F.; Øvstebø, R. Size and concentration analyses of extracellular vesicles by nanoparticle tracking analysis: A variation study. J. Extracell. Vesicles 2017, 6, 1344087.
  72. Dragovic, R.A.; Gardiner, C.; Brooks, A.S.; Tannetta, D.S.; Ferguson, D.J.; Hole, P.; Carr, B.; Redman, C.W.; Harris, A.L.; Dobson, P.J.; et al. Sizing and phenotyping of cellular vesicles using Nanoparticle Tracking Analysis. Nanomed. Nanotechnol. Biol. Med. 2011, 7, 780–788.
  73. van der Pol, E.; Coumans, F.A.; Grootemaat, A.E.; Gardiner, C.; Sargent, I.L.; Harrison, P.; Sturk, A.; van Leeuwen, T.G.; Nieuwland, R. Particle size distribution of exosomes and microvesicles determined by transmission electron microscopy, flow cytometry, nanoparticle tracking analysis, and resistive pulse sensing. J. Thromb. Haemost. 2014, 12, 1182–1192.
  74. Hu, Y.; Tian, Y.; Di, H.; Xue, C.; Zheng, Y.; Hu, B.; Lin, Q.; Yan, X. Noninvasive Diagnosis of Nasopharyngeal Carcinoma Based on Phenotypic Profiling of Viral and Tumor Markers on Plasma Extracellular Vesicles. Anal. Chem. 2022, 94, 9740–9749.
  75. Liu, H.S.; Tian, Y.; Xue, C.F.; Niu, Q.; Chen, C.; Yan, X.M. Analysis of extracellular vesicle DNA at the single-vesicle level by nano-flow cytometry. J. Extracell. Vesicles 2022, 11, e12206.
  76. Ricklefs, F.L.; Maire, C.L.; Reimer, R.; Dührsen, L.; Kolbe, K.; Holz, M.; Schneider, E.; Rissiek, A.; Babayan, A.; Hille, C.; et al. Imaging flow cytometry facilitates multiparametric characterization of extracellular vesicles in malignant brain tumours. J. Extracell. Vesicles 2019, 8, 1588555.
  77. Aibaidula, A.Z.; Fain, C.E.; Garcia, L.C.; Wier, A.; Bouchal, S.M.; Bauman, M.M.; Jung, M.Y.; Sarkaria, J.N.; Johnson, A.J.; Parney, I.F. Spectral flow cytometry identifies distinct nonneoplastic plasma extracellular vesicle phenotype in glioblastoma patients. Neuro-Oncol. Adv. 2023, 5, vdad082.
  78. Betzig, E.; Patterson, G.H.; Sougrat, R.; Lindwasser, O.W.; Olenych, S.; Bonifacino, J.S.; Davidson, M.W.; Lippincott-Schwartz, J.; Hess, H.F. Imaging intracellular fluorescent proteins at nanometer resolution. Science 2006, 313, 1642–1645.
  79. Rust, M.J.; Bates, M.; Zhuang, X. Sub-diffraction-limit imaging by stochastic optical reconstruction microscopy (STORM). Nat. Methods 2006, 3, 793–795.
  80. Jones, S.A.; Shim, S.H.; He, J.; Zhuang, X. Fast, three-dimensional super-resolution imaging of live cells. Nat. Methods 2011, 8, 499–508.
  81. Shim, S.H.; Xia, C.; Zhong, G.; Babcock, H.P.; Vaughan, J.C.; Huang, B.; Wang, X.; Xu, C.; Bi, G.Q.; Zhuang, X. Super-resolution fluorescence imaging of organelles in live cells with photoswitchable membrane probes. Proc. Natl. Acad. Sci. USA 2012, 109, 13978–13983.
  82. Zong, S.; Zong, J.; Chen, C.; Jiang, X.; Zhang, Y.; Wang, Z.; Cui, Y. Single molecule localization imaging of exosomes using blinking silicon quantum dots. Nanotechnology 2018, 29, 065705.
  83. Jungmann, R.; Avendaño, M.S.; Woehrstein, J.B.; Dai, M.; Shih, W.M.; Yin, P. Multiplexed 3D cellular super-resolution imaging with DNA-PAINT and Exchange-PAINT. Nat. Methods 2014, 11, 313–318.
  84. Tholen, M.M.E.; Tas, R.P.; Wang, Y.; Albertazzi, L. Beyond DNA: New probes for PAINT super-resolution microscopy. Chem. Commun. 2023, 59, 8332–8342.
  85. Chen, C.; Zong, S.; Liu, Y.; Wang, Z.; Zhang, Y.; Chen, B.; Cui, Y. Profiling of Exosomal Biomarkers for Accurate Cancer Identification: Combining DNA-PAINT with Machine- Learning-Based Classification. Small 2019, 15, e1901014.
  86. Liu, D.A.; Tao, K.; Wu, B.; Yu, Z.; Szczepaniak, M.; Rames, M.; Yang, C.; Svitkina, T.; Zhu, Y.; Xu, F.; et al. A phosphoinositide switch mediates exocyst recruitment to multivesicular endosomes for exosome secretion. Nat. Commun. 2023, 14, 6883.
  87. Auer, A.; Strauss, M.T.; Schlichthaerle, T.; Jungmann, R. Fast, Background-Free DNA-PAINT Imaging Using FRET-Based Probes. Nano Lett. 2017, 17, 6428–6434.
  88. Zhu, M.; Zhang, L.; Jin, L.; Chen, J.; Zhang, Y.; Xu, Y. DNA-PAINT Imaging Accelerated by Machine Learning. Front. Chem. 2022, 10, 864701.
  89. van Wee, R.; Filius, M.; Joo, C. Completing the canvas: Advances and challenges for DNA-PAINT super-resolution imaging. Trends Biochem. Sci. 2021, 46, 918–930.
  90. McEvoy, A.L.; Greenfield, D.; Bates, M.; Liphardt, J. Q&A: Single-molecule localization microscopy for biological imaging. BMC Biol. 2010, 8, 106.
  91. Nieves, D.J.; Gaus, K.; Baker, M.A.B. DNA-Based Super-Resolution Microscopy: DNA-PAINT. Genes 2018, 9, 621.
  92. Li, W.; Li, C.; Zhou, T.; Liu, X.; Liu, X.; Li, X.; Chen, D. Role of exosomal proteins in cancer diagnosis. Mol. Cancer 2017, 16, 145.
  93. Hell, S.W.; Wichmann, J. Breaking the diffraction resolution limit by stimulated emission: Stimulated-emission-depletion fluorescence microscopy. Opt. Lett. 1994, 19, 780–782.
  94. Sezgin, E. Super-resolution optical microscopy for studying membrane structure and dynamics. J. Phys. Condens. Matter 2017, 29, 273001.
  95. Huang, G.; Liu, Y.; Wang, D.; Zhu, Y.; Wen, S.; Ruan, J.; Jin, D. Upconversion nanoparticles for super-resolution quantification of single small extracellular vesicles. eLight 2022, 2, 20.
  96. Valcz, G.; Buzás, E.I.; Kittel, Á.; Krenács, T.; Visnovitz, T.; Spisák, S.; Török, G.; Homolya, L.; Zsigrai, S.; Kiszler, G.; et al. En bloc release of MVB-like small extracellular vesicle clusters by colorectal carcinoma cells. J. Extracell. Vesicles 2019, 8, 1596668.
  97. Yang, X.; Yang, Z.; Wu, Z.; He, Y.; Shan, C.; Chai, P.; Ma, C.; Tian, M.; Teng, J.; Jin, D.; et al. Mitochondrial dynamics quantitatively revealed by STED nanoscopy with an enhanced squaraine variant probe. Nat. Commun. 2020, 11, 3699.
  98. Choi, D.; Montermini, L.; Jeong, H.; Sharma, S.; Meehan, B.; Rak, J. Mapping Subpopulations of Cancer Cell-Derived Extracellular Vesicles and Particles by Nano-Flow Cytometry. ACS Nano 2019, 13, 10499–10511.
  99. Gustafsson, M.G. Surpassing the lateral resolution limit by a factor of two using structured illumination microscopy. J. Microsc. 2000, 198, 82–87.
  100. Gustafsson, M.G. Nonlinear structured-illumination microscopy: Wide-field fluorescence imaging with theoretically unlimited resolution. Proc. Natl. Acad. Sci. USA 2005, 102, 13081–13086.
  101. Li, D.; Shao, L.; Chen, B.C.; Zhang, X.; Zhang, M.; Moses, B.; Milkie, D.E.; Beach, J.R.; Hammer, J.A., 3rd; Pasham, M.; et al. ADVANCED IMAGING. Extended-resolution structured illumination imaging of endocytic and cytoskeletal dynamics. Science 2015, 349, aab3500.
  102. Boland, M.A.; Cohen, E.A.K.; Flaxman, S.R.; Neil, M.A.A. Improving axial resolution in Structured Illumination Microscopy using deep learning. Philos. Trans. Ser. A Math. Phys. Eng. Sci. 2021, 379, 20200298.
  103. He, Y.; Yao, Y.; He, Y.; Huang, Z.; Luo, F.; Zhang, C.; Qi, D.; Jia, T.; Wang, Z.; Sun, Z.; et al. Surpassing the resolution limitation of structured illumination microscopy by an untrained neural network. Biomed. Opt. Express 2023, 14, 106–117.
  104. Butola, A.; Acuna, S.; Hansen, D.H.; Agarwal, K. Scalable-resolution structured illumination microscopy. Opt. Express 2022, 30, 43752–43767.
More
Video Production Service