We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Characterization and bioassays of extracellular vesicles extracted by tangential flow filtration

    Xiaoli Zeng

    Translational Medicine Research Laboratory, PLA Air Force Hospital of Southern Theatre Command, Guangzhou, 510602, China

    Guangzhou Dude Biotechnology Co., Ltd., Guangzhou, 510320, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Xuerui Yi

    Central Research Laboratory, PLA Air Force Hospital of Southern Theatre Command, Guangzhou, 510602, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Lixuan Chen

    Guangzhou Dude Biotechnology Co., Ltd., Guangzhou, 510320, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Haisong Zhang

    Central Research Laboratory, PLA Air Force Hospital of Southern Theatre Command, Guangzhou, 510602, China

    ,
    Rongcheng Zhou

    Guangzhou Dude Biotechnology Co., Ltd., Guangzhou, 510320, China

    ,
    Jiwei Wu

    Guangzhou Dude Biotechnology Co., Ltd., Guangzhou, 510320, China

    ,
    Yuguang Chen

    Guangzhou Dude Biotechnology Co., Ltd., Guangzhou, 510320, China

    ,
    Wanyi Huang

    Guangzhou Dude Biotechnology Co., Ltd., Guangzhou, 510320, China

    ,
    Linyan Zhang

    Guangzhou Dude Biotechnology Co., Ltd., Guangzhou, 510320, China

    ,
    Jie Zheng

    Guangzhou Dude Biotechnology Co., Ltd., Guangzhou, 510320, China

    ,
    Yang Xiao

    **Author for correspondence: Tel.: +86 020 3195 5320;

    E-mail Address: jdxiao111@163.com

    Guangzhou Dude Biotechnology Co., Ltd., Guangzhou, 510320, China

    Stem Cell Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China

    &
    Fuqiang Yang

    *Author for correspondence: Tel.: +86 020 8737 6240;

    E-mail Address: yangfq23@163.com

    Translational Medicine Research Laboratory, PLA Air Force Hospital of Southern Theatre Command, Guangzhou, 510602, China

    Published Online:https://doi.org/10.2217/rme-2021-0038

    Aim: To evaluate the efficiency of tangential flow filtration (TFF) in improving the yield of human umbilical cord mesenchymal stem cell (hucMSC)-derived extracellular vesicles (EVs) while promoting cell regeneration under oxidative stress. Methods: HucMSC-EVs were extracted from supernatants by ultracentrifugation (UC-EVs) and TFF (TFF-EVs), followed by feature characterization and bioactivity assays. Results: The yield of TFF-EVs increased 18-times compared with that of UC-EVs. TFF-EVs displayed proliferation-promoting ability similar to that of UC-EVs in the damaged HaCaT cell model with ultraviolet radiation B (UVB) and H2O2. Furthermore, the antiapoptotic effects of TFF-EVs were improved, whereby the apoptosis rate exhibited a 3.7-fold decrease. Conclusion: HucMSC-EVs extracted by TFF show a higher yield and rejuvenate the damaged HaCaT cells induced by oxidative stress.

    Plain language summary

    The progresses in regenerative medicine will enable a perfect repair of burns. Stem cells release extracellular vesicles around injured tissues to improve its structural and functional repair. But the current methods for vesicles enrichment are not efficient enough to meet the needs of investigation. Here we isolated the vesicles from the stem cells supernatants by either traditional method or ultrafiltration. We found that the vesicles isolated with ultrafiltration method displayed a similar cell proliferation ability of that with the traditional one. The output of the vesicles or its anti-aging capability has increased 18- or 3.7-times respectively. Therefore, the scalable and effective isolation method described here may facilitate the successful medical translation of the vesicles for clinical use.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Wu YC, Wu GX, Huang HH, Kuo SM. Liposome-encapsulated farnesol accelerated tissue repair in third-degree burns on a rat model. Burns 45(5), 1139–1151 (2019).
    • 2. Lewis CJ, Mardaryev AN, Poterlowicz K et al. Bone morphogenetic protein signaling suppresses wound-induced skin repair by inhibiting keratinocyte proliferation and migration. J. Invest. Dermatol. 134(3), 827–837 (2014).
    • 3. Assis-Ribas T, Forni MF, Winnischofer SMB, Sogayar MC, Trombetta-Lima M. Extracellular matrix dynamics during mesenchymal stem cells differentiation. Dev. Biol. 437(2), 63–74 (2018).
    • 4. Kariminekoo S, Movassaghpour A, Rahimzadeh A, Talebi M, Shamsasenjan K, Akbarzadeh A. Implications of mesenchymal stem cells in regenerative medicine. Artif Cells Nanomed. Biotechnol. 44(3), 749–757 (2016).
    • 5. Han YF, Chai JK, Sun TJ, Li DJ, Tao R. Differentiation of human umbilical cord mesenchymal stem cells into dermal fibroblasts in vitro. Biochem. Biophys. Res. Commun. 413(4), 561–565 (2011).
    • 6. Luo GX, Cheng WG, He WF et al. Promotion of cutaneous wound healing by local application of mesenchymal stem cells derived from human umbilical cord blood. Wound Repair Regen. 18(5), 506–513 (2010).
    • 7. Sasaki M, Abe R, Fujita Y, Ando S, Inokuma D, Shimizu H. Mesenchymal stem cells are recruited into wounded skin and contribute to wound repair by transdifferentiation into multiple skin cell type. J. Immunol. 180(4), 2581–2587 (2008).
    • 8. Wu YJ, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells 25(10), 2648–2659 (2007).
    • 9. Zou Z, Zhang Y, Hao L et al. More insight into mesenchymal stem cells and their effects inside the body. Expert Opin. Biol. Ther. 10(2), 215–230 (2010).
    • 10. Baldari S, Di Rocco G, Piccoli M, Pozzobon M, Muraca M, Toietta G. Challenges and strategies for improving the regenerative effects of mesenchymal stromal cell-based therapies. Int. J. Mol. Sci. 18(10), 2087 (2017).
    • 11. Phinney DG, Prockop DJ. Concise review: mesenchymal stem/multipotent stromal cells: the state of transdifferentiation and modes of tissue repair – current views. Stem Cells 25(11), 2896–2902 (2010).
    • 12. Baker CD, Seedorf GJ, Wisniewski BL et al. Endothelial colony-forming cell conditioned media promote angiogenesis in vitro and prevent pulmonary hypertension in experimental bronchopulmonary dysplasia. Am. J. Physiol. Lung Cell Mol. Physiol. 305(1), L73–L81 (2013).
    • 13. Bjørge I, Kim S, Mano J, Kalionis B, Chrzanowski W. Extracellular vesicles, exosomes and shedding vesicles in regenerative medicine – a new paradigm for tissue repair. Biomater Sci. 6(1), 60–78 (2017). •• Highlights the differences in composition between shedding vesicles and exosomes that depend on the various types of stem cells from which they are derived. The conditions that affect the production of exosomes in different cell types are deliberated. This review also presents the current status of candidate exosomal miRNAs for potential therapeutic use in regenerative medicine, and in applications involving widely studied organs and tissues such as heart, lung, cartilage and bone.
    • 14. Harting MT, Srivastava AK, Zhaorigetu S et al. Inflammation-stimulated mesenchymal stromal cell-derived extracellular vesicles attenuate inflammation. Stem Cells 36, 79–90 (2018). • Demonstrates a specific anti-inflammatory, mesenchymal stem cell (MSC)-extracellular vesicles (EV) mediated response and the capacity to change efficacy in response to inflammatory cues, creating the foundation for enhancing the efficacy of translational efforts using MSC-EV for targeting inflammatory injuries and diseases.
    • 15. Furi I, Momen-Heravi F, Szabo G. Extracellular vesicle isolation: present and future. Annals Translati. Med. 5(12), 263 (2017).
    • 16. Akyurekli C, Le Y, Richardson R, Fergusson D, Tay J, Allan D. A systematic review of preclinical studies on the therapeutic potential of mesenchymal stromal cell-derived microvesicles. Stem Cell Rev. Rep. 11(1), 150–160 (2015).
    • 17. Rani S, Ryan A, Griffin M, Ritter T. Mesenchymal stem cell-derived extracellular vesicles: toward cell-free therapeutic applications. Mol. Ther. 23(5), 812–823 (2015).
    • 18. Katsuda T, Kosaka N, Takeshita F, Ochiya T. The therapeutic potential of mesenchymal stem cell-derived extracellular vesicles. Proteomics 13(10–11), 1637–1653 (2013).
    • 19. Zhang B, Wang M, Gong A et al. HucMSC-exosome mediated-Wnt4 signaling is required for cutaneous wound healing. Stem Cells 33(7), 2158–2168 (2015).
    • 20. Zhang B, Shi Y, Gong A et al. HucMSC exosome-delivered 14-3-3ζ orchestrates self-control of the Wnt response via modulation of YAP during cutaneous regeneration. Stem Cells 34(10), 2485–2500 (2016).
    • 21. Sun Y, Shi H, Yin S et al. Human mesenchymal stem cell derived exosomes alleviate Type 2 diabetes mellitus by reversing peripheral insulin resistance and relieving β-cell destruction. ACS Nano. 12(8), 7613–7628 (2018).
    • 22. Li X, Liu L, Yang J et al. Exosome derived from human umbilical cord mesenchymal stem cell mediates MiR-181c attenuating burn-induced excessive inflammation. EBioMedicine 8, 72–82 (2016). • MSC-derived exosomes have diverse functions in regulating wound healing and inflammation. The miR-181c expression in human umbilical cord MSC-exosomes reduces burn-induced inflammation by down-regulating the TLR4 signaling pathway.
    • 23. Shi Q, Qian Z, Liu D et al. GMSC-derived exosomes combined with a chitosan/silk hydrogel sponge accelerates wound healing in a diabetic rat skin defect model. Front. Physiol. 8, 904 (2017).
    • 24. Gardiner C, Di Vizio D, Sahoo S et al. Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey. J. Extracell. Vesicles 5, 32945 (2016).
    • 25. Busatto S, Giacomini A, Montis C, Ronca R, Bergese P. Uptake profiles of human serum exosomes by murine and human tumor cells through combined use of colloidal nanoplasmonics and flow cytofluorimetric analysis. Anal. Chem. 90(13), 7855–7861 (2018).
    • 26. Furi I, Momen-Heravi F, Szabo G. Extracellular vesicle isolation: present and future. Ann. Transl. Med. 5(12), 263–263 (2017).
    • 27. Gámez-Valero A, Monguió-Tortajada M, Carreras-Planella L, Franquesa M, Beyer K, Borràs F. Size-exclusion chromatography-based isolation minimally alters extracellular vesicles' characteristics compared to precipitating agents. Sci. Rep. 6(33641), 33641 (2016).
    • 28. Reiner A, Witwer K, Van Balkom B et al. Concise review: developing best-practice models for the therapeutic use of extracellular vesicles. Stem Cells Transl. Med. 6(8), 1730–1739 (2017).
    • 29. Colao I, Corteling R, Bracewell D, Wall I. Manufacturing exosomes: a promising therapeutic platform. Trends Mol. Med. 24(3), 242–256 (2018).
    • 30. Jong AY, Wu CH, Li J, Sun J, Seeger RC. Large-scale isolation and cytotoxicity of extracellular vesicles derived from activated human natural killer cells. J. Extracell. Vesicles 6(1), 1294368 (2017).
    • 31. Busatto S, Vilanilam G, Ticer T et al. Tangential flow filtration for highly efficient concentration of extracellular vesicles from large volumes of fluid. Cells 7(12), 273 (2018). •• Comparative assessment of tangential flow filtration (TFF) and ultracentrifugation (UC) of conditioned cell culture media revealed that the former concentrates EVs of comparable physicochemical characteristics, but with higher yield, less single macromolecules and aggregates (<15nm in size), and improved batch-to-batch consistency in half the processing time (1 h). TFF is a more efficient, scalable, and gentler method.
    • 32. Andriolo G, Provasi E, Lo Cicero V et al. Exosomes from human cardiac progenitor cells for therapeutic applications: development of a GMP-grade manufacturing method. Front. Physiol. 9, 1169 (2018). •• A good manufacturing process (GMP)-compliant manufacturing method was set up, based on large-scale cell culture in xeno-free conditions, collection of up to 8 L of exosome-containing conditioned medium and isolation of Exo-cardiac progenitor cell through TFF. The GMP manufacturing method guaranteed high exosome yield (>1013 particles) and consistent removal (≥97%) of contaminating proteins.
    • 33. Wu H, Zeng X, Yu J et al. Comparison of nucleus pulposus stem/progenitor cells isolated from degenerated intervertebral discs with umbilical cord derived mesenchymal stem cells. Exp. Cell Res. 361(2), 324–332 (2017).
    • 34. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J. Cell Biol. 200(4), 373–383 (2013).
    • 35. Candelario KM, Steindler DA. The role of extracellular vesicles in the progression of neurodegenerative disease and cancer. Trends Mol. Med. 20(7), 368–374 (2014).
    • 36. Vader P, Breakefield X, Wood MJA. Extracellular vesicles: emerging targets for cancer therapy. Trends Mol. Med. 20(7), 385–393 (2014).
    • 37. Borrelli D, Yankson K, Shukla N, Vilanilam G, Ticer T, Wolfram J. Extracellular vesicle therapeutics for liver disease. J. Control. Rel. S0168365918300385 (2018).
    • 38. Paolini L, Zendrini A, Radeghieri A. Biophysical properties of extracellular vesicles in diagnostics. Biomark. Med. 12(4), 383–391 (2018).
    • 39. Conlan RS, Pisano S, Oliveira MI, Ferrari M, Pinto IM. Exosomes as reconfigurable therapeutic systems. Trends Mol. Med. 23(7), 636 (2017).
    • 40. Gilligan KE, Dwyer RM. Engineering exosomes for cancer therapy. Int. J. Mol. Sci. 18(6), 1122- (2017).
    • 41. Jiang L, Vader P, Schiffelers RM. Extracellular vesicles for nucleic acid delivery: progress and prospects for safe RNA-based gene therapy. Gene Ther. 24(3), 157 (2017).
    • 42. Kamerkar S, Lebleu V, Sugimoto H et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature 546(7659), 498–503 (2017).
    • 43. Wen S, Dooner M, Cheng Y et al. Mesenchymal stromal cell-derived extracellular vesicles rescue radiation damage to murine marrow hematopoietic cells. Leukemia 30(11), 2221–2231 (2016).
    • 44. Didiot M, Hall L, Coles A et al. Exosome-mediated delivery of hydrophobically modified siRNA for Huntingtin mRNA silencing. Mol. Ther. 24(10), 1836–1847 (2016).
    • 45. Pi F, Binzel D, Lee T et al. Nanoparticle orientation to control RNA loading and ligand display on extracellular vesicles for cancer regression. Nat. Nanotechnol. 13(1), 82–89 (2018).
    • 46. Gardiner C, Vizio DD, Sahoo S et al. Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey. J. Extracell. Vesicles 5(1), 32945 (2016).
    • 47. Haraszti R, 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. 26(12), 2838–2847 (2018). •• In combination with the conventional differential ultracentrifugation, 3D culture yields 20-fold more exosomes (3D-UC-exosomes) than two-dimensional cultures (2D-UC-exosomes). TFF in combination with 3D MSC cultures further improves the yield of exosomes (3D-TFF-exosomes) 7-fold over 3D-UC-exosomes. These findings lift a major roadblock for the clinical utility of MSC exosomes.
    • 48. Potter M, Lins B, Mietzsch M et al. A simplified purification protocol for recombinant adeno-associated virus vectors. Mol. Ther. Methods Clin. Dev. 1, 14034 (2014). • A simple and rapid clarification of cell lysate to remove the bulk of proteins and DNA is accomplished by sodium citrate and citric acid. After the low-speed centrifugation step, the supernatant is subjected to cation exchange chromatography via sulfopropyl column. The eluted virus may then be further concentrated by either centrifugal spin devices or tangential flow filtration yielding material of high titer and GMP grade biochemical purity.
    • 49. Li H, Fan X, Wang Y et al. Extracellular vesicles from human urine-derived stem cells ameliorate particulate polyethylene-induced osteolysis. Internat. J. Nanomed. 16, 7479–7494 (2021).
    • 50. Li SL, Liu J, Liu SY, Jiao WJ, Wang XH. Chitosan oligosaccharides packaged into rat adipose mesenchymal stem cells-derived extracellular vesicles facilitating cartilage injury repair and alleviating osteoarthritis. J. Nanobiotechnol. 19(1), 343 (2021).
    • 51. Xiao X, Xu MQ, Yu HL et al. Mesenchymal stem cell-derived small extracellular vesicles mitigate oxidative stress-induced senescence in endothelial cells via regulation of miR-146a/Src. Signal Transduction Targeted Ther. 6(1), 354 (2021).
    • 52. Bissett D, Chatterjee R, Hannon D. Photoprotective effect of superoxide-scavenging antioxidants against ultraviolet radiation-induced chronic skin damage in the hairless mouse. Photodermatol. Photoimmunol. Photomed. 7(2), 56–62 (1990).
    • 53. Liang C, Ju W, Pei S, Tang Y, Xiao Y. Pharmacological activities and synthesis of esculetin and its derivatives: a mini-review. Molecules 22(3), 387 (2017).
    • 54. Lai R, Arslan F, Lee M et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. Ther. 4(3), 214–222 (2010).
    • 55. Lai R, Yeo R, Tan K, Lim S. Mesenchymal stem cell exosome ameliorates reperfusion injury through proteomic complementation. Regen. Med. 8(2), 197–209 (2013).
    • 56. Chen L, Xu Y, Zhao J et al. Conditioned medium from hypoxic bone marrow-derived mesenchymal stem cells enhances wound healing in mice. PLoS ONE 9(4), e96161 (2014).
    • 57. Jun E, Zhang Q, Yoon B et al. Hypoxic conditioned medium from human amniotic fluid-derived mesenchymal stem cells accelerates skin wound healing through TGF-β/SMAD2 and PI3K/Akt pathways. Int. J. Mol. Sci. 15(1), 605–628 (2014).
    • 58. Liang X, Zhang L, Wang S, Han Q, Zhao RC. Exosomes secreted by mesenchymal stem cells promote endothelial cell angiogenesis by transferring miR-125a. J. Cell Sci. 129(11), 2182–2189 (2016).
    • 59. Maguire G, Friedman P, Mccarthy D, Friedman R, Maniotis A. Stem cell released molecules and exosomes in tissue engineering. Procedia Engin. 59, 270–278 (2013).
    • 60. Kim YJ, Yoo SM, Park HH et al. Exosomes derived from human umbilical cord blood mesenchymal stem cells stimulates rejuvenation of human skin. Biochem. Biophys. Res. Commun. 493(2), 1102-–1108 (2017).
    • 61. Nazihah B, Jeschke MG, Elaine H, Mohammadali S, Saeid AN. Exosomes from acellular Wharton's jelly of the human umbilical cord promotes skin wound healing. Stem Cell Res. Ther. 9(1), 193 (2018). •• Exosomes are being enriched in the native niche of the umbilical cord and can enhance wound healing in vivo through their cargo. Exosomes from the acellular gelatinous Wharton's jelly and the cargo protein α2M have tremendous potential as a noncellular, off-the-shelf therapeutic modality for wound healing.
    • 62. He L, Zhu C, Jia J, Hao XY, Shu MG. ADSC-Exos containing MALAT1 promotes wound healing by targeting miR-124 through activating Wnt/β-catenin pathway. Biosci. Rep. 40(5), 2549 (2020).
    • 63. Calò R, Marabini L. Protective effect of Vaccinium myrtillus extract against UVA- and UVB-induced damage in a human keratinocyte cell line (HaCaT cells) – ScienceDirect. J. Photoch. PhotoBio B. 132(2), 27–35 (2014).
    • 64. Choi J, Cho W, Choi Y et al. Functional recovery in photo-damaged human dermal fibroblasts by human adipose-derived stem cell extracellular vesicles. J. Extracell. Vesicles 8(1), 1565885 (2019). • Investigated the effects of EVs derived from human adipose-derived stem cells (HASCs) on photo-damaged human dermal fibroblasts. It suggested that diverse components that are enriched in HASC-derived EVs could act as a biochemical cue for recovery from skin photoaging.
    • 65. Bielefeld K, Amini-Nik S, Alman B. Cutaneous wound healing: recruiting developmental pathways for regeneration. Cellul. Mol. Life Sci. 70(12), 2059–2081 (2013).
    • 66. Ma T, Fu B, Yang X, Xiao Y, Pan M. Adipose mesenchymal stem cell-derived exosomes promote cell proliferation, migration, and inhibit cell apoptosis via Wnt/β-catenin signaling in cutaneous wound healing. J. Cell. Biochem. 120(6), 10847–10854 (2019).
    • 67. Zhang J, Guan J, Niu X et al. Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. J. Transl. Med. 13, 49 (2015).
    • 68. Zhao G, Liu F, Liu Z et al. MSC-derived exosomes attenuate cell death through suppressing AIF nucleus translocation and enhance cutaneous wound healing. Stem Cell Res. Ther. 11(1), 174 (2020).