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

Exosomes: the latest in regenerative aesthetics

    Krishna S Vyas

    Division of Plastic Surgery, Mayo Clinic, Rochester, MN 55905, USA

    ,
    Joely Kaufman

    Skin Associates of South Florida & Skin Research Institute, Coral Gables, FL 33146, USA

    ,
    Girish S Munavalli

    Dermatology, Laser, & Vein Specialists of the Carolinas, Charlotte, NC 28207, USA

    ,
    Kiran Robertson

    Private Practice, Dallas, TX 75205, USA

    ,
    Atta Behfar

    Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA

    &
    Saranya P Wyles

    *Author for correspondence:

    E-mail Address: wyles.saranya@mayo.edu

    Department of Dermatology, Mayo Clinic, Rochester, MN 55905, USA

    Published Online:https://doi.org/10.2217/rme-2022-0134

    Regenerative aesthetics is a burgeoning field for skin rejuvenation and skin health restoration. Exosomes, or extracellular vesicles, represent a new and minimally invasive addition to the regenerative aesthetic toolbox. These nano-sized vesicles contain bioactive cargo with crucial roles in intercellular communication. Exosome technology, while still in its infancy, is now leveraged in regenerative aesthetic medicine due to its multifaceted role in targeting root causes of skin aging and improving overall tissue homeostasis. The main considerations for practice utilization include variation in exosome purification, isolation, storage, scalability and reproducibility. This review aims at highlighting the current and emerging landscape of exosomes in aesthetic medicine including skin rejuvenation and hair restoration.

    Plain language summary

    What is this article about?

    The purpose of this paper is to review available studies that look at the effects of exosomes in aesthetic medicine and cosmetic surgery. A thorough literature search of all available studies was performed.

    What were the results?

    Topical exosomes, although variable in source and method of isolation, are generally considered safe in humans on intact skin. The current published research literature does not yet provide a clear consensus on long-term use for skin rejuvenation or hair restoration, nor does it delineate which patients would benefit most from this technology. There are no currently US FDA-approved exosome products on the market for medical indications.

    What do the results of this literature review mean?

    More clinical studies with proper regulatory oversight are needed.

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

    References

    • 1. Zarbafian M, Fabi SG, Dayan S, Goldie K. The emerging field of regenerative aesthetics: where we are now. Dermatol. Surg. 48(1), 101–108 (2022).
    • 2. Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell 153(6), 1194–1217 (2013). • A highly cited review enumerating the tentative hallmarks of aging.
    • 3. Marks PW, Witten CM, Califf RM. Clarifying stem-cell therapy's benefits and risks. N. Engl. J. Med. 376(11), 1007–1009 (2017).
    • 4. Kee LT, Ng CY, Al-Masawa ME et al. Extracellular Vesicles in Facial Aesthetics: A Review. Int. J. Mol. Sci. 23(12), 6742 (2022 Jun 16).
    • 5. Harding C, Heuser J, Stahl P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J. Cell Biol. 97(2), 329–339 (1983).
    • 6. Than UTT, Leavesley DI, Parker TJ. Characteristics and roles of extracellular vesicles released by epidermal keratinocytes. J. Eur. Acad. Dermatol. Venereol. 33(12), 2264–2272 (2019).
    • 7. Kalluri R. The biology and function of exosomes in cancer. J. Clin. Invest. 126(4), 1208–1215 (2016). • A highly cited review with illustrative figures enumerating the role of exosomes.
    • 8. Jella KK, Nasti TH, Li Z, Malla SR, Buchwald ZS, Khan MK. Exosomes, Their Biogenesis and Role in Inter-Cellular Communication, Tumor Microenvironment and Cancer Immunotherapy. Vaccines (Basel). 6(4), 69 (2018).
    • 9. Spada S, Rudqvist NP, Wennerberg E. Isolation of DNA from exosomes. Methods Enzymol. 636, 173–183 (2020).
    • 10. Spada S. Methods to purify DNA from extracellular vesicles: focus on exosomes. Methods Enzymol. 645, 109–118 (2020).
    • 11. Li L, Ngo HTT, Hwang E et al. Conditioned medium from human adipose-derived mesenchymal stem cell culture prevents UVB-induced skin aging in human keratinocytes and dermal fibroblasts. Int. J. Mol. Sci. 21(1), 49 (2019).
    • 12. Tang Y, Zhou Y, Li HJ. Advances in mesenchymal stem cell exosomes: a review. Stem Cell Res Ther 12(1), 71 (2021).
    • 13. Villatoro AJ, Alcoholado C, Martin-Astorga MC, Fernandez V, Cifuentes M, Becerra J. Comparative analysis and characterization of soluble factors and exosomes from cultured adipose tissue and bone marrow mesenchymal stem cells in canine species. Vet. Immunol. Immunopathol. 208, 6–15 (2019).
    • 14. Van Balkom BWM, Gremmels H, Giebel B, Lim SK. Proteomic Signature of Mesenchymal Stromal Cell-Derived Small Extracellular Vesicles. Proteomics 19(1-2), e1800163 (2019).
    • 15. Proffer S, Paradise CR, Degrazia E et al. Efficacy and tolerability of topical platelet exosomes for skin rejuvenation: six-week results. Aesthet Surg. J. doi: 10.1093/asj/sjac149 (2022).
    • 16. Sanchez-Gonzalez DJ, Mendez-Bolaina E, Trejo-Bahena NI. Platelet-rich plasma peptides: key for regeneration. Int. J. Pept. 2012, 532519 (2012).
    • 17. Langevin SM, Kuhnell D, Orr-Asman MA et al. Balancing yield, purity and practicality: a modified differential ultracentrifugation protocol for efficient isolation of small extracellular vesicles from human serum. RNA Biol 16(1), 5–12 (2019).
    • 18. Coughlan C, Bruce KD, Burgy O et al. Exosome isolation by ultracentrifugation and precipitation and techniques for downstream analyses. Curr. Protoc. Cell Biol. 88(1), e110 (2020).
    • 19. Liangsupree T, Multia E, Riekkola ML. Modern isolation and separation techniques for extracellular vesicles. J. Chromatogr. A 1636, 461773 (2021).
    • 20. Li K, Wong D, Hong K, Raffai R. Cushioned-Density Gradient Ultracentrifugation (C-DGUC): a refined and high performance method for the isolation, characterization, and use of exosomes. Methods Mol. Biol. 1740, 69–83 (2018).
    • 21. Boing AN, Van Der Pol E, Grootemaat AE, Coumans FA, Sturk A, Nieuwland R. Single-step isolation of extracellular vesicles by size-exclusion chromatography. J. Extracell. Vesicles 8, 3 (2014).
    • 22. Niu Z, Pang RTK, Liu W et al. Polymer-based precipitation preserves biological activities of extracellular vesicles from an endometrial cell line. PLOS ONE. 12(10), e0186534 (2017).
    • 23. Greening D, Xu R, Ji H. A protocol for exosome isolation and characterization: evaluation of ultracentrifugation, density-gradient separation, and immunoaffinity capture methods. Methods Mol. Biol. 1295, 179–209 (2015).
    • 24. Davies R, Kim J, Jang S, Choi E, Gho Y, Park J. Microfluidic filtration system to isolate extracellular vesicles from blood. Lab Chip. 12, 5202–5210 (2012).
    • 25. Wang Z, Wu H, Fine D, Schmulen J, Hu Y, Godin B et al. Ciliated micropillars for the microfluidic-based isolation of nanoscale lipid vesicles. Lab Chip. 13, 2879–2882 (2013).
    • 26. Hong P, Yang H, Wu Y, Li K, Tang Z. The functions and clinical application potential of exosomes derived from adipose mesenchymal stem cells: a comprehensive review. Stem Cell Res. Ther. 10(1), 242 (2019).
    • 27. Nordin J, Lee Y, Vader P et al. Ultrafiltration with size-exclusion liquid chromatography for high yield isolation of extracellular vesicles preserving intact biophysical and functional properties. Nanomedicine 11, 879–883 (2015).
    • 28. Xu P, Xin Y, Zhang Z et al. Extracellular vesicles from adipose-derived stem cells ameliorate ultraviolet B-induced skin photoaging by attenuating reactive oxygen species production and inflammation. Stem Cell Res. Ther. 11, 264 (2020).
    • 29. Geeurickx E, Tulkens J, Dhondt B et al. The generation and use of recombinant extracellular vesicles as biological reference material. Nat. Commun 10(1), 3288 (2019).
    • 30. Multia E, Liangsupree T, Jussila M, Ruiz-Jimenez J, Kemell M, Riekkola ML. Automated on-line isolation and fractionation system for nanosized biomacromolecules from human plasma. Anal. Chem. 92(19), 13058–13065 (2020).
    • 31. Fang X, Duan Y, Adkins GB et al. Highly efficient exosome isolation and protein analysis by an integrated nanomaterial-based platform. Anal. Chem. 90(4), 2787–2795 (2018).
    • 32. 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. 27(5), 796–810 (2010).
    • 33. Jung MK, Mun JY. Sample preparation and imaging of exosomes by transmission electron microscopy. J. Visualized Exp. 131, 1–5 (2018).
    • 34. Dragovic RA, Gardiner C, Brooks AS et al. Sizing and phenotyping of cellular vesicles using nanoparticle tracking analysis. Nanomedicine: Nanotechnol. Biol. Med. 7, 780–788 (2011).
    • 35. Pecora R. Dynamic light scattering measurement of nanometer particles in liquids. J. Nanoparticle Res. 2, 123–131 (2000).
    • 36. Maas SL, De Vrij J, Broekman ML. Quantification and size-profiling of extracellular vesicles using tunable resistive pulse sensing. J. Visualized Exp. 92, 1–7 (2014).
    • 37. Mincheva Nilsson L, Baranov V, Nagaeva O, Dehlin E. Isolation and Characterization of Exosomes from Cultures of Tissue Explants and Cell Lines. Curr. Protoc. Immunol. 115, 14.42.1–14.42.21 (2016).
    • 38. Libregts SFWM, Arkesteijn GJA, Németh A, Noltet Hoen ENM, Wauben MHM. Flow cytometric analysis of extracellular vesicle subsets in plasma: impact of swarm by particles of non-interest. J. Thromb. Haemostasis. 16, 1423–1436 (2020).
    • 39. Kormelink TG, Arkesteijn GJ, Nauwelaers FA, van den Engh G, Nolte-'t Hoen EN, Wauben MH. Prerequisites for the analysis and sorting of extracellular vesicle subpopulations by high-resolution flow cytometry. Cytometry A 89A, 135–147 51 (2016).
    • 40. Arraud N, Gounou C, Turpin D, Brisson AR. Fluorescence triggering: a general strategy for enumerating and phenotyping extracellular vesicles by flow cytometry. Cytometry A 89A, 184–195 (2016).
    • 41. Zouboulis CC, Ganceviciene R, Liakou AI, Theodoridis A, Elewa R, Makrantonaki E. Aesthetic aspects of skin aging, prevention, and local treatment. Clin. Dermatol. 37(4), 365–372 (2019).
    • 42. Waldera Lupa DM, Kalfalah F, Safferling K et al. Characterization of Skin Aging-Associated Secreted Proteins (SAASP) Produced by Dermal Fibroblasts Isolated from Intrinsically Aged Human Skin. J. Invest. Dermatol. 135(8), 1954–1968 (2015).
    • 43. Yoo JK, Kim CH, Jung HY, Lee DR, Kim JK. Discovery and characterization of miRNA during cellular senescence in bone marrow-derived human mesenchymal stem cells. Exp. Gerontol. 58, 139–145 (2014).
    • 44. Wang T, Jian Z, Baskys A et al. MSC-derived exosomes protect against oxidative stress-induced skin injury via adaptive regulation of the NRF2 defense system. Biomaterials 257, 120264 (2020).
    • 45. Li X, Xie X, Lian W et al. Exosomes from adipose-derived stem cells overexpressing NRF2 accelerate cutaneous wound healing by promoting vascularization in a diabetic foot ulcer rat model. Exp. Mol. Med. 50(4), 1–14 (2018).
    • 46. Carrasco E, Soto-Heredero G, Mittelbrunn M. The role of extracellular vesicles in cutaneous remodeling and hair follicle dynamics. Int. J. Mol. Sci 20(11), 23430 (2019).
    • 47. Zhang K, Yu L, Li FR et al. Topical application of exosomes derived from human umbilical cord mesenchymal stem cells in combination with sponge spicules for treatment of photoaging. Int. J. Nanomedicine 15, 2859–2872 (2020).
    • 48. 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).
    • 49. Liu SJ, Meng MY, Han S et al. Umbilical cord mesenchymal stem cell-derived exosomes ameliorate HaCaT cell photo-aging. Rejuvenation Res. 24(4), 283–293 (2021).
    • 50. Choi JS, Cho WL, Choi YJ 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).
    • 51. Gao W, Wang X, Si Y et al. Exosome derived from ADSCs attenuates ultraviolet B-mediated photoaging in human dermal fibroblasts. Photochem. Photobiol 97(4), 795–804 (2021).
    • 52. Liang JX, Liao X, Li SH et al. Antiaging properties of exosomes from adipose-derived mesenchymal stem cells in photoaged rat skin. Biomed. Res. Int. 2020, 6406395 (2020).
    • 53. Kwon HH, Yang SH, Lee J et al. Combination treatment with human adipose tissue stem cell-derived exosomes and fractional CO2 laser for acne scars: a 12-week prospective, double-blind, randomized, split-face study. Acta Derm Venereol 100(18), adv00310 (2020).
    • 54. Oh M, Lee J, Kim YJ, Rhee WJ, Park JH. Exosomes derived from human induced pluripotent stem cells ameliorate the aging of skin fibroblasts. Int. J. Mol. Sci. 19(6), 1715 (2018 Jun 9).
    • 55. Deng M, Yu TZ, Li D et al. Human umbilical cord mesenchymal stem cell-derived and dermal fibroblast-derived extracellular vesicles protect dermal fibroblasts from ultraviolet radiation-induced photoaging in vitro. Photochem Photobiol Sci 19(3), 406–414 (2020).
    • 56. Wu P, Zhang B, Han X et al. HucMSC exosome-delivered 14-3-3ζ alleviates ultraviolet radiation-induced photodamage via SIRT1 pathway modulation. Aging (Albany NY). 13(8), 11542–11563 (2021).
    • 57. Hu S, Li Z, Cores J et al. Needle-free injection of exosomes derived from human dermal fibroblast spheroids ameliorates skin photoaging. ACS Nano 13(10), 11273–11282 (2019).
    • 58. Fusenig NE, Limat A, Stark HJ, Breitkreutz D. Modulation of the differentiated phenotype of keratinocytes of the hair follicle and from epidermis. J. Dermatol. Sci. 7(Suppl. S142-151), S142–S151 (1994).
    • 59. Roh C, Tao Q, Lyle S. Dermal papilla-induced hair differentiation of adult epithelial stem cells from human skin. Physiol. Genomics 19(2), 207–217 (2004).
    • 60. Kishimoto J, Burgeson RE, Morgan BA. Wnt signaling maintains the hair-inducing activity of the dermal papilla. Genes Dev. 14(10), 1181–1185 (2000).
    • 61. Kwack MH, Seo CH, Gangadaran P et al. Exosomes derived from human dermal papilla cells promote hair growth in cultured human hair follicles and augment the hair-inductive capacity of cultured dermal papilla spheres. Exp. Dermatol. 28(7), 854–857 (2019).
    • 62. Yan H, Gao Y, Ding Q et al. Exosomal micro RNAs derived from dermal papilla cells mediate hair follicle stem cell proliferation and differentiation. Int. J. Biol. Sci. 15(7), 1368–1382 (2019).
    • 63. Rajendran RL, Gangadaran P, Bak SS et al. Extracellular vesicles derived from MSCs activates dermal papilla cell in vitro and promotes hair follicle conversion from telogen to anagen in mice. Sci. Rep. 7(1), 15560 (2017).
    • 64. Ha DH, Kim HK, Lee J et al. Mesenchymal stem/stromal cell-derived exosomes for immunomodulatory therapeutics and skin regeneration. Cells 9(5), 1157 (2020).
    • 65. Zhou L, Wang H, Jing J, Yu L, Wu X, Lu Z. Regulation of hair follicle development by exosomes derived from dermal papilla cells. Biochem. Biophys. Res. Commun. 500(2), 325–332 (2018).
    • 66. Kim H, Jang Y, Kim EH et al. Potential of colostrum-derived exosomes for promoting hair regeneration through the transition from telogen to anagen phase. Front. Cell Dev. Biol. 10, 815205 (2022).
    • 67. Li MY, Liu DW, Mao YG. [Advances in the research of effects of exosomes derived from stem cells on wound repair]. Zhonghua Shao Shang Za Zhi 33(3), 180–184 (2017).
    • 68. Li Y, Wang G, Wang Q, Zhang Y, Cui L, Huang X. Exosomes secreted from adipose-derived stem cells are a potential treatment agent for immune-mediated alopecia. J. Immunol. Res 2022, 7471246 (2022).
    • 69. Liu Y, Xue L, Gao H et al. Exosomal miRNA derived from keratinocytes regulates pigmentation in melanocytes. J. Dermatol. Sci. 93(3), 159–167 (2019).
    • 70. Wang XY, Guan XH, Yu ZP et al. Human amniotic stem cells-derived exosmal miR-181a-5p and miR-199a inhibit melanogenesis and promote melanosome degradation in skin hyperpigmentation, respectively. Stem Cell Res. Ther. 12(1), 501 (2021).
    • 71. Wäster P, Eriksson I, Vainikka L, Öllinger K. Extracellular vesicles released by melanocytes after UVA irradiation promote intercellular signaling via miR21. Pigment Cell Melanoma Res. 33(4), 542–555 (2020).
    • 72. Thery C, Witwer KW, Aikawa E et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 7(1), 1535750 (2018). •• The MISEV2018 guidelines include tables and summaries of suggested protocols and steps to document specific EV-associated functional activities.
    • 73. Hahm J, Kim J, Park J. Strategies to enhance extracellular vesicle production. Tiss. Engin. Regen. Med. 18(4), 513–524 (2021).
    • 74. Charoenviriyakul C, Takahashi Y, Nishikawa M, Takakura Y. Preservation of exosomes at room temperature using lyophilization. Int. J. Pharm. 553(1-2), 1–7 (2018).
    • 75. Charoenviriyakul C, Takahashi Y, Nishikawa M, Takakura Y. Erratum to ‘Preservation of exosomes at room temperature using lyophilization’ [International Journal of Pharmaceutics 553 (2018) 1–7]. Int J Pharm. 559, 427–428 (2019).
    • 76. Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat. Nanotechnol. 16(7), 748–759 (2021).