Combination of Biomaterials and Extracellular Vesicles from Mesenchymal Stem-Cells: New Therapeutic Strategies for Skin-Wound Healing
Abstract
:1. Introduction
2. Physiology and Pathology of Cutaneous-Wound Healing
3. Mesenchymal Stem-Cells in Wound Healing
4. MSC-Derived Extracellular Vesicles
5. MSC-EV and Wound Healing
5.1. Hemostasis
5.2. Inflammation
5.3. Proliferation
5.4. Remodeling
6. Extracellular-Vesicle Pharmacokinetics
7. Use of Biomaterials in the Release of Extracellular Vesicles for Treatment of Cutaneous Wounds
7.1. Chitosan Hydrogels
7.2. Alginate Hydrogels
7.3. Hyaluronic-Acid Hydrogels
7.4. Collagen Hydrogels
7.5. Pluronic F127 Hydrogels
7.6. Decellularized Biomaterials
7.7. Complex Multifunctional Scaffolds
8. Conclusions and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Takeo, M.; Lee, W.; Ito, M. Wound healing and skin regeneration. Cold Spring Harb. Perspect. Med. 2015, 5, a023267. [Google Scholar] [CrossRef]
- Gantwerker, E.A.; Hom, D.B. Skin: Histology and physiology of wound healing. Facial Plast. Surg. Clin. N. Am. 2011, 19, 441–453. [Google Scholar] [CrossRef] [PubMed]
- Rodrigues, M.; Kosaric, N.; Bonham, C.A.; Gurtner, G.C. Wound healing: A cellular perspective. Physiol. Rev. 2019, 99, 665–706. [Google Scholar] [CrossRef] [PubMed]
- Beyene, R.T.; Derryberry, S.L.; Barbul, A. The Effect of Comorbidities on Wound Healing. Surg. Clin. N. Am. 2020, 100, 695–705. [Google Scholar] [CrossRef] [PubMed]
- Velnar, T.; Bailey, T.; Smrkolj, V. The wound healing process: An overview of the cellular and molecular mechanisms. J. Int. Med. Res. 2009, 37, 1528–1542. [Google Scholar] [CrossRef]
- Eming, S.A.; Martin, P.; Tomic-Canic, M. Wound repair and regeneration: Mechanisms, signaling, and translation. Sci. Transl. Med. 2014, 6, 265sr6. [Google Scholar] [CrossRef] [Green Version]
- Wild, S.; Roglic, G.; Green, A.; Sicree, R.; King, H. Global Prevalence of Diabetes: Estimates for the year 2000 and projections for 2030. Diabetes Care 2004, 27, 1047–1053. [Google Scholar] [CrossRef] [Green Version]
- Glovaci, D.; Fan, W.; Wong, N.D. Epidemiology of Diabetes Mellitus and Cardiovascular Disease. Curr. Cardiol. Rep. 2019, 21, 21. [Google Scholar] [CrossRef]
- Sen, C.K. Human Wounds and Its Burden: An Updated Compendium of Estimates. Adv. Wound Care 2019, 8, 39–48. [Google Scholar] [CrossRef] [Green Version]
- Raghav, A.; Khan, Z.A.; Labala, R.K.; Ahmad, J.; Noor, S.; Mishra, B.K. Financial burden of diabetic foot ulcers to world: A progressive topic to discuss always. Ther. Adv. Endocrinol. Metab. 2018, 9, 29–31. [Google Scholar] [CrossRef]
- Frykberg, R.G.; Banks, J. Challenges in the Treatment of Chronic Wounds. Adv. Wound Care 2015, 4, 560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guillamat-Prats, R. The role of MSC in wound healing, scarring and regeneration. Cells 2021, 10, 1729. [Google Scholar] [CrossRef]
- Kucharzewski, M.; Rojczyk, E.; Wilemska-Kucharzewska, K.; Wilk, R.; Hudecki, J.; Los, M.J. Novel trends in application of stem cells in skin wound healing. Eur. J. Pharmacol. 2019, 843, 307–315. [Google Scholar] [CrossRef] [PubMed]
- Walter, M.N.M.; Wright, K.T.; Fuller, H.R.; MacNeil, S.; Johnson, W.E.B. Mesenchymal stem cell-conditioned medium accelerates skin wound healing: An in vitro study of fibroblast and keratinocyte scratch assays. Exp. Cell Res. 2010, 316, 1271–1281. [Google Scholar] [CrossRef]
- Yew, T.-L.; Hung, Y.-T.; Li, H.-Y.; Chen, H.-W.; Chen, L.-L.; Tsai, K.-S.; Chiou, S.-H.; Chao, K.-C.; Huang, T.-F.; Chen, H.-L.; et al. Enhancement of wound healing by human multipotent stromal cell conditioned medium: The paracrine factors and p38 MAPK activation. Cell Transplant. 2011, 20, 693–706. [Google Scholar] [CrossRef]
- Abels, E.R.; Breakefield, X.O. Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cell. Mol. Neurobiol. 2016, 36, 301–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Casado-Díaz, A.; Quesada-Gómez, J.M.; Dorado, G. Extracellular Vesicles Derived from Mesenchymal Stem Cells (MSC) in Regenerative Medicine: Applications in Skin Wound Healing. Front. Bioeng. Biotechnol. 2020, 8, e00146. [Google Scholar] [CrossRef] [Green Version]
- Safari, B.; Aghazadeh, M.; Davaran, S.; Roshangar, L. Exosome-loaded hydrogels: A new cell-free therapeutic approach for skin regeneration. Eur. J. Pharm. Biopharm. 2022, 171, 50–59. [Google Scholar] [CrossRef] [PubMed]
- Karppinen, S.M.; Heljasvaara, R.; Gullberg, D.; Tasanen, K.; Pihlajaniemi, T. Toward understanding scarless skin wound healing and pathological scarring. F1000Research 2019, 8, 787. [Google Scholar] [CrossRef] [Green Version]
- Potekaev, N.N.; Borzykh, O.B.; Medvedev, G.V.; Pushkin, D.V.; Petrova, M.M.; Petrov, A.V.; Dmitrenko, D.V.; Karpova, E.I.; Demina, O.M.; Shnayder, N.A. The Role of Extracellular Matrix in Skin Wound Healing. J. Clin. Med. 2021, 10, 5947. [Google Scholar] [CrossRef]
- Reinke, J.M.; Sorg, H. Wound Repair and Regeneration. Eur. Surg. Res. 2012, 49, 35–43. [Google Scholar] [CrossRef] [PubMed]
- Curaj, A.; Schumacher, D.; Rusu, M.; Staudt, M.; Li, X.; Simsekyilmaz, S.; Jankowski, V.; Jankowski, J.; Dumitraşcu, A.R.; Hausenloy, D.J.; et al. Neutrophils Modulate Fibroblast Function and Promote Healing and Scar Formation after Murine Myocardial Infarction †. Int. J. Mol. Sci. 2020, 21, 3685. [Google Scholar] [CrossRef]
- Ha, D.H.; Kim, H.K.; Lee, J.; Kwon, H.H.; Park, G.H.; Yang, S.H.; Jung, J.Y.; Choi, H.; Lee, J.H.; Sung, S.; et al. Mesenchymal Stem/Stromal Cell-Derived Exosomes for Immunomodulatory Therapeutics and Skin Regeneration. Cells 2020, 9, 1157. [Google Scholar] [CrossRef]
- Gurtner, G.C.; Werner, S.; Barrandon, Y.; Longaker, M.T. Wound repair and regeneration. Nature 2008, 453, 314–321. [Google Scholar] [CrossRef]
- Pastar, I.; Stojadinovic, O.; Yin, N.C.; Ramirez, H.; Nusbaum, A.G.; Sawaya, A.; Patel, S.B.; Khalid, L.; Isseroff, R.R.; Tomic-Canic, M. Epithelialization in Wound Healing: A Comprehensive Review. Adv. Wound Care 2014, 3, 445–464. [Google Scholar] [CrossRef] [Green Version]
- Oliveira, G.V.; Hawkins, H.K.; Chinkes, D.; Burke, A.; Tavares, A.L.P.; Ramos-E-Silva, M.; Albrecht, T.B.; Kitten, G.T.; Herndon, D.N. Hypertrophic versus non hypertrophic scars compared by immunohistochemistry and laser confocal microscopy: Type I and III collagens†. Int. Wound J. 2009, 6, 445–452. [Google Scholar] [CrossRef]
- van den Broek, L.J.; Limandjaja, G.C.; Niessen, F.B.; Gibbs, S. Human hypertrophic and keloid scar models: Principles, limitations and future challenges from a tissue engineering perspective. Exp. Dermatol. 2014, 23, 382–386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, R.; Liang, H.; Clarke, E.; Jackson, C.; Xue, M. Inflammation in Chronic Wounds. Int. J. Mol. Sci. 2016, 17, 2085. [Google Scholar] [CrossRef] [PubMed]
- Eming, S.A.; Wynn, T.A.; Martin, P. Inflammation and metabolism in tissue repair and regeneration. Science 2017, 356, 1026–1030. [Google Scholar] [CrossRef] [Green Version]
- Wilgus, T.A. Inflammation as an orchestrator of cutaneous scar formation: A review of the literature. Plast. Aesthetic Res. 2020, 7, 54. [Google Scholar] [CrossRef]
- Eming, S.A.; Koch, M.; Krieger, A.; Brachvogel, B.; Kreft, S.; Bruckner-Tuderman, L.; Krieg, T.; Shannon, J.D.; Fox, J.W. Differential proteomic analysis distinguishes tissue repair biomarker signatures in wound exudates obtained from normal healing and chronic wounds. J. Proteome Res. 2010, 9, 4758–4766. [Google Scholar] [CrossRef] [PubMed]
- Stojadinovic, O.; Minkiewicz, J.; Sawaya, A.; Bourne, J.W.; Torzilli, P.; de Rivero Vaccari, J.P.; Dietrich, W.D.; Keane, R.W.; Tomic-Canic, M. Deep Tissue Injury in Development of Pressure Ulcers: A Decrease of Inflammasome Activation and Changes in Human Skin Morphology in Response to Aging and Mechanical Load. PLoS ONE 2013, 8, e69223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baltzis, D.; Eleftheriadou, I.; Veves, A. Pathogenesis and Treatment of Impaired Wound Healing in Diabetes Mellitus: New Insights. Adv. Ther. 2014, 31, 817–836. [Google Scholar] [CrossRef]
- Sgonc, R.; Gruber, J. Age-related aspects of cutaneous wound healing: A mini-review. Gerontology 2013, 59, 159–164. [Google Scholar] [CrossRef] [PubMed]
- Friedman, S.L.; Sheppard, D.; Duffield, J.S.; Violette, S. Therapy for Fibrotic Diseases: Nearing the Starting Line. Sci. Transl. Med. 2013, 5, e3004700. [Google Scholar] [CrossRef] [Green Version]
- Berman, B.; Maderal, A.; Raphael, B. Keloids and hypertrophic scars: Pathophysiology, classification, and treatment. Dermatol. Surg. 2017, 43, S3–S18. [Google Scholar] [CrossRef]
- Stoica, A.E.; Grumezescu, A.M.; Hermenean, A.O.; Andronescu, E.; Vasile, B.S. Scar-Free Healing: Current Concepts and Future Perspectives. Nanomaterials 2020, 10, 2179. [Google Scholar] [CrossRef] [PubMed]
- Keshtkar, S.; Azarpira, N.; Ghahremani, M.H. Mesenchymal stem cell-derived extracellular vesicles: Novel frontiers in regenerative medicine. Stem Cell Res. Ther. 2018, 9, 63. [Google Scholar] [CrossRef]
- Bray, E.R.; Oropallo, A.R.; Grande, D.A.; Kirsner, R.S.; Badiavas, E.V. Extracellular vesicles as therapeutic tools for the treatment of chronic wounds. Pharmaceutics 2021, 13, 1543. [Google Scholar] [CrossRef]
- Yu, B.; Zhang, X.; Li, X. Exosomes derived from mesenchymal stem cells. Int. J. Mol. Sci. 2014, 15, 4142–4157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Han, Y.; Li, X.; Zhang, Y.; Han, Y.; Chang, F.; Ding, J. Mesenchymal Stem Cells for Regenerative Medicine. Cells 2019, 8, 886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hade, M.D.; Suire, C.N.; Mossell, J.; Suo, Z. Extracellular vesicles: Emerging frontiers in wound healing. Med. Res. Rev. 2022, 42, 2102–2125. [Google Scholar] [CrossRef]
- Riedl, J.; Popp, C.; Eide, C.; Ebens, C.; Tolar, J. Mesenchymal stromal cells in wound healing applications: Role of the secretome, targeted delivery and impact on recessive dystrophic epidermolysis bullosa treatment. Cytotherapy 2021, 23, 961–973. [Google Scholar] [CrossRef] [PubMed]
- Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Doyle, L.; Wang, M. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [Green Version]
- Lu, S.; Lu, L.; Liu, Y.; Li, Z.; Fang, Y.; Chen, Z.; Zhou, J. Native and engineered extracellular vesicles for wound healing. Front. Bioeng. Biotechnol. 2022, 10, 1053217. [Google Scholar] [CrossRef]
- Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
- Akbar, A.; Malekian, F.; Baghban, N.; Kodam, S.P.; Ullah, M. Methodologies to Isolate and Purify Clinical Grade Extracellular Vesicles for Medical Applications. Cells 2022, 11, 186. [Google Scholar] [CrossRef]
- Rani, S.; Ryan, A.E.; Griffin, M.D.; Ritter, T. Mesenchymal stem cell-derived extracellular vesicles: Toward cell-free therapeutic applications. Mol. Ther. 2015, 23, 812–823. [Google Scholar] [CrossRef] [Green Version]
- Baranovskii, D.S.; Klabukov, I.D.; Arguchinskaya, N.V.; Yakimova, A.O.; Kisel, A.A.; Yatsenko, E.M.; Ivanov, S.A.; Shegay, P.V.; Kaprin, A.D. Adverse events, side effects and complications in mesenchymal stromal cell-based therapies. Stem Cell Investig. 2022, 9, 7. [Google Scholar] [CrossRef]
- Narauskaitė, D.; Vydmantaitė, G.; Rusteikaitė, J.; Sampath, R.; Rudaitytė, A.; Stašytė, G.; Calvente, M.I.A.; Jekabsone, A. Extracellular vesicles in skin wound healing. Pharmaceuticals 2021, 14, 811. [Google Scholar] [CrossRef]
- Ramos-Zaldívar, H.M.; Polakovicova, I.; Salas-Huenuleo, E.; Corvalán, A.H.; Kogan, M.J.; Yefi, C.P.; Andia, M.E. Extracellular vesicles through the blood–brain barrier: A review. Fluids Barriers CNS 2022, 19, 60. [Google Scholar] [CrossRef] [PubMed]
- Vizoso, F.J.; Eiro, N.; Cid, S.; Schneider, J.; Perez-Fernandez, R. Mesenchymal stem cell secretome: Toward cell-free therapeutic strategies in regenerative medicine. Int. J. Mol. Sci. 2017, 18, 1852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Silachev, D.N.; Goryunov, K.V.; Shpilyuk, M.A.; Beznoschenko, O.S.; Morozova, N.Y.; Kraevaya, E.E.; Popkov, V.A.; Pevzner, I.B.; Zorova, L.D.; Evtushenko, E.A.; et al. Effect of MSCs and MSC-derived extracellular vesicles on human blood coagulation. Cells 2019, 8, 258. [Google Scholar] [CrossRef] [PubMed]
- Fiedler, T.; Rabe, M.; Mundkowski, R.G.; Oehmcke-Hecht, S.; Peters, K. Adipose-derived mesenchymal stem cells release microvesicles with procoagulant activity. Int. J. Biochem. Cell Biol. 2018, 100, 49–53. [Google Scholar] [CrossRef]
- He, X.; Dong, Z.; Cao, Y.; Wang, H.; Liu, S.; Liao, L.; Jin, Y.; Yuan, L.; Li, B.; Bolontrade, M.F. MSC-Derived Exosome Promotes M2 Polarization and Enhances Cutaneous Wound Healing. Stem Cells Int. 2019, 2019, 7132708. [Google Scholar] [CrossRef] [Green Version]
- Heo, J.S.; Kim, S.; Yang, C.E.; Choi, Y.; Song, S.Y.; Kim, H.O. Human Adipose Mesenchymal Stem Cell-Derived Exosomes: A Key Player in Wound Healing. Tissue Eng. Regen. Med. 2021, 18, 537–548. [Google Scholar] [CrossRef]
- Li, X.; Liu, L.; Yang, J.; Yu, Y.; Chai, J.; Wang, L.; Ma, L.; Yin, H. Exosome Derived from Human Umbilical Cord Mesenchymal Stem Cell Mediates MiR-181c Attenuating Burn-induced Excessive Inflammation. EBioMedicine 2016, 8, 72–82. [Google Scholar] [CrossRef] [Green Version]
- Mardpour, S.; Hamidieh, A.A.; Taleahmad, S.; Sharifzad, F.; Taghikhani, A.; Baharvand, H. Interaction between mesenchymal stromal cell-derived extracellular vesicles and immune cells by distinct protein content. J. Cell. Physiol. 2019, 234, 8249–8258. [Google Scholar] [CrossRef]
- Favaro, E.; Carpanetto, A.; Caorsi, C.; Giovarelli, M.; Angelini, C.; Cavallo-Perin, P.; Tetta, C.; Camussi, G.; Zanone, M.M. Human mesenchymal stem cells and derived extracellular vesicles induce regulatory dendritic cells in type 1 diabetic patients. Diabetologia 2016, 59, 325–333. [Google Scholar] [CrossRef] [Green Version]
- Cao, G.; Chen, B.; Zhang, X.; Chen, H. Human adipose-derived mesenchymal stem cells-derived exosomal microRNA-19b promotes the healing of skin wounds through modulation of the CCL1/TGF-β signaling axis. Clin. Cosmet. Investig. Dermatol. 2020, 13, 957–971. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Xie, X.; Lian, W.; Shi, R.; Han, S.; Zhang, H.; Lu, L.; Li, M. 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. 2018, 50, e0585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, X.; Zhang, L.; Wang, S.; Han, Q.; Zhao, R.C. Exosomes secreted by mesenchymal stem cells promote endothelial cell angiogenesis by transferring miR-125a. J. Cell Sci. 2016, 129, 2182–2189. [Google Scholar] [CrossRef] [Green Version]
- Krawczenko, A.; Bielawska-Pohl, A.; Paprocka, M.; Kraskiewicz, H.; Szyposzynska, A.; Wojdat, E.; Klimczak, A. Microvesicles from Human Immortalized Cell Lines of Endothelial Progenitor Cells and Mesenchymal Stem/Stromal Cells of Adipose Tissue Origin as Carriers of Bioactive Factors Facilitating Angiogenesis. Stem Cells Int. 2020, 2020, 1289380. [Google Scholar] [CrossRef]
- Liu, J.; Yan, Z.; Yang, F.; Huang, Y.; Yu, Y.; Zhou, L.; Sun, Z.; Cui, D.; Yan, Y. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Accelerate Cutaneous Wound Healing by Enhancing Angiogenesis through Delivering Angiopoietin-2. Stem Cell Rev. Rep. 2021, 17, 305–317. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Jiao, Y.; Pan, Y.; Zhang, L.; Gong, H.; Qi, Y.; Wang, M.; Gong, H.; Shao, M.; Wang, X.; et al. Fetal dermal mesenchymal stem cell-derived exosomes accelerate cutaneous wound healing by activating Notch signaling. Stem Cells Int. 2019, 2019, 2402916. [Google Scholar] [CrossRef]
- Qian, L.; Pi, L.; Fang, B.R.; Meng, X.X. Adipose mesenchymal stem cell-derived exosomes accelerate skin wound healing via the lncRNA H19/miR-19b/SOX9 axis. Lab. Investig. 2021, 101, 1254–1266. [Google Scholar] [CrossRef]
- Cheng, S.; Xi, Z.; Chen, G.; Liu, K.; Ma, R.; Zhou, C. Extracellular vesicle-carried microRNA-27b derived from mesenchymal stem cells accelerates cutaneous wound healing via E3 ubiquitin ligase ITCH. J. Cell. Mol. Med. 2020, 24, 11254–11271. [Google Scholar] [CrossRef]
- Ren, S.; Chen, J.; Duscher, D.; Liu, Y.; Guo, G.; Kang, Y.; Xiong, H.; Zhan, P.; Wang, Y.; Wang, C.; et al. Microvesicles from human adipose stem cells promote wound healing by optimizing cellular functions via AKT and ERK signaling pathways. Stem Cell Res. Ther. 2019, 10, e1152. [Google Scholar] [CrossRef]
- Zhou, Y.; Zhao, B.; Zhang, X.L.; Lu, Y.J.; Lu, S.T.; Cheng, J.; Fu, Y.; Lin, L.; Zhang, N.Y.; Li, P.X.; et al. Combined topical and systemic administration with human adipose-derived mesenchymal stem cells (hADSC) and hADSC-derived exosomes markedly promoted cutaneous wound healing and regeneration. Stem Cell Res. Ther. 2021, 12, 257. [Google Scholar] [CrossRef]
- Wang, L.; Hu, L.; Zhou, X.; Xiong, Z.; Zhang, C.; Shehada, H.M.A.; Hu, B.; Song, J.; Chen, L. Exosomes secreted by human adipose mesenchymal stem cells promote scarless cutaneous repair by regulating extracellular matrix remodelling. Sci. Rep. 2017, 7, e12919. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Zhang, J.; Shi, J.; Liu, K.; Wang, X.; Jia, Y.; He, T.; Shen, K.; Wang, Y.; Liu, J.; et al. Exosomes derived from human adipose mesenchymal stem cells attenuate hypertrophic scar fibrosis by miR-192-5p/IL-17RA/Smad axis. Stem Cell Res. Ther. 2021, 12, 221. [Google Scholar] [CrossRef] [PubMed]
- Fang, S.; Xu, C.; Zhang, Y.; Xue, C.; Yang, C.; Bi, H.; Qian, X.; Wu, M.; Ji, K.; Zhao, Y.; et al. Umbilical Cord-Derived Mesenchymal Stem Cell-Derived Exosomal MicroRNAs Suppress Myofibroblast Differentiation by Inhibiting the Transforming Growth Factor-β/SMAD2 Pathway during Wound Healing. Stem Cells Transl. Med. 2016, 5, 1425–1439. [Google Scholar] [CrossRef]
- Nallakumarasamy, A.; Jeyaraman, M.; Maffulli, N.; Jeyaraman, N.; Suresh, V.; Ravichandran, S.; Gupta, M.; Potty, A.G.; El-Amin, S.F.; Khanna, M.; et al. Mesenchymal Stromal Cell-Derived Extracellular Vesicles in Wound Healing. Life 2022, 12, 1733. [Google Scholar] [CrossRef] [PubMed]
- Grange, C.; Tapparo, M.; Bruno, S.; Chatterjee, D.; Quesenberry, P.J.; Tetta, C.; Camussi, G. Biodistribution of mesenchymal stem cell-derived extracellular vesicles in a model of acute kidney injury monitored by optical imaging. Int. J. Mol. Med. 2014, 33, 1055–1063. [Google Scholar] [CrossRef] [Green Version]
- Wiklander, O.P.B.; Nordin, J.Z.; O’Loughlin, A.; Gustafsson, Y.; Corso, G.; Mäger, I.; Vader, P.; Lee, Y.; Sork, H.; Seow, Y.; et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J. Extracell. vesicles 2015, 4, e26316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Munagala, R.; Aqil, F.; Jeyabalan, J.; Gupta, R.C. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2016, 371, 48–61. [Google Scholar] [CrossRef] [Green Version]
- Charoenviriyakul, C.; Takahashi, Y.; Morishita, M.; Matsumoto, A.; Nishikawa, M.; Takakura, Y. Cell type-specific and common characteristics of exosomes derived from mouse cell lines: Yield, physicochemical properties, and pharmacokinetics. Eur. J. Pharm. Sci. 2017, 96, 316–322. [Google Scholar] [CrossRef]
- Takahashi, Y.; Nishikawa, M.; Shinotsuka, H.; Matsui, Y.; Ohara, S.; Imai, T.; Takakura, Y. Visualization and in vivo tracking of the exosomes of murine melanoma B16-BL6 cells in mice after intravenous injection. J. Biotechnol. 2013, 165, 77–84. [Google Scholar] [CrossRef]
- Xiong, Y.; Chen, L.; Liu, P.; Yu, T.; Lin, C.; Yan, C.; Hu, Y.; Zhou, W.; Sun, Y.; Panayi, A.C.; et al. All-in-One: Multifunctional Hydrogel Accelerates Oxidative Diabetic Wound Healing through Timed-Release of Exosome and Fibroblast Growth Factor. Small 2022, 18, e202104229. [Google Scholar] [CrossRef]
- Kwak, G.; Cheng, J.; Kim, H.; Song, S.; Lee, S.J.; Yang, Y.; Jeong, J.H.; Lee, J.E.; Messersmith, P.B.; Kim, S.H. Sustained Exosome-Guided Macrophage Polarization Using Hydrolytically Degradable PEG Hydrogels for Cutaneous Wound Healing: Identification of Key Proteins and MiRNAs, and Sustained Release Formulation. Small 2022, 18, e202200060. [Google Scholar] [CrossRef]
- Imai, T.; Takahashi, Y.; Nishikawa, M.; Kato, K.; Morishita, M.; Yamashita, T.; Matsumoto, A.; Charoenviriyakul, C.; Takakura, Y. Macrophage-dependent clearance of systemically administered B16BL6-derived exosomes from the blood circulation in mice. J. Extracell. Vesicles 2015, 4, e26238. [Google Scholar] [CrossRef]
- Dhivya, S.; Padma, V.V.; Santhini, E. Wound dressings—A review. BioMedicine 2015, 5, 24–28. [Google Scholar] [CrossRef]
- Shi, C.; Wang, C.; Liu, H.; Li, Q.; Li, R.; Zhang, Y.; Liu, Y.; Shao, Y.; Wang, J. Selection of Appropriate Wound Dressing for Various Wounds. Front. Bioeng. Biotechnol. 2020, 8, 182. [Google Scholar] [CrossRef] [Green Version]
- Frazier, T.; Alarcon, A.; Wu, X.; Mohiuddin, O.A.; Motherwell, J.M.; Carlsson, A.H.; Christy, R.J.; Edwards, J.V.; Mackin, R.T.; Prevost, N.; et al. Clinical translational potential in skin wound regeneration for adipose-derived, blood-derived, and cellulose materials: Cells, exosomes, and hydrogels. Biomolecules 2020, 10, 1373. [Google Scholar] [CrossRef] [PubMed]
- Boateng, J.; Catanzano, O. Advanced Therapeutic Dressings for Effective Wound Healing—A Review. J. Pharm. Sci. 2015, 104, 3653–3680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rice, J.J.; Martino, M.M.; De Laporte, L.; Tortelli, F.; Briquez, P.S.; Hubbell, J.A. Engineering the regenerative microenvironment with biomaterials. Adv. Healthc. Mater. 2013, 2, 57–71. [Google Scholar] [CrossRef]
- Khayambashi, P.; Iyer, J.; Pillai, S.; Upadhyay, A.; Zhang, Y.; Tran, S.D. Hydrogel encapsulation of mesenchymal stem cells and their derived exosomes for tissue engineering. Int. J. Mol. Sci. 2021, 22, 684. [Google Scholar] [CrossRef] [PubMed]
- Huang, J.; Xiong, J.; Yang, L.; Zhang, J.; Sun, S.; Liang, Y. Cell-free exosome-laden scaffolds for tissue repair. Nanoscale 2021, 13, 8740–8750. [Google Scholar] [CrossRef]
- Brennan, M.; Layrolle, P.; Mooney, D.J. Biomaterials functionalized with MSC secreted extracellular vesicles and soluble factors for tissue regeneration. Adv. Funct. Mater. 2020, 30, e201909125. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Sun, M.; Wu, S. State-of-the-Art Review of Electrospun Gelatin-Based Nanofiber Dressings for Wound Healing Applications. Nanomaterials 2022, 12, 784. [Google Scholar] [CrossRef] [PubMed]
- Németh, K.; Kazsoki, A.; Visnovitz, T.; Pinke, B.; Mészáros, L.; Buzás, E.I.; Zelkó, R. Nanofiber formation as a promising technology for preservation and easy storage of extracellular vesicles. Sci. Rep. 2022, 12, 22012. [Google Scholar] [CrossRef]
- Wang, L.; Cheng, W.; Zhu, J.; Li, W.; Li, D.; Yang, X.; Zhao, W.; Ren, M.; Ren, J.; Mo, X.; et al. Electrospun nanoyarn and exosomes of adipose-derived stem cells for urethral regeneration: Evaluations in vitro and in vivo. Colloids Surf. B Biointerfaces 2022, 209, 112218. [Google Scholar] [CrossRef] [PubMed]
- Nieuwoudt, M.; Woods, I.; Eichholz, K.F.; Martins, C.; McSweeney, K.; Shen, N.; Hoey, D.A. Functionalization of Electrospun Polycaprolactone Scaffolds with Matrix-Binding Osteocyte-Derived Extracellular Vesicles Promotes Osteoblastic Differentiation and Mineralization. Ann. Biomed. Eng. 2021, 49, 3621–3635. [Google Scholar] [CrossRef]
- Elangwe, C.N.; Morozkina, S.N.; Olekhnovich, R.O.; Krasichkov, A.; Polyakova, V.O.; Uspenskaya, M.V. A Review on Chitosan and Cellulose Hydrogels for Wound Dressings. Polymers 2022, 14, 5163. [Google Scholar] [CrossRef] [PubMed]
- Abolgheit, S.; Abdelkader, S.; Aboushelib, M.; Omar, E.; Mehanna, R. Bone marrow-derived mesenchymal stem cells and extracellular vesicles enriched collagen chitosan scaffold in skin wound healing (a rat model). J. Biomater. Appl. 2021, 36, 128–139. [Google Scholar] [CrossRef]
- Tao, S.-C.; Guo, S.-C.; Li, M.; Ke, Q.-F.; Guo, Y.-P.; Zhang, C.-Q. Chitosan Wound Dressings Incorporating Exosomes Derived from MicroRNA-126-Overexpressing Synovium Mesenchymal Stem Cells Provide Sustained Release of Exosomes and Heal Full-Thickness Skin Defects in a Diabetic Rat Model. Stem Cells Transl. Med. 2017, 6, 736–747. [Google Scholar] [CrossRef] [PubMed]
- Huang, F.; Fang, Z.F.; Hu, X.Q.; Tang, L.; Zhou, S.H.; Huang, J.P. Overexpression of miR-126 promotes the differentiation of mesenchymal stem cells toward endothelial cells via activation of PI3K/Akt and MAPK/ERK pathways and release of paracrine factors. Biol. Chem. 2013, 394, 1223–1233. [Google Scholar] [CrossRef]
- Nooshabadi, V.T.; Khanmohamadi, M.; Valipour, E.; Mahdipour, S.; Salati, A.; Malekshahi, Z.V.; Shafei, S.; Amini, E.; Farzamfar, S.; Ai, J. Impact of exosome-loaded chitosan hydrogel in wound repair and layered dermal reconstitution in mice animal model. J. Biomed. Mater. Res. 2020, 108, 2138–2149. [Google Scholar] [CrossRef]
- Li, Q.; Gong, S.; Yao, W.; Yang, Z.; Wang, R.; Yu, Z.; Wei, M. Exosome loaded genipin crosslinked hydrogel facilitates full thickness cutaneous wound healing in rat animal model. Drug Deliv. 2021, 28, 884–893. [Google Scholar] [CrossRef]
- Wang, C.; Liang, C.; Wang, R.; Yao, X.; Guo, P.; Yuan, W.; Liu, Y.; Song, Y.; Li, Z.; Xie, X. The fabrication of a highly efficient self-healing hydrogel from natural biopolymers loaded with exosomes for the synergistic promotion of severe wound healing. Biomater. Sci. 2020, 8, 313–324. [Google Scholar] [CrossRef] [PubMed]
- Xu, L.; Wang, Y.Y.; Huang, J.; Chen, C.Y.; Wang, Z.X.; Xie, H. Silver nanoparticles: Synthesis, medical applications and biosafety. Theranostics 2020, 10, 8996–9031. [Google Scholar] [CrossRef] [PubMed]
- Qian, Z.; Bai, Y.; Zhou, J.; Li, L.; Na, J.; Fan, Y.; Guo, X.; Liu, H. A moisturizing chitosan-silk fibroin dressing with silver nanoparticles-adsorbed exosomes for repairing infected wounds. J. Mater. Chem. B 2020, 8, 7197–7212. [Google Scholar] [CrossRef]
- Liu, Y.; Liu, Y.; Zhao, Y.; Wu, M.; Mao, S.; Cong, P.; Zou, R.; Hou, M.; Jin, H.; Bao, Y. Application of adipose mesenchymal stem cell-derived exosomes-loaded β-chitin nanofiber hydrogel for wound healing. Folia Histochem. Cytobiol. 2022, 60, 167–178. [Google Scholar] [CrossRef]
- Zhang, M.; Zhao, X. Alginate hydrogel dressings for advanced wound management. Int. J. Biol. Macromol. 2020, 162, 1414–1428. [Google Scholar] [CrossRef]
- Shafei, S.; Khanmohammadi, M.; Heidari, R.; Ghanbari, H.; Taghdiri Nooshabadi, V.; Farzamfar, S.; Akbariqomi, M.; Sanikhani, N.S.; Absalan, M.; Tavoosidana, G. Exosome loaded alginate hydrogel promotes tissue regeneration in full-thickness skin wounds: An in vivo study. J. Biomed. Mater. Res. 2020, 108, 545–556. [Google Scholar] [CrossRef]
- Bari, E.; Di Silvestre, D.; Mastracci, L.; Grillo, F.; Grisoli, P.; Marrubini, G.; Nardini, M.; Mastrogiacomo, M.; Sorlini, M.; Rossi, R.; et al. GMP-compliant sponge-like dressing containing MSC lyo-secretome: Proteomic network of healing in a murine wound model. Eur. J. Pharm. Biopharm. 2020, 155, 37–48. [Google Scholar] [CrossRef]
- Shen, Y.; Xu, G.; Huang, H.; Wang, K.; Wang, H.; Lang, M.; Gao, H.; Zhao, S. Sequential Release of Small Extracellular Vesicles from Bilayered Thiolated Alginate/Polyethylene Glycol Diacrylate Hydrogels for Scarless Wound Healing. ACS Nano 2021, 15, 6352–6368. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.; Li, Z.; Wang, Y.; Li, L.; Wang, D.; Zhang, W.; Liu, L.; Jiang, H.; Yang, J.; Cheng, J. Overexpression of miR-29b reduces collagen biosynthesis by inhibiting heat shock protein 47 during skin wound healing. Transl. Res. 2016, 178, 38–53.e6. [Google Scholar] [CrossRef] [PubMed]
- Yang, S.; Jiang, H.; Qian, M.; Ji, G.; Wei, Y.; He, J.; Tian, H.; Zhao, Q. MSC-derived sEV-loaded hyaluronan hydrogel promotes scarless skin healing by immunomodulation in a large skin wound model. Biomed. Mater. 2022, 17, 034104. [Google Scholar] [CrossRef]
- Henriques-Antunes, H.; Cardoso, R.M.S.; Zonari, A.; Correia, J.; Leal, E.C.; Jiménez-Balsa, A.; Lino, M.M.; Barradas, A.; Kostic, I.; Gomes, C.; et al. The Kinetics of Small Extracellular Vesicle Delivery Impacts Skin Tissue Regeneration. ACS Nano 2019, 13, 8694–8707. [Google Scholar] [CrossRef]
- Sarrigiannidis, S.O.; Rey, J.M.; Dobre, O.; González-García, C.; Dalby, M.J.; Salmeron-Sanchez, M. A tough act to follow: Collagen hydrogel modifications to improve mechanical and growth factor loading capabilities. Mater. Today Bio 2021, 10, 100098. [Google Scholar] [CrossRef]
- Liu, W.; Lin, H.; Zhao, P.; Xing, L.; Li, J.; Wang, Z.; Ju, S.; Shi, X.L.; Liu, Y.; Deng, G.; et al. A regulatory perspective on recombinant collagen-based medical devices. Bioact. Mater. 2021, 12, 198–202. [Google Scholar] [CrossRef]
- Xu, L.; Liu, Y.; Tang, L.; Xiao, H.; Yang, Z.; Wang, S. Preparation of Recombinant Human Collagen III Protein Hydrogels with Sustained Release of Extracellular Vesicles for Skin Wound Healing. Int. J. Mol. Sci. 2022, 23, 6289. [Google Scholar] [CrossRef]
- Diniz, I.M.A.; Chen, C.; Xu, X.; Ansari, S.; Zadeh, H.H.; Marques, M.M.; Shi, S.; Moshaverinia, A. Pluronic F-127 hydrogel as a promising scaffold for encapsulation of dental-derived mesenchymal stem cells. J. Mater. Sci. Mater. Med. 2015, 26, 153. [Google Scholar] [CrossRef] [Green Version]
- Wang, M.; Wang, C.; Chen, M.; Xi, Y.; Cheng, W.; Mao, C.; Xu, T.; Zhang, X.; Lin, C.; Gao, W.; et al. Efficient Angiogenesis-Based Diabetic Wound Healing/Skin Reconstruction through Bioactive Antibacterial Adhesive Ultraviolet Shielding Nanodressing with Exosome Release. ACS Nano 2019, 13, 10279–10293. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.; Wang, M.; Xu, T.; Zhang, X.; Lin, C.; Gao, W.; Xu, H.; Lei, B.; Mao, C. Engineering Bioactive Self-Healing Antibacterial Exosomes Hydrogel for Promoting Chronic Diabetic Wound Healing and Complete Skin Regeneration. Theranostics 2019, 9, 65–76. [Google Scholar] [CrossRef]
- Monavarian, M.; Kader, S.; Moeinzadeh, S.; Jabbari, E. Regenerative Scar-Free Skin Wound Healing. Tissue Eng. Rev. 2019, 25, 294–311. [Google Scholar] [CrossRef]
- Zhou, Y.; Zhang, X.L.; Lu, S.T.; Zhang, N.Y.; Zhang, H.J.; Zhang, J.; Zhang, J. Human adipose-derived mesenchymal stem cells-derived exosomes encapsulated in pluronic F127 hydrogel promote wound healing and regeneration. Stem Cell Res. Ther. 2022, 13, 407. [Google Scholar] [CrossRef]
- Yang, J.; Chen, Z.; Pan, D.; Li, H.; Shen, J. Umbilical cord-derived mesenchymal stem cell-derived exosomes combined pluronic F127 hydrogel promote chronic diabetic wound healing and complete skin regeneration. Int. J. Nanomed. 2020, 15, 5911–5926. [Google Scholar] [CrossRef]
- Guruswamy Damodaran, R.; Vermette, P. Tissue and organ decellularization in regenerative medicine. Biotechnol. Prog. 2018, 34, 1494–1505. [Google Scholar] [CrossRef]
- Andrée, B.; Bär, A.; Haverich, A.; Hilfiker, A. Small intestinal submucosa segments as matrix for tissue engineering: Review. Tissue Eng. Rev. 2013, 19, 279–291. [Google Scholar] [CrossRef]
- Ma, S.; Hu, H.; Wu, J.; Li, X.; Ma, X.; Zhao, Z.; Liu, Z.; Wu, C.; Zhao, B.; Wang, Y.; et al. Functional extracellular matrix hydrogel modified with MSC-derived small extracellular vesicles for chronic wound healing. Cell Prolif. 2022, 55, e13196. [Google Scholar] [CrossRef]
- Fénelon, M.; Catros, S.; Meyer, C.; Fricain, J.C.; Obert, L.; Auber, F.; Louvrier, A.; Gindraux, F. Applications of human amniotic membrane for tissue engineering. Membranes 2021, 11, 387. [Google Scholar] [CrossRef]
- Xiao, S.; Xiao, C.; Miao, Y.; Wang, J.; Chen, R.; Fan, Z.; Hu, Z. Human acellular amniotic membrane incorporating exosomes from adipose-derived mesenchymal stem cells promotes diabetic wound healing. Stem Cell Res. Ther. 2021, 12, 255. [Google Scholar] [CrossRef]
- Wendels, S.; Avérous, L. Biobased polyurethanes for biomedical applications. Bioact. Mater. 2020, 6, 1083–1106. [Google Scholar] [CrossRef]
- Shiekh, P.A.; Singh, A.; Kumar, A. Exosome laden oxygen releasing antioxidant and antibacterial cryogel wound dressing OxOBand alleviate diabetic and infectious wound healing. Biomaterials 2020, 249, 120020. [Google Scholar] [CrossRef]
- Shiekh, P.A.; Singh, A.; Kumar, A. Engineering Bioinspired Antioxidant Materials Promoting Cardiomyocyte Functionality and Maturation for Tissue Engineering Application. ACS Appl. Mater. Interfaces 2018, 10, 3260–3273. [Google Scholar] [CrossRef]
- Shiekh, P.A.; Singh, A.; Kumar, A. Oxygen-Releasing Antioxidant Cryogel Scaffolds with Sustained Oxygen Delivery for Tissue Engineering Applications. ACS Appl. Mater. Interfaces 2018, 10, 18458–18469. [Google Scholar] [CrossRef]
- Wang, J.; Zhang, D.; Zhu, Y.; Mo, X.; McHugh, P.C.; Tong, Q. Astragalus and human mesenchymal stem cells promote wound healing by mediating immunomodulatory effects through paracrine signaling. Regen. Med. 2022, 17, 219–232. [Google Scholar] [CrossRef]
- Rodríguez-Rodríguez, R.; García-Carvajal, Z.Y.; Jiménez-Palomar, I.; Jiménez-Avalos, J.A.; Espinosa-Andrews, H. Development of gelatin/chitosan/PVA hydrogels: Thermal stability, water state, viscoelasticity, and cytotoxicity assays. J. Appl. Polym. Sci. 2019, 136, 47149. [Google Scholar] [CrossRef]
- Tyliszczak, B.; Drabczyk, A.; Kudłacik-Kramarczyk, S.; Bialik-Wąs, K.; Kijkowska, R.; Sobczak-Kupiec, A. Preparation and cytotoxicity of chitosan-based hydrogels modified with silver nanoparticles. Colloids Surf. B Biointerfaces 2017, 160, 325–330. [Google Scholar] [CrossRef]
- Ye, J.; Xiao, Z.; Gao, L.; Zhang, J.; He, L.; Zhang, H.; Liu, Q.; Yang, G. Assessment of the effects of four crosslinking agents on gelatin hydrogel for myocardial tissue engineering applications. Biomed. Mater. 2021, 16, 045026. [Google Scholar] [CrossRef]
Biomaterial | Origin | Culture | Isolation | [EV] | Model | Treatment * | Ref. |
---|---|---|---|---|---|---|---|
Collagen chitosan scaffold | Rat Bone marrow MSC (BMMSC) | 80% confluent cells in LG-DMEM serum-free medium for 48 h | Ultracentrifugation | EV isolated from 106 BMMSC | Sprague-Dawley rats | Not specified | [96] |
Chitosan Hydrogel | Human Synovium MSC (SMSC) and SMSC overexpressing miR-126-3p | 50–60% confluent cells in MesenGro hMSC medium for 48 h | Ultrafiltration-ultracentrifugation | Not specified | Diabetic sprague-Dawley rats (streptozotocin-induced) | Not specified | [97] |
Chitosan Hydrogel | Human endometrial stem cell (hEnSC) | 80% confluent cells in DMEM/F12 + 15% exo-free serum for 24 h | Ultracentrifugation | 100 μg/mL (200 μL/treatment) | BALB/c mice | 0, 3 and 7 days | [99] |
Chitosan genipin crosslinked hydrogel | Human umbilical MSCs (hUCMSC) | Not specified | Ultracentrifugation | 20 μg/mL (300 μL/treatment) | Sprague-Dawley rats | every three days | [100] |
Methylcellulose-chitosan hydrogel | Human placental MSC (hPMSC) | 80% confluent cells. Medium and culture time are not specified | Exosome isolation reagent (Precipitation) | 2 × 1012 EV/mL (100 μL/treatment) | Diabetic mice (C57BLKS-Leprdb) | Not specified | [101] |
Chitosan dressing | Human umbilical MSCs (hUCMSC) | Not specified | Size Exclusion Chromatography-ultrafiltration | Not specified | BALB/c mice | every two days | [103] |
β-chitin nanofiber hydrogel | Mouse adipose MSC (AMSC) | Not specified | Not specified | 200 μg/mL (400 μL/treatment) | Sprague-Dawley rats | Not specified | [104] |
Alginate hydrogel | Rat adipose MSC (AMSC) | 80% confluent cells. Medium and culture time are not specified | Ultracentrifugation | 100 μg/mL (300 μL/treatment) | Wistar rats | Not specified | [106] |
Alginate sponge-like dressing | Human adipose MSC (AMSC) | Sub-confluent cells in DMEM/F12 platelet lysate free for 48 h | Ultrafiltration (isolation of soluble proteins and EV) | EV isolated from 4 × 106 cells/mL (500 μL/treatment) | Mice | Not specified | [107] |
Bilayered Thiolated Alginate/Polyethylene Glycol Diacrylate Hydrogels | Human Bone marrow MSC (hBMMSC) and hBMSC overexpressing miR-29b-3p | 80% confluent cells in LG-DMEM serum-free medium for 48 h | Ultracentrifugation | 1011 EV/mL | Sprague-Dawley rats | Not specified | [108] |
Hyaluronic acid hydrogel | Human placental MSC (hPMSC) | Confluent cell not specified. DMEM serum-free medium for 48 h | Ultracentrifugation | 1000 μg/mL (100 μL/treatment) | C57BL/6J mice | Not specified | [110] |
Hyaluronic acid-light-triggerable hydrogel | Human umbilical Mononuclear cells (hUCMNC) | 2 × 106 cells/mL in X-VIVO 15 serum-free cell culture medium supplemented with Flt-3 and stem-cell factor in hypoxia (0.5% O2) for 18 h | Ultracentrifugation | 200 μg/treatment | Diabetic C57BL/6 mice (streptozotocin-induced) | One time and light-activated each day for 1 min | [111] |
RhCollagen III Hydrogel | Human umbilical MSCs (hUCMSC) | Not specified | Ultracentrifugation | Not specified | Diabetic sprague-Dawley rats (streptozotocin-induced) | every three days | [114] |
Pluronic F127 hydrogel | Mouse adipose MSC (AMSC) | 6 × 104 cells/cm2 in DMEM + 3% exo-free serum for 48 h | Ultracentrifugation | 10 μg/mL | Diabetic ICR mice (streptozotocin-induced) | Not specified | [116] |
Pluronic F127-oxidized hyaluronic acid and poly-ε-lysine hydrogel | Mouse adipose MSC (AMSC) | Not specified | Ultracentrifugation | 10 μg/treatment | Diabetic ICR mice (streptozotocin-induced) | Not specified | [117] |
Pluronic F127 hydrogel | Human adipose MSC (AMSC) | 80–90% confluent cells in α-MEM + 10% UltraGROTM-Advanced medium for 48 h | Ultracentrifugation | 1000 μg/mL (100 μL/treatment) | ICR mice | every three days | [119] |
Pluronic F127 hydrogel | Human umbilical MSCs (hUCMSC) | Not specified | ExoQuick-TC | 1000 μg/mL (100 μL/treatment) | Diabetic sprague-Dawley rats (streptozotocin-induced) | Not specified | [120] |
porcine small intestinal submucosa-based hydrogel | Human umbilical MSCs (hUCMSC) | Culture medium (DMEM + 10% exo-free serum) was collected when the cells reached 80–85% confluency. | Ultracentrifugation | Not specified | Diabetic sprague-Dawley rats (streptozotocin-induced) | Not specified | [123] |
Human acellular amniotic membrane | Human adipose MSC (AMSC) | 60–65% confluent cell in DMEM + 10% exo-free serum for 48 h | Exosome isolation reagent (Precipitation) | 100 μg/treatment | Diabetic BALB/c mice (streptozotocin-induced) | every other day, three times in total | [125] |
Antioxidant polyurethane dressing | Rat adipose MSC (AMSC) | 80% confluent cells in α-MEM serum-free medium for 48 h | Ultrafiltration | 100 μg/treatment | Diabetic wistar rats (streptozotocin-induced) | Not specified | [129] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Pulido-Escribano, V.; Torrecillas-Baena, B.; Dorado, G.; Gálvez-Moreno, M.Á.; Camacho-Cardenosa, M.; Casado-Díaz, A. Combination of Biomaterials and Extracellular Vesicles from Mesenchymal Stem-Cells: New Therapeutic Strategies for Skin-Wound Healing. Appl. Sci. 2023, 13, 2702. https://doi.org/10.3390/app13042702
Pulido-Escribano V, Torrecillas-Baena B, Dorado G, Gálvez-Moreno MÁ, Camacho-Cardenosa M, Casado-Díaz A. Combination of Biomaterials and Extracellular Vesicles from Mesenchymal Stem-Cells: New Therapeutic Strategies for Skin-Wound Healing. Applied Sciences. 2023; 13(4):2702. https://doi.org/10.3390/app13042702
Chicago/Turabian StylePulido-Escribano, Victoria, Bárbara Torrecillas-Baena, Gabriel Dorado, María Ángeles Gálvez-Moreno, Marta Camacho-Cardenosa, and Antonio Casado-Díaz. 2023. "Combination of Biomaterials and Extracellular Vesicles from Mesenchymal Stem-Cells: New Therapeutic Strategies for Skin-Wound Healing" Applied Sciences 13, no. 4: 2702. https://doi.org/10.3390/app13042702
APA StylePulido-Escribano, V., Torrecillas-Baena, B., Dorado, G., Gálvez-Moreno, M. Á., Camacho-Cardenosa, M., & Casado-Díaz, A. (2023). Combination of Biomaterials and Extracellular Vesicles from Mesenchymal Stem-Cells: New Therapeutic Strategies for Skin-Wound Healing. Applied Sciences, 13(4), 2702. https://doi.org/10.3390/app13042702