422
Views
5
CrossRef citations to date
0
Altmetric
REVIEW

Mesenchymal Stem Cell Derived Exosomes Therapy in Diabetic Wound Repair

ORCID Icon, & ORCID Icon
Pages 2707-2720 | Received 25 Mar 2023, Accepted 15 May 2023, Published online: 22 May 2023

References

  • Boulton AJ, Vileikyte L, Ragnarson-Tennvall G, Apelqvist J. The global burden of diabetic foot disease. Lancet. 2005;366(9498):1719–1724. doi:10.1016/s0140-6736(05)67698-2
  • Baltzis D, Eleftheriadou I, Veves A. Pathogenesis and treatment of impaired wound healing in diabetes mellitus: new insights. Adv Ther. 2014;31(8):817–836. doi:10.1007/s12325-014-0140-x
  • Lopes L, Setia O, Aurshina A, et al. Stem cell therapy for diabetic foot ulcers: a review of preclinical and clinical research. Stem Cell Res Ther. 2018;9(1):188. doi:10.1186/s13287-018-0938-6
  • Taverna S, Pucci M, Alessandro R. Extracellular vesicles: small bricks for tissue repair/regeneration. Ann Trans Med. 2017;5(4):83. doi:10.21037/atm.2017.01.53
  • An Y, Lin S, Tan X, et al. Exosomes from adipose-derived stem cells and application to skin wound healing. Cell Prolif. 2021;54(3):e12993. doi:10.1111/cpr.12993
  • Vizoso FJ, 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(9). doi:10.3390/ijms18091852
  • Rani M, Nicholson SE, Zhang Q, Schwacha MG. Damage-associated molecular patterns (DAMPs) released after burn are associated with inflammation and monocyte activation. Burns. 2017;43(2):297–303. doi:10.1016/j.burns.2016.10.001
  • Barrientos S, Stojadinovic O, Golinko MS, Brem H, Tomic-Canic M. Growth factors and cytokines in wound healing. Wound Repair Regen. 2008;16(5):585–601. doi:10.1111/j.1524-475X.2008.00410.x
  • Yunna C, Mengru H, Lei W, Weidong C. Macrophage M1/M2 polarization. Eur J Pharmacol. 2020;877:173090. doi:10.1016/j.ejphar.2020.173090
  • den Dekker A, Davis FM, Kunkel SL, Gallagher KA. Targeting epigenetic mechanisms in diabetic wound healing. Transl Res. 2019;204:39–50. doi:10.1016/j.trsl.2018.10.001
  • Diegelmann RF, Evans MC. Wound healing: an overview of acute, fibrotic and delayed healing. Front Biosci. 2004;9:283–289. doi:10.2741/1184
  • Gabbiani G. The myofibroblast in wound healing and fibrocontractive diseases. J Pathol. 2003;200(4):500–503. doi:10.1002/path.1427
  • Aitcheson SM, Frentiu FD, Hurn SE, Edwards K, Murray RZ. Skin wound healing: normal macrophage function and macrophage dysfunction in diabetic wounds. Molecules. 2021;26(16). doi:10.3390/molecules26164917
  • Brem H, Tomic-Canic M. Cellular and molecular basis of wound healing in diabetes. J Clin Invest. 2007;117(5):1219–1222. doi:10.1172/jci32169
  • Tecilazich F, Dinh TL, Veves A. Emerging drugs for the treatment of diabetic ulcers. Expert Opin Emerg Drugs. 2013;18(2):207–217. doi:10.1517/14728214.2013.802305
  • Matoori S, Veves A, Mooney DJ. Advanced bandages for diabetic wound healing. Sci Transl Med. 2021;13(585). doi:10.1126/scitranslmed.abe4839
  • Ekstrand AJ, Cao R, Bjorndahl M, et al. Deletion of neuropeptide Y (NPY) 2 receptor in mice results in blockage of NPY-induced angiogenesis and delayed wound healing. Proc Natl Acad Sci USA. 2003;100(10):6033–6038. doi:10.1073/pnas.1135965100
  • Toda M, Suzuki T, Hosono K, et al. Roles of calcitonin gene-related peptide in facilitation of wound healing and angiogenesis. Biomed Pharmacother. 2008;62(6):352–359. doi:10.1016/j.biopha.2008.02.003
  • Pradhan L, Nabzdyk C, Andersen ND, LoGerfo FW, Veves A. Inflammation and neuropeptides: the connection in diabetic wound healing. Expert Rev Mol Med. 2009;11:e2. doi:10.1017/s1462399409000945
  • Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015;25(6):364–372. doi:10.1016/j.tcb.2015.01.004
  • Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367:6478. doi:10.1126/science.aau6977
  • Soltani S, Mansouri K, Parvaneh S, Thakor AS, Pociot F, Yarani R. Diabetes complications and extracellular vesicle therapy. Rev Endocr Metab Disord. 2022;23(3):357–385. doi:10.1007/s11154-021-09680-y
  • Keerthikumar S, Chisanga D, Ariyaratne D, et al. ExoCarta: a web-based compendium of exosomal cargo. J Mol Biol. 2016;428(4):688–692. doi:10.1016/j.jmb.2015.09.019
  • Pathan M, Fonseka P, Chitti SV, et al. Vesiclepedia 2019: a compendium of RNA, proteins, lipids and metabolites in extracellular vesicles. Nucleic Acids Res. 2019;47(D1):D516–d519. doi:10.1093/nar/gky1029
  • Xu YX, Pu SD, Li X, et al. Exosomal ncRNAs: novel therapeutic target and biomarker for diabetic complications. Pharmacol Res. 2022;178:106135. doi:10.1016/j.phrs.2022.106135
  • Wen SW, Lima LG, Lobb RJ, et al. Breast cancer-derived exosomes reflect the cell-of-origin phenotype. Proteomics. 2019;19(8):e1800180. doi:10.1002/pmic.201800180
  • Álvarez-Viejo M. Mesenchymal stem cells from different sources and their derived exosomes: a pre-clinical perspective. World J Stem Cells. 2020;12(2):100–109. doi:10.4252/wjsc.v12.i2.100
  • Xue M, Zhuo Y, Shan B. MicroRNAs, long noncoding RNAs, and their functions in human disease. Methods Mol Biol. 2017;1617:1–25. doi:10.1007/978-1-4939-7046-9_1
  • Bochon B, Kozubska M, Surygała G, et al. Mesenchymal stem cells-potential applications in kidney diseases. Int J Mol Sci. 2019;20(10). doi:10.3390/ijms20102462
  • Vahedi P, Moghaddamshahabi R, Webster TJ, et al. The use of infrapatellar fat pad-derived mesenchymal stem cells in articular cartilage regeneration: a review. Int J Mol Sci. 2021;22(17). doi:10.3390/ijms22179215
  • Pomatto M, Gai C, Negro F, et al. Differential therapeutic effect of extracellular vesicles derived by bone marrow and adipose mesenchymal stem cells on wound healing of diabetic ulcers and correlation to their cargoes. Int J Mol Sci. 2021;22(8):8. doi:10.3390/ijms22083851
  • Barlow S, Brooke G, Chatterjee K, et al. Comparison of human placenta- and bone marrow-derived multipotent mesenchymal stem cells. Stem Cells Dev. 2008;17(6):1095–1107. doi:10.1089/scd.2007.0154
  • Mathew SA, Naik C, Cahill PA, Bhonde RR. Placental mesenchymal stromal cells as an alternative tool for therapeutic angiogenesis. Cell Mol Life Sci. 2020;77(2):253–265. doi:10.1007/s00018-019-03268-1
  • Alcayaga-Miranda F, Cuenca J, Luz-Crawford P, et al. Characterization of menstrual stem cells: angiogenic effect, migration and hematopoietic stem cell support in comparison with bone marrow mesenchymal stem cells. Stem Cell Res Ther. 2015;6(1):32. doi:10.1186/s13287-015-0013-5
  • Las Heras K, Royo F, Garcia-Vallicrosa C, et al. Extracellular vesicles from hair follicle-derived mesenchymal stromal cells: isolation, characterization and therapeutic potential for chronic wound healing. Stem Cell Res Ther. 2022;13(1):147. doi:10.1186/s13287-022-02824-0
  • Chen CY, Rao SS, Ren L, et al. Exosomal DMBT1 from human urine-derived stem cells facilitates diabetic wound repair by promoting angiogenesis. Theranostics. 2018;8(6):1607–1623. doi:10.7150/thno.22958
  • 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. 2017;8:904. doi:10.3389/fphys.2017.00904
  • Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997;276(5309):71–74. doi:10.1126/science.276.5309.71
  • Shabbir A, Cox A, Rodriguez-Menocal L, Salgado M, Van Badiavas E. Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro. Stem Cells Dev. 2015;24(14):1635–1647. doi:10.1089/scd.2014.0316
  • Born LJ, Chang KH, Shoureshi P, et al. HOTAIR-loaded mesenchymal stem/stromal cell extracellular vesicles enhance angiogenesis and wound healing. Adv Healthc Mater. 2022;11(5):e2002070. doi:10.1002/adhm.202002070
  • Lamichhane TN, Leung CA, Douti LY, Jay SM. Ethanol induces enhanced vascularization bioactivity of endothelial cell-derived extracellular vesicles via regulation of MicroRNAs and long non-coding RNAs. Sci Rep. 2017;7(1):13794. doi:10.1038/s41598-017-14356-2
  • Han ZF, Cao JH, Liu ZY, Yang Z, Qi RX, Xu HL. Exosomal lncRNA KLF3-AS1 derived from bone marrow mesenchymal stem cells stimulates angiogenesis to promote diabetic cutaneous wound healing. Diabetes Res Clin Pract. 2022;183:109126. doi:10.1016/j.diabres.2021.109126
  • Li B, Luan S, Chen J, et al. The MSC-derived exosomal lncRNA H19 promotes wound healing in diabetic foot ulcers by upregulating PTEN via MicroRNA-152-3p. Mol Ther Nucleic Acids. 2020;19:814–826. doi:10.1016/j.omtn.2019.11.034
  • Ding J, Wang X, Chen B, Zhang J, Xu J. Exosomes derived from human bone marrow mesenchymal stem cells stimulated by deferoxamine accelerate cutaneous wound healing by promoting angiogenesis. Biomed Res Int. 2019;2019:9742765. doi:10.1155/2019/9742765
  • Zernecke A, Bidzhekov K, Noels H, et al. Delivery of microRNA-126 by apoptotic bodies induces CXCL12-dependent vascular protection. Sci Signal. 2009;2(100):ra81. doi:10.1126/scisignal.2000610
  • Moura J, Sørensen A, Leal EC, et al. microRNA-155 inhibition restores fibroblast growth factor 7 expression in diabetic skin and decreases wound inflammation. Sci Rep. 2019;9(1):5836. doi:10.1038/s41598-019-42309-4
  • Petkovic M, Sørensen AE, Leal EC, Carvalho E, Dalgaard LT. Mechanistic actions of microRNAs in diabetic wound healing. Cells. 2020;9(10). doi:10.3390/cells9102228
  • Gondaliya P, Sayyed AA, Bhat P, et al. Mesenchymal stem cell-derived exosomes loaded with miR-155 inhibitor ameliorate diabetic wound healing. Mol Pharm. 2022;19(5):1294–1308. doi:10.1021/acs.molpharmaceut.1c00669
  • He F, Ru X, Wen T. NRF2, a transcription factor for stress response and beyond. Int J Mol Sci. 2020;21(13). doi:10.3390/ijms21134777
  • Wang L, Cai Y, Zhang Q, Zhang Y. Pharmaceutical activation of Nrf2 accelerates diabetic wound healing by exosomes from bone marrow mesenchymal stem cells. Int J Stem Cells. 2022;15(2):164–172. doi:10.15283/ijsc21067
  • Tsugawa K, Jones MK, Sugimachi K, Sarfeh IJ, Tarnawski AS. Biological role of phosphatase PTEN in cancer and tissue injury healing. Front Biosci. 2002;7:e245–51. doi:10.2741/tsugawa
  • Zhang W, Bai X, Zhao B, et al. Cell-free therapy based on adipose tissue stem cell-derived exosomes promotes wound healing via the PI3K/Akt signaling pathway. Exp Cell Res. 2018;370(2):333–342. doi:10.1016/j.yexcr.2018.06.035
  • Chen YG, Li Z, Wang XF. Where PI3K/Akt meets Smads: the crosstalk determines human embryonic stem cell fate. Cell Stem Cell. 2012;10(3):231–232. doi:10.1016/j.stem.2012.02.008
  • Yu M, Liu W, Li J, et al. Exosomes derived from atorvastatin-pretreated MSC accelerate diabetic wound repair by enhancing angiogenesis via AKT/eNOS pathway. Stem Cell Res Ther. 2020;11(1):350. doi:10.1186/s13287-020-01824-2
  • Zhang Y, Liu NM, Wang Y, Youn JY, Cai H. Endothelial cell calpain as a critical modulator of angiogenesis. Biochim Biophys Acta Mol Basis Dis. 2017;1863(6):1326–1335. doi:10.1016/j.bbadis.2017.03.021
  • Hu Y, Tao R, Chen L, et al. Exosomes derived from pioglitazone-pretreated MSCs accelerate diabetic wound healing through enhancing angiogenesis. J Nanobiotechnology. 2021;19(1):150. doi:10.1186/s12951-021-00894-5
  • Vergadi E, Ieronymaki E, Lyroni K, Vaporidi K, Tsatsanis C. Akt signaling pathway in macrophage activation and M1/M2 polarization. J Immunol. 2017;198(3):1006–1014. doi:10.4049/jimmunol.1601515
  • Liu W, Yu M, Xie D, et al. Melatonin-stimulated MSC-derived exosomes improve diabetic wound healing through regulating macrophage M1 and M2 polarization by targeting the PTEN/AKT pathway. Stem Cell Res Ther. 2020;11(1):259. doi:10.1186/s13287-020-01756-x
  • Hu L, Wang J, Zhou X, et al. Exosomes derived from human adipose mensenchymal stem cells accelerates cutaneous wound healing via optimizing the characteristics of fibroblasts. Sci Rep. 2016;6:32993. doi:10.1038/srep32993
  • Deng L, Du C, Song P, et al. The role of oxidative stress and antioxidants in diabetic wound healing. Oxid Med Cell Longev. 2021;2021:8852759. doi:10.1155/2021/8852759
  • Wolff SP, Jiang ZY, Hunt JV. Protein glycation and oxidative stress in diabetes mellitus and ageing. Free Radic Biol Med. 1991;10(5):339–352. doi:10.1016/0891-5849(91)90040-a
  • Zhang Y, Bai X, Shen K, et al. Exosomes derived from adipose mesenchymal stem cells promote diabetic chronic wound healing through SIRT3/SOD2. Cells. 2022;11(16). doi:10.3390/cells11162568
  • Shi R, Jin Y, Hu W, et al. Exosomes derived from mmu_circ_0000250-modified adipose-derived mesenchymal stem cells promote wound healing in diabetic mice by inducing miR-128-3p/SIRT1-mediated autophagy. Am J Physiol Cell Physiol. 2020;318(5):C848–c856. doi:10.1152/ajpcell.00041.2020
  • Pires Da Silva J, Monceaux K, Guilbert A, et al. SIRT1 protects the heart from ER stress-induced injury by promoting eEF2K/eEF2-dependent autophagy. Cells. 2020;9(2). doi:10.3390/cells9020426
  • Shi R, Jin Y, Zhao S, Yuan H, Shi J, Zhao H. Hypoxic ADSC-derived exosomes enhance wound healing in diabetic mice via delivery of circ-Snhg11 and induction of M2-like macrophage polarization. Biomed Pharmacother. 2022;153:113463. doi:10.1016/j.biopha.2022.113463
  • Liang ZH, Pan NF, Lin SS, et al. Exosomes from mmu_circ_0001052-modified adipose-derived stem cells promote angiogenesis of DFU via miR-106a-5p and FGF4/p38MAPK pathway. Stem Cell Res Ther. 2022;13(1):336. doi:10.1186/s13287-022-03015-7
  • Lv Q, Deng J, Chen Y, Wang Y, Liu B, Liu J. Engineered human adipose stem-cell-derived exosomes loaded with miR-21-5p to promote diabetic cutaneous wound healing. Mol Pharm. 2020;17(5):1723–1733. doi:10.1021/acs.molpharmaceut.0c00177
  • Goyal N, Kesharwani D, Datta M. Lnc-ing non-coding RNAs with metabolism and diabetes: roles of lncRNAs. Cell Mol Life Sci. 2018;75(10):1827–1837. doi:10.1007/s00018-018-2760-9
  • Qiu J, Shu C, Li X, Ye C, Zhang WC. Exosomes from linc00511-overexpressing ADSCs accelerates angiogenesis in diabetic foot ulcers healing by suppressing PAQR3-induced Twist1 degradation. Diabetes Res Clin Pract. 2021;180:109032. doi:10.1016/j.diabres.2021.109032
  • 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. 2018;50(4):1–14. doi:10.1038/s12276-018-0058-5
  • Hsu HH, Wang AYL, Loh CYY, Pai AA, Kao HK. Therapeutic potential of exosomes derived from diabetic adipose stem cells in cutaneous wound healing of db/db mice. Pharmaceutics. 2022;14(6):Jun. doi:10.3390/pharmaceutics14061206
  • Chen CY, Liu SH, Chen CY, Chen PC, Chen CP. Human placenta-derived multipotent mesenchymal stromal cells involved in placental angiogenesis via the PDGF-BB and STAT3 pathways. Biol Reprod. 2015;93(4):103. doi:10.1095/biolreprod.115.131250
  • Wei P, Zhong C, Yang X, et al. Exosomes derived from human amniotic epithelial cells accelerate diabetic wound healing via PI3K-AKT-mTOR-mediated promotion in angiogenesis and fibroblast function. Burns Trauma. 2020;8:tkaa020. doi:10.1093/burnst/tkaa020
  • Zhang E, Gao B, Yang L, Wu X, Wang Z. Notoginsenoside Ft1 promotes fibroblast proliferation via PI3K/Akt/mTOR signaling pathway and benefits wound healing in genetically diabetic mice. J Pharmacol Exp Ther. 2016;356(2):324–332. doi:10.1124/jpet.115.229369
  • Zhang J, Chen C, Hu B, et al. Exosomes derived from human endothelial progenitor cells accelerate cutaneous wound healing by promoting angiogenesis through Erk1/2 signaling. Int J Biol Sci. 2016;12(12):1472–1487. doi:10.7150/ijbs.15514
  • Hade MD, Suire CN, Mossell J, Suo Z. Extracellular vesicles: emerging frontiers in wound healing. Med Res Rev. 2022;42(6):2102–2125. doi:10.1002/med.21918
  • Teng L, Maqsood M, Zhu M, et al. Exosomes derived from human umbilical cord mesenchymal stem cells accelerate diabetic wound healing via promoting M2 macrophage polarization, angiogenesis, and collagen deposition. Int J Mol Sci. 2022;23(18). doi:10.3390/ijms231810421
  • Yan C, Xv Y, Lin Z, et al. Human umbilical cord mesenchymal stem cell-derived exosomes accelerate diabetic wound healing via ameliorating oxidative stress and promoting angiogenesis. Front Bioeng Biotechnol. 2022;10:829868. doi:10.3389/fbioe.2022.829868
  • Bao MH, Feng X, Zhang YW, Lou XY, Cheng Y, Zhou HH. Let-7 in cardiovascular diseases, heart development and cardiovascular differentiation from stem cells. Int J Mol Sci. 2013;14(11):23086–23102. doi:10.3390/ijms141123086
  • Gao S, Mao F, Zhang B, et al. Mouse bone marrow-derived mesenchymal stem cells induce macrophage M2 polarization through the nuclear factor-κB and signal transducer and activator of transcription 3 pathways. Exp Biol Med. 2014;239(3):366–375. doi:10.1177/1535370213518169
  • Taetzsch T, Levesque S, McGraw C, et al. Redox regulation of NF-κB p50 and M1 polarization in microglia. Glia. 2015;63(3):423–440. doi:10.1002/glia.22762
  • Rao J, Qian X, Li G, et al. ATF3-mediated NRF2/HO-1 signaling regulates TLR4 innate immune responses in mouse liver ischemia/reperfusion injury. Am J Transplant. 2015;15(1):76–87. doi:10.1111/ajt.12954
  • Ke B, Shen XD, Ji H, et al. HO-1-STAT3 axis in mouse liver ischemia/reperfusion injury: regulation of TLR4 innate responses through PI3K/PTEN signaling. J Hepatol. 2012;56(2):359–366. doi:10.1016/j.jhep.2011.05.023
  • Ti D, Hao H, Tong C, et al. LPS-preconditioned mesenchymal stromal cells modify macrophage polarization for resolution of chronic inflammation via exosome-shuttled let-7b. J Transl Med. 2015;13:308. doi:10.1186/s12967-015-0642-6
  • Allickson JG, Sanchez A, Yefimenko N, Borlongan CV, Sanberg PR. Recent studies assessing the proliferative capability of a novel adult stem cell identified in menstrual blood. Open Stem Cell J. 2011;3(2011):4–10. doi:10.2174/1876893801103010004
  • Dalirfardouei R, Jamialahmadi K, Jafarian AH, Mahdipour E. Promising effects of exosomes isolated from menstrual blood-derived mesenchymal stem cell on wound-healing process in diabetic mouse model. J Tissue Eng Regen Med. 2019;13(4):555–568. doi:10.1002/term.2799
  • 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. 2016;129(11):2182–2189. doi:10.1242/jcs.170373
  • Martino PA, Heitman N, Rendl M. The dermal sheath: an emerging component of the hair follicle stem cell niche. Exp Dermatol. 2021;30(4):512–521. doi:10.1111/exd.14204
  • Wang B, Liu XM, Liu ZN, et al. Human hair follicle-derived mesenchymal stem cells: isolation, expansion, and differentiation. World J Stem Cells. 2020;12(6):462–470. doi:10.4252/wjsc.v12.i6.462
  • Al-Awqati Q. Terminal differentiation in epithelia: the role of integrins in hensin polymerization. Annu Rev Physiol. 2011;73:401–412. doi:10.1146/annurev-physiol-012110-142253
  • Renner M, Bergmann G, Krebs I, et al. DMBT1 confers mucosal protection in vivo and a deletion variant is associated with Crohn’s disease. Gastroenterology. 2007;133(5):1499–1509. doi:10.1053/j.gastro.2007.08.007
  • Jones BA, Pei M. Synovium-derived stem cells: a tissue-specific stem cell for cartilage engineering and regeneration. Tissue Eng Part B Rev. 2012;18(4):301–311. doi:10.1089/ten.TEB.2012.0002
  • He F, Chen X, Pei M. Reconstruction of an in vitro tissue-specific microenvironment to rejuvenate synovium-derived stem cells for cartilage tissue engineering. Tissue Eng Part A. 2009;15(12):3809–3821. doi:10.1089/ten.TEA.2009.0188
  • Qu K, Wang Z, Lin XL, Zhang K, He XL, Zhang H. MicroRNAs: key regulators of endothelial progenitor cell functions. Clin Chim Acta. 2015;448:65–73. doi:10.1016/j.cca.2015.06.017
  • Tao SC, Guo SC, Li M, Ke QF, Guo YP, Zhang CQ. 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(3):736–747. doi:10.5966/sctm.2016-0275
  • Takahashi Y, Nishikawa M, Shinotsuka H, et al. Visualization and in vivo tracking of the exosomes of murine melanoma B16-BL6 cells in mice after intravenous injection. J Biotechnol. 2013;165(2):77–84. doi:10.1016/j.jbiotec.2013.03.013
  • Naseri-Nosar M, Ziora ZM. Wound dressings from naturally-occurring polymers: a review on homopolysaccharide-based composites. Carbohydr Polym. 2018;189:379–398. doi:10.1016/j.carbpol.2018.02.003
  • Dai T, Tanaka M, Huang YY, Hamblin MR. Chitosan preparations for wounds and burns: antimicrobial and wound-healing effects. Expert Rev Anti Infect Ther. 2011;9(7):857–879. doi:10.1586/eri.11.59
  • Geng X, Qi Y, Liu X, Shi Y, Li H, Zhao L. A multifunctional antibacterial and self-healing hydrogel laden with bone marrow mesenchymal stem cell-derived exosomes for accelerating diabetic wound healing. Biomater Adv. 2022;133:112613. doi:10.1016/j.msec.2021.112613
  • Wu Z, Liu X, Yuan D, Zhao J. Human acellular amniotic membrane is adopted to treat venous ulcers. Exp Ther Med. 2018;16(2):1285–1289. doi:10.3892/etm.2018.6331
  • Zhou H, Wang L, Zhang C, et al. Feasibility of repairing full-thickness skin defects by iPSC-derived epithelial stem cells seeded on a human acellular amniotic membrane. Stem Cell Res Ther. 2019;10(1):155. doi:10.1186/s13287-019-1234-9
  • Xiao S, Xiao C, Miao Y, et al. Human acellular amniotic membrane incorporating exosomes from adipose-derived mesenchymal stem cells promotes diabetic wound healing. Stem Cell Res Ther. 2021;12(1):255. doi:10.1186/s13287-021-02333-6
  • Shiekh PA, 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. doi:10.1016/j.biomaterials.2020.120020
  • Shi M, Gao Y, Lee L, et al. Adaptive gelatin microspheres enhanced stem cell delivery and integration with diabetic wounds to activate skin tissue regeneration. Front Bioeng Biotechnol. 2022;10:813805. doi:10.3389/fbioe.2022.813805
  • Wang C, Wang M, Xu T, et al. Engineering bioactive self-healing antibacterial exosomes hydrogel for promoting chronic diabetic wound healing and complete skin regeneration. Theranostics. 2019;9(1):65–76. doi:10.7150/thno.29766
  • Shafei S, Khanmohammadi M, Heidari R, et al. Exosome loaded alginate hydrogel promotes tissue regeneration in full-thickness skin wounds: an in vivo study. J Biomed Mater Res A. 2020;108(3):545–556. doi:10.1002/jbm.a.36835
  • Jiang T, Liu S, Wu Z, et al. ADSC-exo@MMP-PEG smart hydrogel promotes diabetic wound healing by optimizing cellular functions and relieving oxidative stress. Mater Today Bio. 2022;16:100365. doi:10.1016/j.mtbio.2022.100365
  • 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 Nanomedicine. 2020;15:5911–5926. doi:10.2147/ijn.S249129
  • Zhang Y, Zhang P, Gao X, Chang L, Chen Z, Mei X. Preparation of exosomes encapsulated nanohydrogel for accelerating wound healing of diabetic rats by promoting angiogenesis. Mater Sci Eng C Mater Biol Appl. 2021;120:111671. doi:10.1016/j.msec.2020.111671
  • Kapoor S, Kundu SC. Silk protein-based hydrogels: promising advanced materials for biomedical applications. Acta Biomater. 2016;31:17–32. doi:10.1016/j.actbio.2015.11.034
  • Li M, Ke QF, Tao SC, Guo SC, Rui BY, Guo YP. Fabrication of hydroxyapatite/chitosan composite hydrogels loaded with exosomes derived from miR-126-3p overexpressed synovial mesenchymal stem cells for diabetic chronic wound healing. J Mater Chem B. 2016;4(42):6830–6841. doi:10.1039/c6tb01560c
  • Meagher MJ, Weiss-Bilka HE, Best ME, Boerckel JD, Wagner DR, Roeder RK. Acellular hydroxyapatite-collagen scaffolds support angiogenesis and osteogenic gene expression in an ectopic murine model: effects of hydroxyapatite volume fraction. J Biomed Mater Res A. 2016;104(9):2178–2188. doi:10.1002/jbm.a.35760
  • Khezri K, Maleki Dizaj S, Rahbar Saadat Y, et al. Osteogenic differentiation of mesenchymal stem cells via curcumin-containing nanoscaffolds. Stem Cells Int. 2021;2021:1520052. doi:10.1155/2021/1520052
  • Ramazanli V, Ahmadov I. Synthesis of silver nanoparticles by using extract of olive leaves; 2022.