1932

Abstract

Chronic skin wounds are commonly found in older individuals who have impaired circulation due to diabetes or are immobilized due to physical disability. Chronic wounds pose a severe burden to the health-care system and are likely to become increasingly prevalent in aging populations. Various treatment approaches exist to help the healing process, although the healed tissue does not generally recapitulate intact skin but rather forms a scar that has inferior mechanical properties and that lacks appendages such as hair or sweat glands. This article describes new experimental avenues for attempting to improve the regenerative response of skin using biophysical techniques as well as biochemical methods, in some cases by trying to harness the potential of stem cells, either endogenous to the host or provided exogenously, to regenerate the skin. These approaches primarily address the local wound environment and should likely be combined with other modalities to address regional and systemic disease, as well as social determinants of health.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-bioeng-010220-113008
2022-06-06
2024-04-19
Loading full text...

Full text loading...

/deliver/fulltext/bioeng/24/1/annurev-bioeng-010220-113008.html?itemId=/content/journals/10.1146/annurev-bioeng-010220-113008&mimeType=html&fmt=ahah

Literature Cited

  1. 1.
    Sen CK. 2019. Human wounds and its burden: an updated compendium of estimates. Adv. Wound Care 8:39–48
    [Google Scholar]
  2. 2.
    Nussbaum SR, Carter MJ, Fife CE, DaVanzo J, Haught R et al. 2018. An economic evaluation of the impact, cost, and Medicare policy implications of chronic nonhealing wounds. Value Health 21:27–32
    [Google Scholar]
  3. 3.
    Eming SA, Martin P, Tomic-Canic M. 2014. Wound repair and regeneration: mechanisms, signaling, and translation. Sci. Transl. Med. 6:265sr6
    [Google Scholar]
  4. 4.
    Frykberg RG, Banks J. 2015. Challenges in the treatment of chronic wounds. Adv. Wound Care 4:560–82
    [Google Scholar]
  5. 5.
    Hetzler PT3rd, Dash BC, Guo S, Hsia HC. 2019. Targeting fibrotic signaling: a review of current literature and identification of future therapeutic targets to improve wound healing. Ann. Plast. Surg. 83:e92–95
    [Google Scholar]
  6. 6.
    Moore AL, Marshall CD, Barnes LA, Murphy MP, Ransom RC, Longaker MT 2018. Scarless wound healing: transitioning from fetal research to regenerative healing. Wiley Interdiscip. Rev. Dev. Biol. 7:e309
    [Google Scholar]
  7. 7.
    Marshall CD, Hu MS, Leavitt T, Barnes LA, Lorenz HP, Longaker MT. 2018. Cutaneous scarring: basic science, current treatments, and future directions. Adv. Wound Care 7:29–45
    [Google Scholar]
  8. 8.
    Correa-Gallegos D, Jiang D, Christ S, Ramesh P, Ye H et al. 2019. Patch repair of deep wounds by mobilized fascia. Nature 576:287–92
    [Google Scholar]
  9. 9.
    Mascharak S, desJardins-Park HE, Davitt MF, Griffin M, Borrelli MR et al. 2021. Preventing Engrailed-1 activation in fibroblasts yields wound regeneration without scarring. Science 372:eaba2374
    [Google Scholar]
  10. 10.
    Shook BA, Wasko RR, Rivera-Gonzalez GC, Salazar-Gatzimas E, Lopez-Giraldez F et al. 2018. Myofibroblast proliferation and heterogeneity are supported by macrophages during skin repair. Science 362:eaar2971
    [Google Scholar]
  11. 11.
    Haydont V, Bernard BA, Fortunel NO. 2019. Age-related evolutions of the dermis: clinical signs, fibroblast and extracellular matrix dynamics. Mech. Ageing Dev. 177:150–56
    [Google Scholar]
  12. 12.
    Phan QM, Fine GM, Salz L, Herrera GG, Wildman B et al. 2020. Lef1 expression in fibroblasts maintains developmental potential in adult skin to regenerate wounds. Elife 9:e60066
    [Google Scholar]
  13. 13.
    Rognoni E, Gomez C, Pisco AO, Rawlins EL, Simons BD et al. 2016. Inhibition of β-catenin signalling in dermal fibroblasts enhances hair follicle regeneration during wound healing. Development 143:2522–35
    [Google Scholar]
  14. 14.
    Wier EM, Garza LA. 2020. Through the lens of hair follicle neogenesis, a new focus on mechanisms of skin regeneration after wounding. Semin. Cell Dev. Biol. 100:122–29
    [Google Scholar]
  15. 15.
    Zakrzewski W, Dobrzynski M, Szymonowicz M, Rybak Z. 2019. Stem cells: past, present, and future. Stem Cell Res. Ther. 10:68
    [Google Scholar]
  16. 16.
    Jimenez F, Poblet E, Izeta A 2015. Reflections on how wound healing-promoting effects of the hair follicle can be translated into clinical practice. Exp. Dermatol. 24:91–94
    [Google Scholar]
  17. 17.
    Motegi SI, Ishikawa O. 2017. Mesenchymal stem cells: the roles and functions in cutaneous wound healing and tumor growth. J. Dermatol. Sci. 86:83–89
    [Google Scholar]
  18. 18.
    Zhu J, Huang H, Chen H, Zhang X, Li Z et al. 2018. Plerixafor and granulocyte-colony-stimulating factor for mobilization of hematopoietic stem cells for autologous transplantation in Chinese patients with non-Hodgkin's lymphoma: a randomized Phase 3 study. Transfusion 58:81–87
    [Google Scholar]
  19. 19.
    Guenou H, Nissan X, Larcher F, Feteira J, Lemaitre G et al. 2009. Human embryonic stem-cell derivatives for full reconstruction of the pluristratified epidermis: a preclinical study. Lancet 374:1745–53
    [Google Scholar]
  20. 20.
    de Miguel-Beriain I. 2015. The ethics of stem cells revisited. Adv. Drug Deliv. Rev. 82–83:176–80
    [Google Scholar]
  21. 21.
    Takahashi K, Yamanaka S. 2006. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:663–76
    [Google Scholar]
  22. 22.
    Dash BC, Korutla L, Vallabhajosyula P, Hsia HC. 2021. Unlocking the potential of induced pluripotent stem cells for wound healing: the next frontier of regenerative medicine. Adv. Wound Care In press. https://doi.org/10.1089/wound.2021.0049
    [Crossref] [Google Scholar]
  23. 23.
    Mandai M, Watanabe A, Kurimoto Y, Hirami Y, Morinaga C et al. 2017. Autologous induced stem-cell-derived retinal cells for macular degeneration. N. Engl. J. Med. 376:1038–46
    [Google Scholar]
  24. 24.
    Gorecka J, Kostiuk V, Fereydooni A, Gonzalez L, Luo J et al. 2019. The potential and limitations of induced pluripotent stem cells to achieve wound healing. Stem Cell Res. Ther. 10:87
    [Google Scholar]
  25. 25.
    Kosaric N, Kiwanuka H, Gurtner GC. 2019. Stem cell therapies for wound healing. Expert Opin. Biol. Ther. 19:575–85
    [Google Scholar]
  26. 26.
    Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F et al. 2006. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8:315–17
    [Google Scholar]
  27. 27.
    Kim I, Bang SI, Lee SK, Park SY, Kim M, Ha H 2014. Clinical implication of allogenic implantation of adipogenic differentiated adipose-derived stem cells. Stem Cells Transl. Med. 3:1312–21
    [Google Scholar]
  28. 28.
    Malhotra P, Shukla M, Meena P, Kakkar A, Khatri N et al. 2022. Mesenchymal stem cells are prospective novel off-the-shelf wound management tools. Drug Deliv. Transl. Res. 12:179–104
    [Google Scholar]
  29. 29.
    Dash BC, Xu Z, Lin L, Koo A, Ndon S et al. 2018. Stem cells and engineered scaffolds for regenerative wound healing. Bioengineering 5:23
    [Google Scholar]
  30. 30.
    Gorecka J, Gao X, Fereydooni A, Dash BC, Luo J et al. 2020. Induced pluripotent stem cell-derived smooth muscle cells increase angiogenesis and accelerate diabetic wound healing. Regen. Med. 15:1277–93
    [Google Scholar]
  31. 31.
    Chen D, Hou Q, Zhong L, Zhao Y, Li M, Fu X 2019. Bioactive molecules for skin repair and regeneration: progress and perspectives. Stem Cells Int. 2019:6789823
    [Google Scholar]
  32. 32.
    Zhou J, Sun J. 2019. A revolution in reprogramming: small molecules. Curr. Mol. Med. 19:77–90
    [Google Scholar]
  33. 33.
    Horinouchi CDS, Oostendorp C, Schade D, van Kuppevelt TH, Daamen WF. 2021. Growth factor mimetics for skin regeneration: in vitro profiling of primary human fibroblasts and keratinocytes. Arch. Pharm. 354:e2100082
    [Google Scholar]
  34. 34.
    Yuan ZD, Zhu WN, Liu KZ, Huang ZP, Han YC. 2020. Small molecule epigenetic modulators in pure chemical cell fate conversion. Stem Cells Int. 2020:8890917
    [Google Scholar]
  35. 35.
    Williams IM, Wu JC. 2019. Generation of endothelial cells from human pluripotent stem cells. Arterioscler. Thromb. Vasc. Biol. 39:1317–29
    [Google Scholar]
  36. 36.
    Xu M, He J, Zhang C, Xu J, Wang Y. 2019. Strategies for derivation of endothelial lineages from human stem cells. Stem Cell Res. Ther. 10:200
    [Google Scholar]
  37. 37.
    Lee S, Park C, Han JW, Kim JY, Cho K et al. 2017. Direct reprogramming of human dermal fibroblasts into endothelial cells using ER71/ETV2. Circ. Res. 120:848–61
    [Google Scholar]
  38. 38.
    Wary A, Wary N, Baruah J, Mastej V, Wary KK 2017. Chromatin-modifying agents convert fibroblasts to OCT4+ and VEGFR-2+ capillary tube-forming cells. PLOS ONE 12:e0176496
    [Google Scholar]
  39. 39.
    Song W, Kaufman DS, Shen W. 2016. Efficient generation of endothelial cells from human pluripotent stem cells and characterization of their functional properties. J. Biomed. Mater. Res. A 104:678–87
    [Google Scholar]
  40. 40.
    Tao R, Sun TJ, Han YQ, Xu G, Liu J, Han YF. 2014. Epimorphin-induced differentiation of human umbilical cord mesenchymal stem cells into sweat gland cells. Eur. Rev. Med. Pharmacol. Sci. 18:1404–10
    [Google Scholar]
  41. 41.
    Xu Y, Hong Y, Xu M, Ma K, Fu X et al. 2016. Role of keratinocyte growth factor in the differentiation of sweat gland-like cells from human umbilical cord-derived mesenchymal stem cells. Stem Cells Transl. Med. 5:106–16
    [Google Scholar]
  42. 42.
    Yang WS, Kang S, Sung J, Kleinman HK 2019. Thymosin β4: potential to treat epidermolysis bullosa and other severe dermal injuries. Eur. J. Dermatol. 29:459–67
    [Google Scholar]
  43. 43.
    Al Haj Zen A, Nawrot DA, Howarth A, Caporali A, Ebner D et al. 2016. The retinoid agonist tazarotene promotes angiogenesis and wound healing. Mol. Ther. 24:1745–59
    [Google Scholar]
  44. 44.
    Gao SQ, Chang C, Li JJ, Li Y, Niu XQ et al. 2018. Co-delivery of deferoxamine and hydroxysafflor yellow A to accelerate diabetic wound healing via enhanced angiogenesis. Drug Deliv. 25:1779–89
    [Google Scholar]
  45. 45.
    Guenat OT, Geiser T, Berthiaume F. 2020. Clinically relevant tissue scale responses as new readouts from organs-on-a-chip for precision medicine. Annu. Rev. Anal. Chem. 13:111–33
    [Google Scholar]
  46. 46.
    Huang C, Leavitt T, Bayer LR, Orgill DP. 2014. Effect of negative pressure wound therapy on wound healing. Curr. Probl. Surg. 51:301–31
    [Google Scholar]
  47. 47.
    Baldwin C, Potter M, Clayton E, Irvine L, Dye J 2009. Topical negative pressure stimulates endothelial migration and proliferation: a suggested mechanism for improved integration of Integra. Ann. Plast. Surg. 62:92–96
    [Google Scholar]
  48. 48.
    Argenta LC, Morykwas MJ. 1997. Vacuum-assisted closure: a new method for wound control and treatment: clinical experience. Ann. Plast. Surg. 38:563–77
    [Google Scholar]
  49. 49.
    Zhang L, Weng T, Wu P, Li Q, Han C, Wang X 2020. The combined use of negative-pressure wound therapy and dermal substitutes for tissue repair and regeneration. Biomed. Res. Int. 2020:8824737
    [Google Scholar]
  50. 50.
    Thakral G, Lafontaine J, Najafi B, Talal TK, Kim P, Lavery LA 2013. Electrical stimulation to accelerate wound healing. Diabet Foot Ankle 4::10.3402/dfa.v4i0.22081
    [Google Scholar]
  51. 51.
    Rajendran SB, Challen K, Wright KL, Hardy JG 2021. Electrical stimulation to enhance wound healing. J. Funct. Biomater. 12:40
    [Google Scholar]
  52. 52.
    Lv H, Liu J, Zhen C, Wang Y, Wei Y et al. 2021. Magnetic fields as a potential therapy for diabetic wounds based on animal experiments and clinical trials. Cell Prolif. 54:e12982
    [Google Scholar]
  53. 53.
    Long Y, Wei H, Li J, Yao G, Yu B et al. 2018. Effective wound healing enabled by discrete alternative electric fields from wearable nanogenerators. ACS Nano 12:12533–40
    [Google Scholar]
  54. 54.
    Yao G, Jiang D, Li J, Kang L, Chen S et al. 2019. Self-activated electrical stimulation for effective hair regeneration via a wearable omnidirectional pulse generator. ACS Nano 13:12345–56
    [Google Scholar]
  55. 55.
    Rubin AE, Usta OB, Schloss R, Yarmush M, Golberg A 2019. Selective inactivation of Pseudomonas aeruginosa and Staphylococcus epidermidis with pulsed electric fields and antibiotics. Adv. Wound Care 8:136–48
    [Google Scholar]
  56. 56.
    Das B, Berthiaume F. 2021. Irreversible electroporation as an alternative to wound debridement surgery. Surg. Technol. Int. 39:sti39–1452
    [Google Scholar]
  57. 57.
    Das B, Shrirao A, Golberg A, Berthiaume F, Schloss R, Yarmush ML. 2020. Differential cell death and regrowth of dermal fibroblasts and keratinocytes after application of pulsed electric fields. Bioelectricity 2:175–85
    [Google Scholar]
  58. 58.
    Li X, Saeidi N, Villiger M, Albadawi H, Jones JD et al. 2018. Rejuvenation of aged rat skin with pulsed electric fields. J. Tissue Eng. Regen. Med. 12:2309–18
    [Google Scholar]
  59. 59.
    Yannas IV, Tzeranis DS. 2021. Mammals fail to regenerate organs when wound contraction drives scar formation. NPJ Regen. Med. 6:39
    [Google Scholar]
  60. 60.
    Tabares FL, Junkar I. 2021. Cold plasma systems and their application in surface treatments for medicine. Molecules 26:1903
    [Google Scholar]
  61. 61.
    Gan L, Jiang J, Duan JW, Wu XJZ, Zhang S et al. 2021. Cold atmospheric plasma applications in dermatology: a systematic review. J. Biophotonics 14:e202000415
    [Google Scholar]
  62. 62.
    Wen X, Xin Y, Hamblin MR, Jiang X. 2021. Applications of cold atmospheric plasma for transdermal drug delivery: a review. Drug Deliv. Transl. Res. 11:741–47
    [Google Scholar]
  63. 63.
    Bourke P, Ziuzina D, Han L, Cullen PJ, Gilmore BF. 2017. Microbiological interactions with cold plasma. J. Appl. Microbiol. 123:308–24
    [Google Scholar]
  64. 64.
    Arndt S, Unger P, Berneburg M, Bosserhoff AK, Karrer S. 2018. Cold atmospheric plasma (CAP) activates angiogenesis-related molecules in skin keratinocytes, fibroblasts and endothelial cells and improves wound angiogenesis in an autocrine and paracrine mode. J. Dermatol. Sci. 89:181–90
    [Google Scholar]
  65. 65.
    Schmidt A, Liebelt G, Niessner F, von Woedtke T, Bekeschus S. 2021. Gas plasma-spurred wound healing is accompanied by regulation of focal adhesion, matrix remodeling, and tissue oxygenation. Redox Biol. 38:101809
    [Google Scholar]
  66. 66.
    Gorbanev Y, Soriano R, O'Connell D, Chechik V. 2016. An atmospheric pressure plasma setup to investigate the reactive species formation. J. Vis. Exp. 117:54765
    [Google Scholar]
  67. 67.
    Mohamed H, Gebski E, Reyes R, Beane S, Wigdahl B et al. 2021. Differential effect of non-thermal plasma RONS on two human leukemic cell populations. Cancers 13:2437
    [Google Scholar]
  68. 68.
    Xie J, Chen Q, Suresh P, Roy S, White JF, Mazzeo AD. 2017. Paper-based plasma sanitizers. PNAS 114:5119–24
    [Google Scholar]
  69. 69.
    Larouche J, Sheoran S, Maruyama K, Martino MM 2018. Immune regulation of skin wound healing: mechanisms and novel therapeutic targets. Adv. Wound Care 7:7209–31
    [Google Scholar]
  70. 70.
    Menon R, Krzyszczyk P, Berthiaume F. 2017. Pro-resolution potency of resolvins D1, D2 and E1 on neutrophil migration and in dermal wound healing. Nano Life 7:11750002
    [Google Scholar]
  71. 71.
    Krzyszczyk P, Schloss R, Palmer A, Berthiaume F 2018. The role of macrophages in acute and chronic wound healing and interventions to promote pro-wound healing phenotypes. Front. Physiol. 9:419
    [Google Scholar]
  72. 72.
    Olekson MP, Faulknor RA, Hsia HC, Schmidt AM, Berthiaume F. 2016. Soluble receptor for advanced glycation end products improves stromal cell-derived factor-1 activity in model diabetic environments. Adv. Wound Care 5:527–38
    [Google Scholar]
  73. 73.
    Nurkesh A, Jaguparov A, Jimi S, Saparov A 2020. Recent advances in the controlled release of growth factors and cytokines for improving cutaneous wound healing. Front. Cell Dev. Biol. 8:638
    [Google Scholar]
  74. 74.
    Blume P, Bowlby M, Schmidt B, Donegan R. 2014. Safety and efficacy of Becaplermin gel in the treatment of diabetic foot ulcers. Chronic Wound Care Manag. Res. 1:11–14
    [Google Scholar]
  75. 75.
    Kang HJ, Kumar S, D'Elia A, Dash B, Nanda V et al. 2021. Self-assembled elastin-like polypeptide fusion protein coacervates as competitive inhibitors of advanced glycation end-products enhance diabetic wound healing. J. Control Release 333:176–87
    [Google Scholar]
  76. 76.
    Jimi S, Jaguparov A, Nurkesh A, Sultankulov B, Saparov A. 2020. Sequential delivery of cryogel released growth factors and cytokines accelerates wound healing and improves tissue regeneration. Front. Bioeng. Biotechnol. 8:345
    [Google Scholar]
  77. 77.
    Lombardi F, Palumbo P, Augello FR, Cifone MG, Cinque B, Giuliani M 2019. Secretome of adipose tissue-derived stem cells (ASCs) as a novel trend in chronic non-healing wounds: an overview of experimental in vitro and in vivo studies and methodological variables. Int. J. Mol. Sci. 20:3721
    [Google Scholar]
  78. 78.
    Xu J, Liu X, Zhao F, Zhang Y, Wang Z. 2020. HIF1α overexpression enhances diabetic wound closure in high glucose and low oxygen conditions by promoting adipose-derived stem cell paracrine function and survival. Stem Cell Res. Ther. 11:148
    [Google Scholar]
  79. 79.
    An YH, Kim DH, Lee EJ, Lee D, Park MJ et al. 2021. High-efficient production of adipose-derived stem cell (ADSC) secretome through maturation process and its non-scarring wound healing applications. Front. Bioeng. Biotechnol. 9:681501
    [Google Scholar]
  80. 80.
    Cai Y, Li J, Jia C, He Y, Deng C. 2020. Therapeutic applications of adipose cell-free derivatives: a review. Stem Cell Res. Ther. 11:312
    [Google Scholar]
  81. 81.
    Kim MH, Wu WH, Choi JH, Kim J, Jun JH et al. 2018. Galectin-1 from conditioned medium of three-dimensional culture of adipose-derived stem cells accelerates migration and proliferation of human keratinocytes and fibroblasts. Wound Repair Regen. 26:Suppl. 1S9–18
    [Google Scholar]
  82. 82.
    Lou P, Liu S, Xu X, Pan C, Lu Y, Liu J 2021. Extracellular vesicle-based therapeutics for the regeneration of chronic wounds: current knowledge and future perspectives. Acta Biomater. 119:42–56
    [Google Scholar]
  83. 83.
    French KC, Antonyak MA, Cerione RA. 2017. Extracellular vesicle docking at the cellular port: extracellular vesicle binding and uptake. Semin. Cell Dev. Biol. 67:48–55
    [Google Scholar]
  84. 84.
    Zhang B, Wang M, Gong A, Zhang X, Wu X et al. 2015. HucMSC-exosome mediated-Wnt4 signaling is required for cutaneous wound healing. Stem Cells 33:2158–68
    [Google Scholar]
  85. 85.
    Hahm J, Kim J, Park J 2021. Strategies to enhance extracellular vesicle production. Tissue Eng. Regen. Med. 18:513–24
    [Google Scholar]
  86. 86.
    Patel DB, Santoro M, Born LJ, Fisher JP, Jay SM. 2018. Towards rationally designed biomanufacturing of therapeutic extracellular vesicles: impact of the bioproduction microenvironment. Biotechnol. Adv. 36:2051–59
    [Google Scholar]
  87. 87.
    Grangier A, Branchu J, Volatron J, Piffoux M, Gazeau F et al. 2021. Technological advances towards extracellular vesicles mass production. Adv. Drug Deliv. Rev. 176:113843
    [Google Scholar]
  88. 88.
    Gurunathan S, Kang MH, Jeyaraj M, Qasim M, Kim JH. 2019. Review of the isolation, characterization, biological function, and multifarious therapeutic approaches of exosomes. Cells 8:307
    [Google Scholar]
  89. 89.
    Yuan F, Li YM, Wang Z. 2021. Preserving extracellular vesicles for biomedical applications: consideration of storage stability before and after isolation. Drug Deliv. 28:1501–9
    [Google Scholar]
  90. 90.
    Trenkenschuh E, Richter M, Heinrich E, Koch M, Fuhrmann G, Friess W 2022. Enhancing the stabilization potential of lyophilization for extracellular vesicles. Adv. Healthc. Mater. 11:e2100538
    [Google Scholar]
  91. 91.
    An Y, Lin S, Tan X, Zhu S, Nie F et al. 2021. Exosomes from adipose-derived stem cells and application to skin wound healing. Cell Prolif. 54:e12993
    [Google Scholar]
  92. 92.
    Hu Y, Tao R, Chen L, Xiong Y, Xue H et al. 2021. Exosomes derived from pioglitazone-pretreated MSCs accelerate diabetic wound healing through enhancing angiogenesis. J. Nanobiotechnol. 19:150
    [Google Scholar]
  93. 93.
    Chen L, Qin L, Chen C, Hu Q, Wang J, Shen J 2021. Serum exosomes accelerate diabetic wound healing by promoting angiogenesis and ECM formation. Cell Biol. Int. 45:1976–85
    [Google Scholar]
  94. 94.
    Yin H, Chen CY, Liu YW, Tan YJ, Deng ZL et al. 2019. Synechococcus elongatus PCC7942 secretes extracellular vesicles to accelerate cutaneous wound healing by promoting angiogenesis. Theranostics 9:2678–93
    [Google Scholar]
  95. 95.
    Li B, Luan S, Chen J, Zhou Y, Wang T et al. 2020. The MSC-derived exosomal lncRNA H19 promotes wound healing in diabetic foot ulcers by upregulating PTEN via microRNA-152–3p. Mol. Ther. Nucleic Acids 19:814–26
    [Google Scholar]
  96. 96.
    Li X, Xie X, Lian W, Shi R, Han S et al. 2018. 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. 50:1–14
    [Google Scholar]
  97. 97.
    Elsharkasy OM, Nordin JZ, Hagey DW, de Jong OG, Schiffelers RM et al. 2020. Extracellular vesicles as drug delivery systems: why and how?. Adv. Drug Deliv. Rev. 159:332–43
    [Google Scholar]
  98. 98.
    Sutaria DS, Badawi M, Phelps MA, Schmittgen TD. 2017. Achieving the promise of therapeutic extracellular vesicles: The devil is in details of therapeutic loading. Pharm. Res. 34:1053–66
    [Google Scholar]
  99. 99.
    Lv Q, Deng J, Chen Y, Wang Y, Liu B, Liu J. 2020. Engineered human adipose stem-cell-derived exosomes loaded with miR-21-5p to promote diabetic cutaneous wound healing. Mol. Pharm. 17:1723–33
    [Google Scholar]
  100. 100.
    Berger AG, Chou JJ, Hammond PT. 2021. Approaches to modulate the chronic wound environment using localized nucleic acid delivery. Adv. Wound Care 10:503–28
    [Google Scholar]
  101. 101.
    Herter EK, Xu Landén N. 2017. Non-coding RNAs: new players in skin wound healing. Adv. Wound Care 6:93–107
    [Google Scholar]
  102. 102.
    Picanco-Castro V, Pereira CG, Covas DT, Porto GS, Athanassiadou A, Figueiredo ML 2020. Emerging patent landscape for non-viral vectors used for gene therapy. Nat. Biotechnol. 38:151–57
    [Google Scholar]
  103. 103.
    Caley MP, Martins VL, O'Toole EA. 2015. Metalloproteinases and wound healing. Adv. Wound Care 4:225–34
    [Google Scholar]
  104. 104.
    Castleberry SA, Almquist BD, Li W, Reis T, Chow J et al. 2016. Self-assembled wound dressings silence MMP-9 and improve diabetic wound healing in vivo. Adv. Mater. 28:1809–17
    [Google Scholar]
  105. 105.
    Li N, Yang L, Pan C, Saw PE, Ren M et al. 2020. Naturally-occurring bacterial cellulose-hyperbranched cationic polysaccharide derivative/MMP-9 siRNA composite dressing for wound healing enhancement in diabetic rats. Acta Biomater. 102:298–314
    [Google Scholar]
  106. 106.
    Kim HS, Yoo HS. 2013. Matrix metalloproteinase-inspired suicidal treatments of diabetic ulcers with siRNA-decorated nanofibrous meshes. Gene Ther. 20:378–85
    [Google Scholar]
  107. 107.
    Kasiewicz LN, Whitehead KA. 2019. Lipid nanoparticles silence tumor necrosis factor α to improve wound healing in diabetic mice. Bioeng. Transl. Med. 4:75–82
    [Google Scholar]
  108. 108.
    Tezgel Ö, DiStasio N, Laghezza-Masci V, Taddei A-R, Szarpak-Jankowska A et al. 2020. Collagen scaffold-mediated delivery of NLC/siRNA as wound healing materials. J. Drug Deliv. Sci. Technol. 55:101421
    [Google Scholar]
  109. 109.
    Ban E, Jeong S, Park M, Kwon H, Park J et al. 2020. Accelerated wound healing in diabetic mice by miRNA-497 and its anti-inflammatory activity. Biomed. Pharmacother. 121:109613
    [Google Scholar]
  110. 110.
    Sener G, Hilton SA, Osmond MJ, Zgheib C, Newsom JP et al. 2020. Injectable, self-healable zwitterionic cryogels with sustained microRNA–cerium oxide nanoparticle release promote accelerated wound healing. Acta Biomater. 101:262–72
    [Google Scholar]
  111. 111.
    Saleh B, Dhaliwal HK, Portillo-Lara R, Shirzaei Sani E, Abdi R et al. 2019. Local immunomodulation using an adhesive hydrogel loaded with miRNA-laden nanoparticles promotes wound healing. Small 15:e1902232
    [Google Scholar]
  112. 112.
    Ozdemir D, Feinberg MW. 2019. MicroRNAs in diabetic wound healing: pathophysiology and therapeutic opportunities. Trends Cardiovasc. Med. 29:131–37
    [Google Scholar]
  113. 113.
    Mulholland EJ, Dunne N, McCarthy HO 2017. MicroRNA as therapeutic targets for chronic wound healing. Mol. Ther. Nucleic Acids 8:46–55
    [Google Scholar]
  114. 114.
    Icli B, Wu W, Ozdemir D, Li H, Cheng HS et al. 2019. MicroRNA-615-5p regulates angiogenesis and tissue repair by targeting AKT/eNOS (protein kinase B/endothelial nitric oxide synthase) signaling in endothelial cells. Arterioscler. Thromb. Vasc. Biol. 39:1458–74
    [Google Scholar]
  115. 115.
    Icli B, Wu W, Ozdemir D, Li H, Haemmig S et al. 2019. MicroRNA-135a-3p regulates angiogenesis and tissue repair by targeting p38 signaling in endothelial cells. FASEB J. 33:5599–614
    [Google Scholar]
  116. 116.
    Gallant-Behm CL, Piper J, Dickinson BA, Dalby CM, Pestano LA, Jackson AL. 2018. A synthetic microRNA-92a inhibitor (MRG-110) accelerates angiogenesis and wound healing in diabetic and nondiabetic wounds. Wound Repair Regen. 26:311–23
    [Google Scholar]
  117. 117.
    Lucas T, Schafer F, Muller P, Eming SA, Heckel A, Dimmeler S. 2017. Light-inducible antimiR-92a as a therapeutic strategy to promote skin repair in healing-impaired diabetic mice. Nat. Commun. 8:15162
    [Google Scholar]
  118. 118.
    Shi R, Lian W, Han S, Cao C, Jin Y et al. 2018. Nanosphere-mediated co-delivery of VEGF-A and PDGF-B genes for accelerating diabetic foot ulcers healing in rats. Gene Ther. 25:425–38
    [Google Scholar]
  119. 119.
    Wang S, Yan C, Zhang X, Shi D, Chi L et al. 2018. Antimicrobial peptide modification enhances the gene delivery and bactericidal efficiency of gold nanoparticles for accelerating diabetic wound healing. Biomater. Sci. 6:2757–72
    [Google Scholar]
  120. 120.
    Mo Y, Guo R, Zhang Y, Xue W, Cheng B, Zhang Y. 2017. Controlled dual delivery of angiogenin and curcumin by electrospun nanofibers for skin regeneration. Tissue Eng. Part A 23:597–608
    [Google Scholar]
  121. 121.
    Sun N, Ning B, Hansson KM, Bruce AC, Seaman SA et al. 2018. Modified VEGF-A mRNA induces sustained multifaceted microvascular response and accelerates diabetic wound healing. Sci. Rep. 8:17509
    [Google Scholar]
  122. 122.
    Thiersch M, Rimann M, Panagiotopoulou V, Ozturk E, Biedermann T et al. 2013. The angiogenic response to PLL-g-PEG-mediated HIF-1α plasmid DNA delivery in healthy and diabetic rats. Biomaterials 34:4173–82
    [Google Scholar]
  123. 123.
    Martin JR, Nelson CE, Gupta MK, Yu F, Sarett SM et al. 2016. Local delivery of PHD2 siRNA from ROS-degradable scaffolds to promote diabetic wound healing. Adv. Healthc. Mater. 5:2751–57
    [Google Scholar]
  124. 124.
    Rabbani PS, Zhou A, Borab ZM, Frezzo JA, Srivastava N et al. 2017. Novel lipoproteoplex delivers Keap1 siRNA based gene therapy to accelerate diabetic wound healing. Biomaterials 132:1–15
    [Google Scholar]
  125. 125.
    Kobsa S, Kristofik NJ, Sawyer AJ, Bothwell AL, Kyriakides TR, Saltzman WM. 2013. An electrospun scaffold integrating nucleic acid delivery for treatment of full-thickness wounds. Biomaterials 34:3891–901
    [Google Scholar]
  126. 126.
    Ghatak S, Li J, Chan YC, Gnyawali SC, Steen E et al. 2016. AntihypoxamiR functionalized gramicidin lipid nanoparticles rescue against ischemic memory improving cutaneous wound healing. Nanomedicine 12:1827–31
    [Google Scholar]
  127. 127.
    Gilmartin DJ, Soon A, Thrasivoulou C, Phillips AR, Jayasinghe SN, Becker DL. 2016. Sustained release of Cx43 antisense oligodeoxynucleotides from coated collagen scaffolds promotes wound healing. Adv. Healthc. Mater. 5:1786–99
    [Google Scholar]
  128. 128.
    Li H, Chang L, Du WW, Gupta S, Khorshidi A et al. 2014. Anti-microRNA-378a enhances wound healing process by upregulating integrin beta-3 and vimentin. Mol. Ther. 22:1839–50
    [Google Scholar]
  129. 129.
    Miscianinov V, Martello A, Rose L, Parish E, Cathcart B et al. 2018. MicroRNA-148b targets the TGF-β pathway to regulate angiogenesis and endothelial-to-mesenchymal transition during skin wound healing. Mol. Ther. 26:1996–2007
    [Google Scholar]
  130. 130.
    Li J, Wu C, Wang W, He Y, Elkayam E et al. 2018. Structurally modulated codelivery of siRNA and Argonaute 2 for enhanced RNA interference. PNAS 115:E2696–705
    [Google Scholar]
  131. 131.
    Shah JB. 2011. The history of wound care. J. Am. Col. Certif. Wound Spec. 3:65–66
    [Google Scholar]
  132. 132.
    Zhang X, Shu W, Yu Q, Qu W, Wang Y, Li R 2020. Functional biomaterials for treatment of chronic wound. Front. Bioeng. Biotechnol. 8:516
    [Google Scholar]
  133. 133.
    Hosseini M, Shafiee A. 2021. Engineering bioactive scaffolds for skin regeneration. Small 17:e2101384
    [Google Scholar]
  134. 134.
    Nikolova MP, Chavali MS. 2019. Recent advances in biomaterials for 3D scaffolds: a review. Bioact. Mater. 4:271–92
    [Google Scholar]
  135. 135.
    Liang Y, Zhao X, Hu T, Chen B, Yin Z et al. 2019. Adhesive hemostatic conducting injectable composite hydrogels with sustained drug release and photothermal antibacterial activity to promote full-thickness skin regeneration during wound healing. Small 15:e1900046
    [Google Scholar]
  136. 136.
    Rahmani Del Bakhshayesh A, Mostafavi E, Alizadeh E, Asadi N, Akbarzadeh A, Davaran S 2018. Fabrication of three-dimensional scaffolds based on nano-biomimetic collagen hybrid constructs for skin tissue engineering. ACS Omega 3:8605–11
    [Google Scholar]
  137. 137.
    Wong VW, Rustad KC, Galvez MG, Neofytou E, Glotzbach JP et al. 2011. Engineered pullulan-collagen composite dermal hydrogels improve early cutaneous wound healing. Tissue Eng. Part A 17:631–44
    [Google Scholar]
  138. 138.
    Griffin DR, Archang MM, Kuan CH, Weaver WM, Weinstein JS et al. 2021. Activating an adaptive immune response from a hydrogel scaffold imparts regenerative wound healing. Nat. Mater. 20:560–69
    [Google Scholar]
  139. 139.
    Assi R, Foster TR, He H, Stamati K, Bai H et al. 2016. Delivery of mesenchymal stem cells in biomimetic engineered scaffolds promotes healing of diabetic ulcers. Regen. Med. 11:245–60
    [Google Scholar]
  140. 140.
    Bhowmick S, Thanusha AV, Kumar A, Scharnweber D, Rother S, Koul V 2018. Nanofibrous artificial skin substitute composed of mPEG–PCL grafted gelatin/hyaluronan/chondroitin sulfate/sericin for 2nd degree burn care: in vitro and in vivo study. RSC Adv. 8:16420–32
    [Google Scholar]
  141. 141.
    Farokhi M, Mottaghitalab F, Fatahi Y, Khademhosseini A, Kaplan DL 2018. Overview of silk fibroin use in wound dressings. Trends Biotechnol. 36:907–22
    [Google Scholar]
  142. 142.
    Jin G, Prabhakaran MP, Ramakrishna S. 2014. Photosensitive and biomimetic core-shell nanofibrous scaffolds as wound dressing. Photochem. Photobiol. 90:673–81
    [Google Scholar]
  143. 143.
    Ozpur MA, Guneren E, Canter HI, Karaaltin MV, Ovali E et al. 2016. Generation of skin tissue using adipose tissue-derived stem cells. Plast. Reconstr. Surg. 137:134–43
    [Google Scholar]
  144. 144.
    Bahrami H, Keshel SH, Chari AJ, Biazar E. 2016. Human unrestricted somatic stem cells loaded in nanofibrous PCL scaffold and their healing effect on skin defects. Artif. Cells Nanomed. Biotechnol. 44:1556–60
    [Google Scholar]
  145. 145.
    Griffin DR, Weaver WM, Scumpia PO, Di Carlo D, Segura T. 2015. Accelerated wound healing by injectable microporous gel scaffolds assembled from annealed building blocks. Nat. Mater. 14:737–44
    [Google Scholar]
  146. 146.
    Lima AC, Mano JF, Concheiro A, Alvarez-Lorenzo C. 2015. Fast and mild strategy, using superhydrophobic surfaces, to produce collagen/platelet lysate gel beads for skin regeneration. Stem Cell Rev. Rep. 11:161–79
    [Google Scholar]
  147. 147.
    Kim BS, Kwon YW, Kong JS, Park GT, Gao G et al. 2018. 3D cell printing of in vitro stabilized skin model and in vivo pre-vascularized skin patch using tissue-specific extracellular matrix bioink: a step towards advanced skin tissue engineering. Biomaterials 168:38–53
    [Google Scholar]
  148. 148.
    Skardal A, Mack D, Kapetanovic E, Atala A, Jackson JD et al. 2012. Bioprinted amniotic fluid-derived stem cells accelerate healing of large skin wounds. Stem Cells Transl. Med. 1:792–802
    [Google Scholar]
  149. 149.
    Masri S, Fauzi MB. 2021. Current insight of printability quality improvement strategies in natural-based bioinks for skin regeneration and wound healing. Polymers 13:1011
    [Google Scholar]
  150. 150.
    Vojtassak J, Danisovic L, Kubes M, Bakos D, Jarabek L et al. 2006. Autologous biograft and mesenchymal stem cells in treatment of the diabetic foot. Neuro Endocrinol. Lett. 27:Suppl. 2134–37
    [Google Scholar]
  151. 151.
    Kosol W, Kumar S, Marrero-Berrĺos I, Berthiaume F. 2020. Medium conditioned by human mesenchymal stromal cells reverses low serum and hypoxia-induced inhibition of wound closure. Biochem. Biophys. Res. Commun. 522:335–41
    [Google Scholar]
  152. 152.
    Faulknor RA, Olekson MA, Ekwueme EC, Krzyszczyk P, Freeman JW, Berthiaume F 2017. Hypoxia impairs mesenchymal stromal cell-induced macrophage M1 to M2 transition. Technology 5:81–86
    [Google Scholar]
  153. 153.
    Kleinman DV, Pronk N, Gomez CA, Wrenn Gordon GL, Ochiai E et al. 2021. Addressing health equity and social determinants of health through Healthy People 2030. J. Public Health Manag. Pract. 27:Suppl. 6S249–57
    [Google Scholar]
  154. 154.
    Lucyk K, McLaren L. 2017. Taking stock of the social determinants of health: a scoping review. PLOS ONE 12:e0177306
    [Google Scholar]
  155. 155.
    Sasson DC, Duan K, Patel SM, Junn A, Hsia HC 2021. The impact of social determinants of health on pressure ulcer prognosis: a retrospective chart and systematic review Poster presented at the 38th Annual Meeting of the Northeastern Society of Plastic Surgeons Philadelphia:
/content/journals/10.1146/annurev-bioeng-010220-113008
Loading
/content/journals/10.1146/annurev-bioeng-010220-113008
Loading

Data & Media loading...

Supplemental Material

Supplementary Data

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error