Review Article

用于慢性伤口愈合的细胞疗法:再生医学的未来

卷 23, 期 16, 2022

发表于: 20 October, 2022

页: [1489 - 1504] 页: 16

弟呕挨: 10.2174/138945012309220623144620

价格: $65

摘要

慢性创伤与显著的发病率和死亡率有关,需要长期有效的治疗,并对全球卫生保健系统造成巨大的财政压力。用干细胞的再生药物最近已成为一种有前途的方法,并且是一个活跃的研究领域,它们有可能分化成特定类型的细胞,从而具有自我更新,再生和免疫调节作用。此外,随着技术的兴起,各种细胞疗法和细胞类型,如骨髓和脂肪来源的间充质细胞(ADMSC),内皮祖细胞(EPC),胚胎干细胞(ESC),间充质干细胞(MSCs)和多能干细胞(PSC)被研究其对修复过程和组织再生的治疗影响。细胞疗法已被证明对提高皮肤再生和伤口修复的质量和速度具有实质性的控制作用。文献综述揭示了伤口愈合的机制,导致慢性伤口的异常以及伤口护理研究人员面临的障碍,从而探索了众多潜在改进的机会。此外,本综述的重点是在临床试验的背景下提供有关可能的细胞衍生治疗选择及其在愈合方面的相关挑战的详细信息,因为这些挑战的解决方案将为改进研究设计提供新的和更好的未来机会,从而为开发更专业的治疗方法提供大量数据。

关键词: 细胞疗法,细胞功能障碍,慢性创伤,创伤愈合,干细胞,再生医学

图形摘要
[1]
Dong R, Guo B. Smart wound dressings for wound healing. Nano Today 2021; 1(41): 101290.
[http://dx.doi.org/10.1016/j.nantod.2021.101290]
[2]
Lloyd G, Friedman G, Jafri S, Schultz G, Fridman A, Harding K. Gas plasma: Medical uses and developments in wound care. Plasma Process Polym 2010; 7(3-4): 194-211.
[http://dx.doi.org/10.1002/ppap.200900097]
[3]
Chandan K. Human wounds and its burden: An updated compendium of estimates. Adv Wound Care (New Rochelle) 2019; 8(2): 39-48.
[http://dx.doi.org/10.1089/wound.2019.0946]
[4]
Centres for disease control and prevention. New CDC report: More than 100 million have diabetes or prediabetes. 2018. Available from: https://www.cdc.gov/media/releases/2017/p0718-diabetes-report.html
[5]
World Health Organization. Available from: https://www.who.int/topics/injuries/en/ (Accessed on December 12, 2020.)
[6]
Li Y, Ye Z, Yang W, Zhang Q, Zeng J. An update on the potential of mesenchymal stem cell therapy for cutaneous diseases. Stem Cells Int 2021; 2021: 8834590.
[http://dx.doi.org/10.1155/2021/8834590] [PMID: 33505474]
[7]
Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: Mechanisms, signaling, and translation. Sci Transl Med 2014; 6(265): 265sr6.
[http://dx.doi.org/10.1126/scitranslmed.3009337] [PMID: 25473038]
[8]
Naghibi M, Smith RP, Baltch AL, et al. The effect of diabetes mellitus on chemotactic and bactericidal activity of human polymorphonuclear leukocytes. Diabetes Res Clin Pract 1987; 4(1): 27-35.
[http://dx.doi.org/10.1016/S0168-8227(87)80030-X] [PMID: 3121272]
[9]
Logan RM, Al-Azri AR, Bossi P, et al. Systematic review of growth factors and cytokines for the management of oral mucositis in cancer patients and clinical practice guidelines. Support Care Cancer 2020; 28(5): 2485-98.
[http://dx.doi.org/10.1007/s00520-019-05170-9] [PMID: 32080767]
[10]
Dar TM, Samin KA. Role of growth factors-rich plasma, gel and membrane in dermal wound healing and injured tissue restoration and regeneration. Eur J Med Health Sci 2021; 3(6): 14-23.
[http://dx.doi.org/10.24018/ejmed.2021.3.6.1105]
[11]
Larouche J, Sheoran S, Maruyama K, Martino MM. Immune regulation of skin wound healing: Mechanisms and novel therapeutic targets. Adv Wound Care (New Rochelle) 2018; 7(7): 209-31.
[http://dx.doi.org/10.1089/wound.2017.0761] [PMID: 29984112]
[12]
Dorantes LC. Cañedo-Ayala, Skin acute wound healing. Int J Inflamm 2019; 2090-8040.
[http://dx.doi.org/10.1155/2019/3706315]
[13]
Cappuzzello C, Doni A, Dander E, et al. Mesenchymal stromal cell-derived PTX3 promotes wound healing via fibrin remodeling. J Invest Dermatol 2016; 136(1): 293-300.
[http://dx.doi.org/10.1038/JID.2015.346] [PMID: 26763449]
[14]
Liu Y, Holmes C. Tissue regeneration capacity of extracellular vesicles isolated from bone marrow-derived and adipose-derived mesenchymal stromal/stem cells. Frontier Cell Develop Biol 2021; 26: 308.
[http://dx.doi.org/10.3389/fcell.2021.648098]
[15]
Wu Y, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells 2007; 25(10): 2648-59.
[http://dx.doi.org/10.1634/stemcells.2007-0226] [PMID: 17615264]
[16]
Wang CG, Lou YT, Tong MJ, et al. Asperosaponin VI promotes angiogenesis and accelerates wound healing in rats via up-regulating HIF-1α/VEGF signaling. Acta Pharmacol Sin 2018; 39(3): 393-404.
[http://dx.doi.org/10.1038/aps.2017.161] [PMID: 29219948]
[17]
Li D, Shen C, Sun T, Zhang L, Deng H, Chai J. Mesenchymal stem cells promote incision wound repair in a mouse model. Trop J Pharm Res 2017; 16(6): 1317-23.
[http://dx.doi.org/10.4314/tjpr.v16i6.15]
[18]
Davis SC, Li J, Gil J, et al. The wound-healing effects of a next-generation anti-biofilm silver Hydrofiber wound dressing on deep partial-thickness wounds using a porcine model. Int Wound J 2018; 15(5): 834-9.
[http://dx.doi.org/10.1111/iwj.12935] [PMID: 29893025]
[19]
Cortés OL, Salazar-Beltrán LD, Rojas-Castañeda YA, Alvarado-Muriel A, Serna-Restrepo A, Grinspun D. Use of hydrocolloid dressings in preventing pressure ulcers in high-risk patients: A retrospective cohort. Invest Educ Enferm 2018; 36(1): e11.
[http://dx.doi.org/10.17533/udea.iee.v36n1e11] [PMID: 29898350]
[20]
Lei J, Sun L, Li P, et al. The wound dressings and their applications in wound healing and management. Health Sci J 2019; 13(4): 1-8.
[21]
Kumar A, Kaur H. Sprayed in-situ synthesis of polyvinyl alcohol/chitosan loaded silver nanocomposite hydrogel for improved antibacterial effects. Int J Biol Macromol 2020; 145: 950-64.
[http://dx.doi.org/10.1016/j.ijbiomac.2019.09.186] [PMID: 31669274]
[22]
Kumar A, Behl T, Chadha S. Synthesis of physically crosslinked PVA/Chitosan loaded silver nanoparticles hydrogels with tunable mechanical properties and antibacterial effects. Int J Biol Macromol 2020; 149: 1262-74.
[http://dx.doi.org/10.1016/j.ijbiomac.2020.02.048] [PMID: 32044364]
[23]
Sharma P, Kumar A, Dey AD, Behl T, Chadha S. Stem cells and growth factors-based delivery approaches for chronic wound repair and regeneration: A promise to heal from within. Life Sci 2021; 268(Mar): 118932-, 268, 118932.
[http://dx.doi.org/10.1016/j.lfs.2020.118932] [PMID: 33400933]
[24]
Jones RE, Foster DS, Hu MS, Longaker MT. Wound healing and fibrosis: Current stem cell therapies. Transfusion 2019; 59(S1): 884-92.
[http://dx.doi.org/10.1111/trf.14836] [PMID: 30737822]
[25]
Glat P, Orgill DP, Galiano R, et al. Placental membrane provides improved healing efficacy and lower cost versus a tissue-engineered human skin in the treatment of diabetic foot ulcerations. Plast Reconstr Surg Glob Open 2019; 7(8): e2371.
[http://dx.doi.org/10.1097/GOX.0000000000002371] [PMID: 31592387]
[26]
Ji ST, Kim H, Yun J, Chung JS, Kwon SM. Promising therapeutic strategies for mesenchymal stem cell-based cardiovascular regeneration: From cell priming to tissue engineering. Stem Cells Int 2017; 2017: 3945403.
[http://dx.doi.org/10.1155/2017/3945403] [PMID: 28303152]
[27]
Henríquez CM, Guerra GD, Vallejos MA, de la Fuente SD, Flores MT, Jimenez LM. In situ silver nanoparticle formation embedded into a photopolymerized hydrogel with biocide properties. J Nanostructure Chem 2014; 4(4): 119-32.
[http://dx.doi.org/10.1007/s40097-014-0125-y]
[28]
Ngandu T, Lehtisalo J, Solomon A, et al. A 2 year multidomain intervention of diet, exercise, cognitive training, and vascular risk monitoring versus control to prevent cognitive decline in at-risk elderly people (FINGER): A randomised controlled trial. Lancet 2015; 385(9984): 2255-63.
[http://dx.doi.org/10.1016/S0140-6736(15)60461-5] [PMID: 25771249]
[29]
Ravari H. Treatment of non-healing wounds with autologous bone marrow cells, platelets, fibrin glue and collagen matrix. Cytotherapy 2011; 13(6): 705-11.
[http://dx.doi.org/10.3109/14653249.2011.553594]
[30]
Wettstein R, Savic M, Pierer G, et al. Progenitor cell therapy for sacral pressure sore: A pilot study with a novel human chronic wound model. Stem Cell Res Ther 2014; 5(1): 18.
[http://dx.doi.org/10.1186/scrt407] [PMID: 24476740]
[31]
Huang YZ, Gou M, Da LC, Zhang WQ, Xie HQ. Mesenchymal stem cells for chronic wound healing: Current status of preclinical and clinical studies. Tissue Eng Part B Rev 2020; 26(6): 555-70.
[http://dx.doi.org/10.1089/ten.teb.2019.0351] [PMID: 32242479]
[32]
Garcia-Fernandez C, Lopez-Fernandez A, Borrós S, Lecina M, Vives J. Strategies for large-scale expansion of clinical-grade human multipotent mesenchymal stromal cells. Biochem Eng J 2020; 159: 15-, 159, 107601.
[http://dx.doi.org/10.1016/j.bej.2020.107601]
[33]
Badiavas EV, Ford D, Liu P, et al. Long-term bone marrow culture and its clinical potential in chronic wound healing. Wound Rep Reg 15(6): 856-65.
[http://dx.doi.org/10.1111/j.1524-475X.2007.00305.x]
[34]
Nussbaum J, Minami E, Laflamme MA, et al. Transplantation of undifferentiated murine embryonic stem cells in the heart: Teratoma formation and immune response. FASEB J 2007; 21(7): 1345-57.
[http://dx.doi.org/10.1096/fj.06-6769com] [PMID: 17284483]
[35]
Yan M, Sun S, Xu K, et al. Cardiac aging: From basic research to therapeutics. Oxid Med Cell Longev 2021; 2021: 9570325.
[http://dx.doi.org/10.1155/2021/9570325] [PMID: 33777324]
[36]
Arjmand B, Payab M, Goodarzi P. Learning Materials in Biosciences Eds.Biomedical Product Development: Bench to Bedside. (1st Edition.). Springer 2020; p. 157.
[http://dx.doi.org/10.1007/978-3-030-35626-2]
[37]
Baptista LS, Silva KR, Borojevic R. Obesity and weight loss could alter the properties of adipose stem cells? World J Stem Cells 2015; 7(1): 165-73.
[http://dx.doi.org/10.4252/wjsc.v7.i1.165] [PMID: 25621116]
[38]
Hayes PD, Alzuhir N, Curran G, Loftus IM. Topical oxygen therapy promotes the healing of chronic diabetic foot ulcers: A pilot study. J Wound Care 26(11): 652-60.
[http://dx.doi.org/10.12968/jowc.2017.26.11.652]
[39]
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): 1723-33.
[http://dx.doi.org/10.1021/acs.molpharmaceut.0c00177]
[40]
Loots MA, Lamme EN, Zeegelaar J, Mekkes JR, Bos JD, Middelkoop E. Differences in cellular infiltrate and extracellular matrix of chronic diabetic and venous ulcers versus acute wounds. J Invest Dermatol 1998; 111(5): 850-7.
[http://dx.doi.org/10.1046/j.1523-1747.1998.00381.x] [PMID: 9804349]
[41]
Keylock KT, Vieira VJ, Wallig MA, DiPietro LA, Schrementi M, Woods JA. Exercise accelerates cutaneous wound healing and decreases wound inflammation in aged mice. Am J Physiol Regul Integr Comp Physiol 2008; 294(1): R179-84.
[http://dx.doi.org/10.1152/ajpregu.00177.2007] [PMID: 18003791]
[42]
Nourbakhsh E, Mohammadi A, Salemizadeh Parizi M, Mansouri A, Ebrahimzadeh F. Role of Myeloid-derived suppressor cell (MDSC) in autoimmunity and its potential as a therapeutic target. Inflammopharmacol 2021; 29(5): 1307-15.
[http://dx.doi.org/10.1007/s10787-021-00846-3] [PMID: 34283371]
[43]
Miao M, Niu Y, Xie T, Yuan B, Qing C, Lu S. Diabetes-impaired wound healing and altered macrophage activation: A possible pathophysiologic correlation. Wound Repair Regen 2012; 20(2): 203-13.
[http://dx.doi.org/10.1111/j.1524-475X.2012.00772.x] [PMID: 22380690]
[44]
Bin Y, Sha H, Dongyun G, Jiangfan X, Nanbo L, Xiaobing F. Age-associated changes in regenerative capabilities of Mesenchymal stem cell: Impact on chronic wound repair. Int Wound J 2015; 13: 1252-9.
[http://dx.doi.org/10.1111/iwj.12491]
[45]
Helfinger V, Von GFF, Henke N, et al. Genetic deletion of Nox4 enhances cancerogen-induced formation of solid tumors. Proc Natl Acad Sci 16(11): 118.
[http://dx.doi.org/10.1073/pnas.2020152118]
[46]
Demidova-Rice TN, Durham JT, Herman IM. Wound healing angiogenesis: Innovations and challenges in acute and chronic wound healing. Adv Wound Care 2012; 1(1): 17-22.
[http://dx.doi.org/10.1089/wound.2011.0308]
[47]
Tejada S, Batle JM, Ferrer MD, et al. Therapeutic effects of hyperbaric oxygen in the process of wound healing. Curr Pharm Des 2019; 25(15): 1682-93.
[http://dx.doi.org/10.2174/1381612825666190703162648]
[48]
Xiao J, Kuang X, Dai L, Zhang L, He B. Anti-tumour effects of Keratin 6A in lung adenocarcinoma. Clin Respir J 2020; 14(7): 667-74.
[http://dx.doi.org/10.1111/crj.13182] [PMID: 32162441]
[49]
Szabowski A, Maas-Szabowski N, Andrecht S, et al. c-Jun and JunB antagonistically control cytokine-regulated mesenchymal–epidermal interaction in skin. Cell 2000; 103(5): 745-55.
[50]
Rennert RC, Januszyk M, Sorkin M, et al. Microfluidic single-cell transcriptional analysis rationally identifies novel surface marker profiles to enhance cell-based therapies. Nat Commun 2016; 7(1): 11945.
[http://dx.doi.org/10.1038/ncomms11945] [PMID: 27324848]
[51]
Kumar A, Jaiswal M. Design and in vitro investigation of nanocomposite hydrogel based in situ spray dressing for chronic wounds and synthesis of silver nanoparticles using green chemistry. J Appl Polym Sci 2016; 133(14): 43260.
[http://dx.doi.org/10.1002/app.43260]
[52]
Al‐Shaibani MB, Wang XN, Lovat PE, Dickinson AM. Cellular therapy for wounds: Applications of mesenchymal stem cells in wound healing. Wound healing-new insights into ancient challenges. London. InTech 2016; 12: 99-131.
[53]
Kucharzewski M, Rojczyk E, Wilemska-Kucharzewska K, Wilk R, Hudecki J, Los MJ. Novel trends in application of stem cells in skin wound healing. Eur J Pharmacol 2019; 843: 307-15.
[http://dx.doi.org/10.1016/j.ejphar.2018.12.012] [PMID: 30537490]
[54]
Keirouz A, Chung M, Kwon J, Fortunato G, Radacsi N. 2D and 3D electrospinning technologies for the fabrication of nanofibrous scaffolds for skin tissue engineering: A review. WIREs Interdiscip Rev Nanomed Nanobiotechnol 2020; 12(4): e1626.
[http://dx.doi.org/10.1002/wnan.1626] [PMID: 32166881]
[55]
Jessen KR, Mirsky R, Lloyd AC. Schwann cells: Development and role in nerve repair. Cold Spring Harb Perspect Biol 2015; 7(7): a020487.
[http://dx.doi.org/10.1101/cshperspect.a020487] [PMID: 25957303]
[56]
Kim HS, Sun X, Lee JH, Kim HW, Fu X, Leong KW. Advanced drug delivery systems and artificial skin grafts for skin wound healing. Adv Drug Deliv Rev 2019; 146: 209-39.
[http://dx.doi.org/10.1016/j.addr.2018.12.014] [PMID: 30605737]
[57]
Lim SK, Khoo BY. An overview of mesenchymal stem cells and their potential therapeutic benefits in cancer therapy. Oncol Lett 2021; 22(5): 1-4.
[http://dx.doi.org/10.3892/ol.2021.13046]
[58]
Zhang Y, Xing Y, Jia L, et al. An in vitro comparative study of multisource derived human mesenchymal stem cells for bone tissue engineering. Stem Cells Develop 2018; 27(23): 1634-45.
[http://dx.doi.org/10.1089/scd.2018.0119]
[59]
Nishikawa G, Kawada K, Nakagawa J, et al. Bone marrow-derived mesenchymal stem cells promote colorectal cancer progression via CCR5. Cell Death Dis 2019; 10(4): 264.
[http://dx.doi.org/10.1038/s41419-019-1508-2] [PMID: 30890699]
[60]
Martin I, Galipeau J, Kessler C, Le Blanc K, Dazzi F. Challenges for mesenchymal stromal cell therapies. Science Transl Med 2019; 11: 480.
[http://dx.doi.org/10.1126/scitranslmed.aat2189]
[61]
Wilson A, Hodgson-Garms M, Frith JE, Genever P. Multiplicity of mesenchymal stromal cells: Finding the right route to therapy. Frontiers Immunol 2019; 16(10): 1112.
[http://dx.doi.org/10.3389/fimmu.2019.01112]
[62]
Kouchakian MR, Baghban N, Moniri SF, et al. The clinical trials of mesenchymal stromal cells therapy. Stem Cells Int 2021; 2021: 1634782.
[http://dx.doi.org/10.1155/2021/1634782] [PMID: 34745268]
[63]
Huo J, Sun S, Geng Z, et al. Bone marrow-derived mesenchymal stem cells promoted cutaneous wound healing by regulating keratinocyte migration via β2-adrenergic receptor signaling. Mol Pharmaceutics 2018; 15(7): 2513-27.
[http://dx.doi.org/10.1021/acs.molpharmaceut.7b01138]
[64]
Humpert PM, Bärtsch U, Konrade I, et al. Locally applied mononuclear bone marrow cells restore angiogenesis and promote wound healing in a type 2 diabetic patient. Exp Clin Endocrinol Diabetes 2005; 113(9): 538-40.
[http://dx.doi.org/10.1055/s-2005-872886] [PMID: 16235157]
[65]
Falanga V, Iwamoto S, Chartier M, et al. Autologous bone marrow-derived cultured mesenchymal stem cells delivered in a fibrin spray accelerate healing in murine and human cutaneous wounds. Tissue Eng 2007; 13(6): 1299-312.
[http://dx.doi.org/10.1089/ten.2006.0278] [PMID: 17518741]
[66]
Chen L, Qu J, Xiang C. The multi-functional roles of menstrual blood-derived stem cells in regenerative medicine. Stem Cell Res Ther 2019; 10(1): 1.
[http://dx.doi.org/10.1186/s13287-018-1105-9] [PMID: 30606242]
[67]
Cuenca J, Le-Gatt A, Castillo V, et al. the reparative abilities of menstrual stem cells modulate the wound matrix signals and improve cutaneous regeneration. Front Physiol 2018; 9: 464.
[http://dx.doi.org/10.3389/fphys.2018.00464] [PMID: 29867527]
[68]
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.
[http://dx.doi.org/10.1186/s13287-015-0013-5]
[69]
Mirzadegan E, Golshahi H, Kazemnejad S. Current evidence on immunological and regenerative effects of menstrual blood stem cells seeded on scaffold consisting of amniotic membrane and silk fibroin in chronic wound. Int Immunopharmacol 2020; 85: 106595.
[http://dx.doi.org/10.1016/j.intimp.2020.106595] [PMID: 32454419]
[70]
Zeppieri M, Salvetat ML, Beltrami A, et al. Adipose derived stem cells for corneal wound healing after laser induced corneal lesions in mice. J Clin Med 2017; 6(12): E115.
[http://dx.doi.org/10.3390/jcm6120115] [PMID: 29206194]
[71]
Chu J, Shi P, Deng X, et al. Dynamic multiphoton imaging of acellular dermal matrix scaffolds seeded with mesenchymal stem cells in diabetic wound healing. J Biophotonics 2018; 11(7): e201700336.
[http://dx.doi.org/10.1002/jbio.201700336] [PMID: 29575792]
[72]
Zhang M, Cao Y, Li X, et al. Cd271 mediates proliferation and differentiation of epidermal stem cells to support cutaneous burn wound healing. Cell Tissue Res 2018; 371(2): 273-82.
[http://dx.doi.org/10.1007/s00441-017-2723-8] [PMID: 29150821]
[73]
Yang K, Song HF, He S, et al. Effect of neuron-derived neurotrophic factor on rejuvenation of human adipose-derived stem cells for cardiac repair after myocardial infarction. J Cell Mol Med 2019; 23(9): 5981-93.
[http://dx.doi.org/10.1111/jcmm.14456] [PMID: 31287219]
[74]
Kosaraju R, Rennert RC, Maan ZN, et al. Adipose-derived stem cell–seeded hydrogels increase endogenous progenitor cell recruitment and neovascularization in wounds 1. Tissue Eng Part A 2016; 22(3-4): 295-305.
[http://dx.doi.org/10.1089/ten.tea.2015.0277] [PMID: 26871860]
[75]
Barrera JA, Trotsyuk AA, Maan ZN, et al. Adipose-derived stromal cells seeded in pullulan-collagen hydrogels improve healing in murine burns. Tissue Eng Part A 2021; 27(11-12): 844-56.
[http://dx.doi.org/10.1089/ten.tea.2020.0320] [PMID: 33789446]
[76]
Kaisang L, Siyu W, Lijun F, Daoyan P, Xian CJ, Jie S. Adipose-derived stem cells seeded in Pluronic F-127 hydrogel promotes diabetic wound healing. J Surg Res 2017; 217: 63-74.
[http://dx.doi.org/10.1016/j.jss.2017.04.032] [PMID: 28595815]
[77]
Nurkesh A, Jaguparov A, Jimi S, Saparov A. Recent advances in the controlled release of growth factors and cytokines for improving cutaneous wound healing. Front Cell Dev Biol 2020; 14: 638.
[http://dx.doi.org/10.3389/fcell.2020.00638]
[78]
Yamakawa S, Hayashida K. Advances in surgical applications of growth factors for wound healing. Burns Trauma 2019; 7: 10.
[http://dx.doi.org/10.1186/s41038-019-0148-1] [PMID: 30993143]
[79]
Sultankulov B, Berillo D, Sultankulova K, Tokay T, Saparov A. Progress in the development of chitosan-based biomaterials for tissue engineering and regenerative medicine. Biomolecules 2019; 9(9): 470.
[http://dx.doi.org/10.3390/biom9090470]
[80]
Chen G, Ren J, Deng Y, et al. An injectable, wound-adapting, self-healing hydrogel for fibroblast growth factor 2 delivery system in tissue repair applications. J Biomed Nanotechnol 2017; 13(12): 1660-72.
[http://dx.doi.org/10.1166/jbn.2017.2443]
[81]
Ornitz DM, Itoh N. Fibroblast growth factors. Genome Biol 2001; 2(3)
[http://dx.doi.org/10.1186/gb-2001-2-3-reviews3005]
[82]
Micera A, Vigneti E, Pickholtz D, et al. Nerve growth factor displays stimulatory effects on human skin and lung fibroblasts, demonstrating a direct role for this factor in tissue repair. Proc Natl Acad Sci USA 2001; 98(11): 6162-7.
[http://dx.doi.org/10.1073/pnas.101130898] [PMID: 11344264]
[83]
Heldin CH, Westermark B. Mechanism of action and in vivo role of platelet-derived growth factor. Physiol Rev 1999; 79(4): 1283-316.
[http://dx.doi.org/10.1152/physrev.1999.79.4.1283] [PMID: 10508235]
[84]
Yager DR, Chen SM, Ward SI, Olutoye OO, Diegelmann RF, Kelman Cohen I. Ability of chronic wound fluids to degrade peptide growth factors is associated with increased levels of elastase activity and diminished levels of proteinase inhibitors. Wound Repair Regen 1997; 5(1): 23-32.
[http://dx.doi.org/10.1046/j.1524-475X.1997.50108.x] [PMID: 16984454]
[85]
Sugiyama K, Ishii G, Ochiai A, Esumi H. Improvement of the breaking strength of wound by combined treatment with recombinant human G-CSF, recombinant human M-CSF, and a TGF-beta1 receptor kinase inhibitor in rat skin. Cancer Sci 2008; 99(5): 1021-8.
[http://dx.doi.org/10.1111/j.1349-7006.2008.00761.x] [PMID: 18380792]
[86]
Bodnar RJ. Epidermal growth factor and epidermal growth factor receptor: The Yin and Yang in the treatment of cutaneous wounds and cancer. Adv Wound Care 2013; 2(1): 24-9.
[http://dx.doi.org/10.1089/wound.2011.0326]
[87]
Steenfos HH. Growth factors and wound healing. Scand J Plast Reconstr Surg Hand Surg 1994; 28(2): 95-105.
[http://dx.doi.org/10.3109/02844319409071186] [PMID: 8079129]
[88]
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.
[http://dx.doi.org/10.1111/j.1524-475X.2008.00410.x] [PMID: 19128254]
[89]
Romano Di Peppe S, Mangoni A, Zambruno G, et al. Adenovirus-mediated VEGF(165) gene transfer enhances wound healing by promoting angiogenesis in CD1 diabetic mice. Gene Ther 2002; 9(19): 1271-7.
[http://dx.doi.org/10.1038/sj.gt.3301798] [PMID: 12224009]
[90]
Duncan MR, Frazier KS, Abramson S, et al. Connective tissue growth factor mediates transforming growth factor β-induced collagen synthesis: Down-regulation by cAMP. FASEB J 1999; 13(13): 1774-86.
[http://dx.doi.org/10.1096/fasebj.13.13.1774] [PMID: 10506580]
[91]
Chen L, Tredget EE, Wu PY, Wu Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One 2008; 3(4): e1886.
[http://dx.doi.org/10.1371/journal.pone.0001886] [PMID: 18382669]
[92]
Coalson E, Bishop E, Liu W, et al. Stem cell therapy for chronic skin wounds in the era of personalized medicine: From bench to bedside. Genes Diseases 2019; 6(4): 342-58.
[93]
Kandhwal M, Behl T, Kumar A, Arora S. Understanding the potential role and delivery approaches of nitric oxide in chronic wound healing management. Curr Pharm Des 2021; 27(17): 1999-2014.
[http://dx.doi.org/10.2174/1381612826666201026152209] [PMID: 33106138]
[94]
Kumar A, Behl T, Chadha S. A rationalized and innovative perspective of nanotechnology and nanobiotechnology in chronic wound management. J Drug Deliv Sci Technol 2020; 60: 101930.
[http://dx.doi.org/10.1016/j.jddst.2020.101930]
[95]
Blot SI, Monstrey SJ, Hoste EA, et al. Cultured epithelial autografts in extensive burn coverage of severely traumatized patients: A five year single-center experience with 30 patients. Burns 2001; 27(4): 418-9.
[http://dx.doi.org/10.1016/S0305-4179(00)00123-6] [PMID: 11417518]
[96]
Parenteau N. Skin: The first tissue-engineered products. Sci Am 1999; 280(4): 83-4.
[http://dx.doi.org/10.1038/scientificamerican0499-83] [PMID: 10232963]
[97]
Cooper ML, Hansbrough JF, Spielvogel RL, Cohen R, Bartel RL, Naughton G. In vivo optimization of a living dermal substitute employing cultured human fibroblasts on a biodegradable polyglycolic acid or polyglactin. Mesh Biomaterials 1991; 12(2): 243-8.
[98]
Cardinal M, Eisenbud DE, Armstrong DG, et al. Serial surgical debridement: A retrospective study on clinical outcomes in chronic lower extremity wounds. Wound Repair Regen 2009; 17(3): 306-11.
[http://dx.doi.org/10.1111/j.1524-475X.2009.00485.x] [PMID: 19660037]
[99]
Tabasi H, Babaei M, Abnous K, et al. Metal-polymer-coordinated complexes as potential nanovehicles for drug delivery. J Nanostruct Chem 2021; 11: 501-26.
[http://dx.doi.org/10.1007/s40097-021-00432-7]
[100]
Murphy PS, Evans GRD. Advances in wound healing: A review of current wound healing products. Plast Surg Int 2012; 2012: 190436.
[http://dx.doi.org/10.1155/2012/190436] [PMID: 22567251]
[101]
Kim PJ, Heilala M, Steinberg JS, Weinraub GM. Bioengineered alternative tissues and hyperbaric oxygen in lower extremity wound healing. Clin Podiatr Med Surg 2007; 24(3): 529-46.
[http://dx.doi.org/10.1016/j.cpm.2007.03.011] [PMID: 17613390]
[102]
Gurtner GC, Chapman MA. Regenerative medicine: charting a new course in wound healing. Adv Wound Care (New Rochelle) 2016; 5(7): 314-28.
[http://dx.doi.org/10.1089/wound.2015.0663] [PMID: 27366592]
[103]
Tottoli EM, Dorati R, Genta I, Chiesa E, Pisani S, Conti B. Skin wound healing process and new emerging technologies for skin wound care and regeneration. Pharmaceutics 2020; 12(8): 735.
[http://dx.doi.org/10.3390/pharmaceutics12080735] [PMID: 32764269]
[104]
Nourian DA, Mirahmadi BF, Chehelgerdi M, Raeisi DS. Skin tissue engineering: Wound healing based on stem-cell-based therapeutic strategies. Stem Cell Res Ther 2019; 10(1): 111.
[http://dx.doi.org/10.1186/s13287-019-1212-2] [PMID: 30922387]
[105]
Park SR, Kim JW, Jun HS, Roh JY, Lee HY, Hong IS. Stem cell secretome and its effect on cellular mechanisms relevant to wound healing. Mol Ther 2018; 26(2): 606-17.
[http://dx.doi.org/10.1016/j.ymthe.2017.09.023] [PMID: 29066165]
[106]
Chitturi RT, Balasubramaniam AM, Parameswar RA, Kesavan G, Haris KTM, Mohideen K. The role of myofibroblasts in wound healing, contraction and its clinical implications in cleft palate repair. J Int Oral Health 2015; 7(3): 75-80.
[PMID: 25878485]
[107]
Dick MK, Miao JH, Limaiem F. Histology, fibroblast. StatPearls 2021.
[108]
Lee DK, Song SU. Immunomodulatory mechanisms of mesenchymal stem cells and their therapeutic applications. Cell Immunol 2018; 326(326): 68-76.
[http://dx.doi.org/10.1016/j.cellimm.2017.08.009] [PMID: 28919171]
[109]
Villamil BAC, Segura PHR, Lopez-Garcia JA, et al. Bovine decellularized amniotic membrane: Extracellular matrix as scaffold for mammalian skin. Polymers (Basel) 2020; 12(3): 590.
[http://dx.doi.org/10.3390/polym12030590] [PMID: 32151022]
[110]
Shoji-Pietraszkiewicz A, Sakamoto M, Katsube M, et al. Treatment of giant congenital melanocytic nevi with cultured epithelial autografts: Clinical and histopathological analysis. Regen Ther 2021; 18(18): 1-6.
[http://dx.doi.org/10.1016/j.reth.2021.02.003] [PMID: 33778134]
[111]
Bi H, Li H, Zhang C, et al. Stromal vascular fraction promotes migration of fibroblasts and angiogenesis through regulation of extracellular matrix in the skin wound healing process. Stem Cell Res Ther 2019; 10(1): 302.
[http://dx.doi.org/10.1186/s13287-019-1415-6] [PMID: 31623669]
[112]
Zahorec P, Koller J, Danisovic L, Bohac M. Mesenchymal stem cells for chronic wounds therapy. Cell Tissue Bank 2015; 16(1): 19-26.
[http://dx.doi.org/10.1007/s10561-014-9440-2] [PMID: 24651970]
[113]
Joseph A, Baiju I, Bhat IA, et al. Mesenchymal stem cell-conditioned media: A novel alternative of stem cell therapy for quality wound healing. J Cell Physiol 2020; 235(7-8): 5555-69.
[http://dx.doi.org/10.1002/jcp.29486] [PMID: 31960454]
[114]
Yi H, Wang Y, Yang Z, Xie Z. Efficacy assessment of mesenchymal stem cell transplantation for burn wounds in animals: A systematic review. Stem Cell Res Ther 2020; 11(1): 372.
[http://dx.doi.org/10.1186/s13287-020-01879-1] [PMID: 32859266]
[115]
Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: Past, present, and future. Stem Cell Res Ther 2019; 10(1): 68.
[http://dx.doi.org/10.1186/s13287-019-1165-5]
[116]
Zhao G, Liu F, Lan S, et al. Large-scale expansion of Wharton’s jelly-derived mesenchymal stem cells on gelatin microbeads, with retention of self-renewal and multipotency characteristics and the capacity for enhancing skin wound healing. Stem Cell Res Ther 2015; 6(1): 38.
[http://dx.doi.org/10.1186/s13287-015-0031-3] [PMID: 25889402]
[117]
Pedram ZR, Mokhtari J, Abbasi M. Biopolymer based three‐dimensional biomimetic micro/nanofibers scaffolds with porous structures via tailored charge repulsions for skin tissue regeneration. Polym Adv Technol 2021; 32(9): 3535-48.
[http://dx.doi.org/10.1002/pat.5364]
[118]
Dzierzak E, Bigas A. Blood development: Hematopoietic stem cell dependence and independence. Cell Stem Cell 2018; 22(5): 639-51.
[http://dx.doi.org/10.1016/j.stem.2018.04.015]
[119]
Ma G, Jiang Y, Liang M, et al. Dynamic monitoring of CD45-/CD31+/DAPI+ circulating endothelial cells aneuploid for chromosome 8 during neoadjuvant chemotherapy in locally advanced breast cancer. Ther Adv Med Oncol 2020; 12: 1758835920918470.
[http://dx.doi.org/10.1177/1758835920918470] [PMID: 32489429]
[120]
Yoder MC. Human endothelial progenitor cells. Cold Spring Harb Perspect Med 2012; 2(7): a006692.
[http://dx.doi.org/10.1101/cshperspect.a006692] [PMID: 22762017]
[121]
Patel J, Seppanen EJ, Rodero MP, et al. Functional definition of progenitors versus mature endothelial cells reveals key SoxF-dependent differentiation process. Circulation 2017; 135(8): 786-805.
[http://dx.doi.org/10.1161/CIRCULATIONAHA.116.024754]
[122]
Zhang J, Guan J, Niu X, et al. Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. J Trans Med 2015; 13(1): 49.
[http://dx.doi.org/10.1186/s12967-015-0417-0]
[123]
Coll M, Perea L, Boon R, et al. Generation of hepatic stellate cells from human pluripotent stem cells enables in vitro modeling of liver fibrosis. Cell Stem Cell 2018; 23(1): 101-13.
[http://dx.doi.org/10.1016/j.stem.2018.05.027]
[124]
Lee MJ, Kim J, Lee KI, Shin JM, Chae JI, Chung HM. Enhancement of wound healing by secretory factors of endothelial precursor cells derived from human embryonic stem cells. Cytotherapy 2011; 13(2): 165-78.
[http://dx.doi.org/10.3109/14653249.2010.512632] [PMID: 21235296]
[125]
Kosaric N, Kiwanuka H, Gurtner GC. Stem cell therapies for wound healing. Expert Opin Biol Ther 2019; 19(6): 575-85.
[http://dx.doi.org/10.1080/14712598.2019.1596257] [PMID: 30900481]
[126]
van den Broek LJ, Bergers LIJC, Reijnders CMA, Gibbs S. Progress and future prospectives in skin-on-chip development with emphasis on the use of different cell types and technical challenges. Stem Cell Rev Rep 2017; 13(3): 418-29.
[http://dx.doi.org/10.1007/s12015-017-9737-1] [PMID: 28536890]
[127]
Guha P, Morgan JW, Mostoslavsky G, Rodrigues NP, Boyd AS. Lack of immune response to differentiated cells derived from syngeneic induced pluripotent stem cells. Cell Stem Cell 2017; 21(1): 144-8.
[http://dx.doi.org/10.1016/j.stem.2017.03.012] [PMID: 28686864]
[128]
Gledhill K, Guo Z, Umegaki-Arao NC, Higgins A, Christiano AM. Melanin transfer in human 3D skin equivalents generated exclusively from induced pluripotent stem cells. PLoS One 2015; 10(8): e0136713.
[129]
Kanji S, Das H. Advances of stem cell therapeutics in cutaneous wound healing and regeneration. Mediators Inflamm 2017; 2017: 5217967.
[http://dx.doi.org/10.1155/2017/5217967] [PMID: 29213192]
[130]
Vijayavenkataraman S, Yan WC, Lu WF, Wang CH, Fuh JYH. 3D bioprinting of tissues and organs for regenerative medicine. Adv Drug Deliv Rev 2018; 132(132): 296-332.
[http://dx.doi.org/10.1016/j.addr.2018.07.004] [PMID: 29990578]
[131]
Mirjalili M, Abbasipour M. Comparison between antibacterial activity of some natural dyes and silver nanoparticles. J Nanostructure Chem 2013; 3(1): 37.
[http://dx.doi.org/10.1186/2193-8865-3-37]
[132]
Magne TM, de Oliveira VT, Alencar LMR, et al. Graphene and its derivatives: Understanding the main chemical and medicinal chemistry roles for biomedical applications. J Nanostructure Chem 2021; 1-35.
[http://dx.doi.org/10.1007/s40097-021-00444-3] [PMID: 34512930]
[133]
Weigelt MA, Sivamani R, Lev-Tov H. The therapeutic potential of cannabinoids for integumentary wound management. Exp Dermatol 2021; 30(2): 201-11.
[http://dx.doi.org/10.1111/exd.14241] [PMID: 33205468]
[134]
Wang S, Qu X, Zhao RC. Clinical applications of mesenchymal stem cells. J Hematol Oncol 2012; 5(1): 19.
[http://dx.doi.org/10.1186/1756-8722-5-19] [PMID: 22546280]
[135]
Abedi M, Alavi-Moghadam S, Payab M, et al. Mesenchymal stem cell as a novel approach to systemic sclerosis; current status and future perspectives. Cell Regen (Lond) 2020; 9(1): 20.
[http://dx.doi.org/10.1186/s13619-020-00058-0] [PMID: 33258056]
[136]
Squillaro T, Peluso G, Galderisi U. Clinical trials with mesenchymal stem cells: An update. Cell Transplant 2016; 25(5): 829-48.
[http://dx.doi.org/10.3727/096368915X689622] [PMID: 26423725]
[137]
Matthay MA, Pati S, Lee JW. Concise review: Mesenchymal stem (stromal) cells: Biology and preclinical evidence for therapeutic potential for organ dysfunction following trauma or sepsis. Stem Cells 2017; 35(2): 316-24.
[http://dx.doi.org/10.1002/stem.2551] [PMID: 27888550]
[138]
Chu DT, Phuong TNT, Tien NLB, et al. An update on the progress of isolation, culture, storage, and clinical application of human bone marrow mesenchymal stem/stromal cells. Int J Mol Sci 2020; 21(3): 708.
[http://dx.doi.org/10.3390/ijms21030708] [PMID: 31973182]
[139]
Dash NR, Dash SN, Routray P, Mohapatra S, Mohapatra PC. Targeting nonhealing ulcers of lower extremity in human through autologous bone marrow-derived mesenchymal stem cells. Rejuvenation Res 2009; 12(5): 359-66.
[http://dx.doi.org/10.1089/rej.2009.0872] [PMID: 19929258]
[140]
Sarasúa JG, López SP, Viejo MÁ, et al. Treatment of pressure ulcers with autologous bone marrow nuclear cells in patients with spinal cord injury. J Spinal Cord Med 2011; 34(3): 301-7.
[http://dx.doi.org/10.1179/2045772311Y.0000000010] [PMID: 21756569]
[141]
Ichioka S, Kouraba S, Sekiya N, Ohura N, Nakatsuka T. Bone marrow-impregnated collagen matrix for wound healing: Experimental evaluation in a microcirculatory model of angiogenesis, and clinical experience. Br J Plast Surg 2005; 58(8): 1124-30.
[http://dx.doi.org/10.1016/j.bjps.2005.04.054] [PMID: 16043157]
[142]
ClinicalTrials.gov. Available from: https://clinicaltrials.gov
[143]
Lataillade JJ, Doucet C, Bey E, et al. New approach to radiation burn treatment by dosimetry-guided surgery combined with autologous mesenchymal stem cell therapy. Regen Med 2007; 2(5): 785-94.
[http://dx.doi.org/10.2217/17460751.2.5.785] [PMID: 17907931]
[144]
U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/ (Accessed on December 14, 2020).
[145]
Covidence is used by world-leading evidence organizations. Available from: https://www.covidence.org (Accessed on December 14, 2020).
[146]
ClinicalTrials.gov. Available from: https://clinicaltrials.gov/ct2/show/NCT02104713 (Accessed on December 14, 2020).
[147]
ClinicalTrials.gov. Available from: https://clinicaltrials.gov/ct2/show/NCT01443689 (Accessed on December 14, 2020).
[148]
Ojeh N, Pastar I, Tomic-Canic M, Stojadinovic O. Stem cells in skin regeneration, wound healing, and their clinical applications. Int J Mol Sci 2015; 16(10): 25476-501.
[http://dx.doi.org/10.3390/ijms161025476] [PMID: 26512657]
[149]
Brodell RT. Dermatoethics: Contemporary ethics and professionalism in dermatology. J Am Acad Dermatol 2021; 85(3): e205.
[http://dx.doi.org/10.1016/j.jaad.2021.04.031] [PMID: 33852923]
[150]
Amani S, Shahrooz R, Hobbenaghi R, et al. Angiogenic effects of cell therapy within a biomaterial scaffold in a rat hind limb ischemia model. Sci Rep 2021; 11(1): 20545.
[http://dx.doi.org/10.1038/s41598-021-99579-0] [PMID: 34654868]
[151]
Huang W, Chen Y, Wang N, Yin G, Wei C, Xu W. Effectiveness and safety of human amnion/chorion membrane therapy for diabetic foot ulcers: An updated meta-analysis of randomized clinical trials. Wound Repair Regen 2020; 28(6): 739-50.
[http://dx.doi.org/10.1111/wrr.12851] [PMID: 32715574]
[152]
Rayman G, Vas P, Dhatariya K, et al. Guidelines on use of interventions to enhance healing of chronic foot ulcers in diabetes (IWGDF 2019 update). Diabetes Metab Res Rev 2020; 36(S1): e3283.
[http://dx.doi.org/10.1002/dmrr.3283] [PMID: 32176450]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy