Home From current landscape to future horizon in stem cell therapy for tissue regeneration and wound healing: bridging the gap
Article
Licensed
Unlicensed Requires Authentication

From current landscape to future horizon in stem cell therapy for tissue regeneration and wound healing: bridging the gap

  • Kashvy R. Morakhia ORCID logo , Aayushi C. Shah ORCID logo , Mannat P. Patel ORCID logo , Jainam K. Shah ORCID logo , Rajanikant Patel ORCID logo and Mehul R. Chorawala ORCID logo EMAIL logo
Published/Copyright: May 23, 2025
Become an author with De Gruyter Brill

Abstract

Stem cell therapy has emerged as a groundbreaking approach in regenerative medicine, offering immense potential for tissue regeneration and wound healing. Stem cells, with their ability to self-renew and differentiate into specialized cell types, provide innovative therapeutic strategies for variety of medical conditions. Key stem cell types, including embryonic, induced pluripotent, and adult stem cells such as mesenchymal and hematopoietic stem cells, play pivotal roles in regenerative processes and wound repair. In tissue regeneration, stem cells replenish damaged or necrotic cells by differentiating into specialized cell types like bone, muscle, or nerve cells, thus restoring the structural and functional integrity of tissues. In wound healing, stem cells stimulate angiogenesis, generate new skin cells, and modulate immune responses to enhance repair. This multifaceted therapeutic potential has paved the way for clinical applications in cardiovascular, neurological, musculoskeletal, and autoimmune disorders, as well as skin and burn injuries. This review highlights recent advancements in stem cell therapy, exploring its clinical applications and addressing challenges such as immune rejection, ethical concerns, scalability, and the need for long-term clinical trials. The article underscores the importance of continued research to fully realize the transformative potential of stem cell therapy in modern medicine.


Corresponding author: Mehul R. Chorawala, Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India, E-mail:
Kashvy R. Morakhia and Aayushi C. Shah contributed equally to this work.

Acknowledgments

The authors are grateful to Prof. Gaurang B. Shah, Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India for kind support and guidance in manuscript preparation. The authors also extend their appreciation to L. M. College of Pharmacy, Ahmedabad, India for providing continuous library resources support throughout literature survey and data collection. The authors are also thankful to Sharaman Sci-Med Writing Association, India for designing Figure 4 and providing continuous support for improvising all figures dpi during revision 1.

  1. Research ethics: Not applicable.

  2. Informed consent: Not applicable.

  3. Author contributions: KRM, ACS – Manuscript original first draft preparation and subsequent editing, Literature and data survey, Figures; MPP, JKS – Data acquisition and curation, Tables, Graphical abstract; RP – Definition of intellectual content, Manuscript draft review and editing, Validation of acquired data, Referencing; MRC – Review topic conception, Design of content and skeleton, Manuscript draft review, editing and approval; Figures and Tables conception, Overall monitoring and co-ordination.

  4. Use of Large Language Models, AI and Machine Learning Tools: We confirm that no generative AI, large language models (LLMs), or machine learning tools were used in the writing, editing, or analysis of the manuscript content. All authors contributed intellectually and manually to the preparation of the manuscript.

  5. Conflicts of interest: The authors declare no conflict of interest to report.

  6. Research funding: The authors declare that no funds, grants, or other support have been received during or for the preparation of this manuscript.

  7. Data availability: As our manuscript is review article, no new data were created or analyzed in this study. Data sharing does not apply to this article. However, the datasets/tables/figures generated during during the preparation of current manuscript are available from the corresponding author on reasonable request.

  8. Supplementary Materials: No available.

References

1. Guo, J, Huang, X, Dou, L, Yan, M, Shen, T, Tang, W, et al.. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Targeted Ther 2022;7:391. https://doi.org/10.1038/s41392-022-01251-0.Search in Google Scholar PubMed PubMed Central

2. Beyene, RT, Derryberry, SL, Barbul, A. The effect of comorbidities on wound healing. Surg Clin North Am 2020;100:695–705. https://doi.org/10.1016/j.suc.2020.05.002.Search in Google Scholar PubMed

3. Leung, E, Wongrakpanich, S, Munshi, MN. Diabetes management in the elderly. Diabetes Spectr 2018;31:245–53. https://doi.org/10.2337/ds18-0033.Search in Google Scholar PubMed PubMed Central

4. Zakrzewski, W, Dobrzyński, M, Szymonowicz, M, Rybak, Z. Stem cells: past, present, and future. Stem Cell Res Ther 2019;10:68. https://doi.org/10.1186/s13287-019-1165-5.Search in Google Scholar PubMed PubMed Central

5. Nourian Dehkordi, A, Mirahmadi Babaheydari, F, Chehelgerdi, M, Raeisi, DS. Skin tissue engineering: wound healing based on stem-cell-based therapeutic strategies. Stem Cell Res Ther 2019;10:111. https://doi.org/10.1186/s13287-019-1212-2.Search in Google Scholar PubMed PubMed Central

6. Maheswary, T, Nurul, AA, Fauzi, MB. The insights of microbes’ roles in wound healing: a comprehensive review. Pharmaceutics 2021;13:981. https://doi.org/10.3390/pharmaceutics13070981.Search in Google Scholar PubMed PubMed Central

7. Han, G, Ceilley, R. Chronic wound healing: a review of current management and treatments. Adv Ther 2017;34:599–610. https://doi.org/10.1007/s12325-017-0478-y.Search in Google Scholar PubMed PubMed Central

8. Wilkinson, HN, Hardman, MJ. Wound healing: cellular mechanisms and pathological outcomes. Open Biol 2020;10:200223. https://doi.org/10.1098/rsob.200223.Search in Google Scholar PubMed PubMed Central

9. Sorg, H, Tilkorn, DJ, Hager, S, Hauser, J, Mirastschijski, U. Skin wound healing: an update on the current knowledge and concepts. Eur Surg Res 2017;58:81–94. https://doi.org/10.1159/000454919.Search in Google Scholar PubMed

10. Soliman, AM, Barreda, DR. Acute inflammation in tissue healing. Int J Mol Sci 2023;24:641. https://doi.org/10.3390/ijms24010641.Search in Google Scholar PubMed PubMed Central

11. Mahla, RS. Stem cells applications in regenerative medicine and disease therapeutics. Int J Cell Biol 2016;2016:6940283. https://doi.org/10.1155/2016/6940283.Search in Google Scholar PubMed PubMed Central

12. Piovani, D, Nikolopoulos, GK, Bonovas, S. Non-communicable diseases: the invisible epidemic. J Clin Med 2022;11:5939. https://doi.org/10.3390/jcm11195939.Search in Google Scholar PubMed PubMed Central

13. Mehanna, RA, Essawy, MM, Barkat, MA, Awaad, AK, Thabet, EH, Hamed, HA, et al.. Cardiac stem cells: current knowledge and future prospects. World J Stem Cells 2022;14:1–40. https://doi.org/10.4252/wjsc.v14.i1.1.Search in Google Scholar PubMed PubMed Central

14. Moon, KC, Suh, HS, Kim, KB, Han, SK, Young, KW, Lee, JW, et al.. Potential of allogeneic adipose-derived stem cell–hydrogel complex for treating diabetic foot ulcers. Diabetes 2019;68:837–46. https://doi.org/10.2337/db18-0699.Search in Google Scholar PubMed

15. Aly, RM. Current state of stem cell-based therapies: an overview. Stem Cell Invest 2020;7:8. https://doi.org/10.21037/SCI-2020-001.Search in Google Scholar

16. Bahari, M, Mokhtari, H, Yeganeh, F. Stem cell therapy, the market, the opportunities and the threat. Int J Mol Cell Med 2023;12:310–19. https://doi.org/10.22088/IJMCM.BUMS.12.3.310.Search in Google Scholar PubMed PubMed Central

17. Stem cell and exosome products. Centers for Disease Control and Prevention. https://archive.cdc.gov/www_cdc_gov/hai/outbreaks/stem-cell-products.html [Accepted 5 Sep 2023].Search in Google Scholar

18. Approved cellular and gene therapy products. FDA. https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/approved-cellular-and-gene-therapy-products [Accepted 5 Sep 2023].Search in Google Scholar

19. Desgres, M, Menasché, P. Clinical translation of pluripotent stem cell therapies: challenges and considerations. Cell Stem Cell 2019;25:594–606. https://doi.org/10.1016/j.stem.2019.10.001.Search in Google Scholar PubMed

20. Abdul-Al, M, Kyeremeh, GK, Saeinasab, M, Heidari Keshel, S, Sefat, F. Stem cell niche microenvironment: review. Bioengineering (Basel) 2021;8:108. https://doi.org/10.3390/bioengineering8080108.Search in Google Scholar PubMed PubMed Central

21. Baldari, S, Di Rocco, G, Piccoli, M, Pozzobon, M, Muraca, M, Toietta, G. Challenges and strategies for improving the regenerative effects of mesenchymal stromal cell-based therapies. Int J Mol Sci 2017;18:2087. https://doi.org/10.3390/IJMS18102087.Search in Google Scholar PubMed PubMed Central

22. Lindley, LE, Stojadinovic, O, Pastar, I, Tomic-Canic, M. Biology and biomarkers for wound healing. Plast Reconstr Surg 2016;138:18S–28S. https://doi.org/10.1097/PRS.0000000000002682.Search in Google Scholar PubMed PubMed Central

23. LaPelusa, A, Dave, HD. Physiology, Hemostasis. [Updated on: May 1, 2023]. In: StatPearls; 2019. http://europepmc.org/books/NBK545263 [Accessed 27 Jul 2024].Search in Google Scholar

24. Feletou, M. Chapter 4, Endothelium-dependent regulation of vascular tone. In: The endothelium: Part 1: Multiple functions of the endothelial cells—Focus on endothelium-derived vasoactive mediators. San Rafael (CA): Morgan & Claypool Life Sciences; 2011. https://www.ncbi.nlm.nih.gov/books/NBK57147/ [Accessed 27 Jul 2024].10.4199/C00031ED1V01Y201105ISP019Search in Google Scholar PubMed

25. Periayah, MH, Halim, AS, Saad, AZM. Mechanism action of platelets and crucial blood coagulation pathways in hemostasis. Int J Hematol Oncol Stem Cell Res 2017;11:319–27.Search in Google Scholar

26. Chaudhry, R, Usama, SM, Babiker, HM. Physiology, coagulation pathways. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2018. http://europepmc.org/books/NBK482253 [Accessed 28 Jul 2024].Search in Google Scholar

27. Grover, SP, Mackman, N. Intrinsic pathway of coagulation and thrombosis. Arterioscler Thromb Vasc Biol 2019;39:331–8. https://doi.org/10.1161/ATVBAHA.118.312130.Search in Google Scholar PubMed

28. Göbel, K, Eichler, S, Wiendl, H, Chavakis, T, Kleinschnitz, C, Meuth, SG. The coagulation factors fibrinogen, thrombin, and factor XII in inflammatory disorders-a systematic review. Front Immunol 2018;9:1731. https://doi.org/10.3389/fimmu.2018.01731.Search in Google Scholar PubMed PubMed Central

29. Wilhelm, G, Mertowska, P, Mertowski, S, Przysucha, A, Strużyna, J, Grywalska, E, et al.. The crossroads of the coagulation system and the immune system: interactions and connections. Int J Mol Sci 2023;24:12563. https://doi.org/10.3390/ijms241612563.Search in Google Scholar PubMed PubMed Central

30. Rodrigues, M, Kosaric, N, Bonham, CA, Gurtner, GC. Wound healing: a cellular perspective. Physiol Rev 2019;99:665–706. https://doi.org/10.1152/physrev.00067.2017.Search in Google Scholar PubMed PubMed Central

31. Chen, L, DiPietro, LA. Toll-like receptor function in acute wounds. Adv Wound Care (New Rochelle) 2017;6:344–55. https://doi.org/10.1089/wound.2017.0734.10.1089/wound.2017.0734Search in Google Scholar PubMed PubMed Central

32. Dunnill, C, Patton, T, Brennan, J, Barrett, J, Dryden, M, Cooke, J, et al.. Reactive oxygen species (ROS) and wound healing: the functional role of ROS and emerging ROS-modulating technologies for augmentation of the healing process. Int Wound J 2017;14:89–96. https://doi.org/10.1111/iwj.12557.Search in Google Scholar PubMed PubMed Central

33. Hughes, CE, Nibbs, RJB. A guide to chemokines and their receptors. FEBS J 2018;285:2944–71. https://doi.org/10.1111/febs.14466.Search in Google Scholar PubMed PubMed Central

34. Fine, N, Tasevski, N, McCulloch, CA, Tenenbaum, HC, Glogauer, M. The neutrophil: constant defender and first responder. Front Immunol 2020;11:571085. https://doi.org/10.3389/fimmu.2020.571085.Search in Google Scholar PubMed PubMed Central

35. Vorobjeva, NV, Chernyak, BV. NETosis: molecular mechanisms, role in physiology and pathology. Biochemistry (Mosc) 2020;85:1178–90. https://doi.org/10.1134/S0006297920100065.Search in Google Scholar PubMed PubMed Central

36. Jorch, SK, Kubes, P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat Med 2017;23:279–87. https://doi.org/10.1038/nm.4294.Search in Google Scholar PubMed

37. Manfredi, AA, Ramirez, GA, Rovere-Querini, P, Maugeri, N. The neutrophil’s choice: phagocytose vs make neutrophil extracellular traps. Front Immunol 2018;9:288. https://doi.org/10.3389/fimmu.2018.00288.Search in Google Scholar PubMed PubMed Central

38. Singhal, A, Kumar, S. Neutrophil and remnant clearance in immunity and inflammation. Immunology 2022;165:22–43. https://doi.org/10.1111/imm.13423.Search in Google Scholar PubMed

39. Chen, Y, Jiang, W, Yong, H, He, M, Yang, Y, Deng, Z, et al.. Macrophages in osteoarthritis: pathophysiology and therapeutics. Am J Transl Res 2020;12:261–68.Search in Google Scholar

40. Cavinato, L, Genise, E, Luly, FR, Di Domenico, EG, Del Porto, P, Ascenzioni, F. Escaping the phagocytic oxidative burst: the role of SODB in the survival of Pseudomonas aeruginosa within macrophages. Front Microbiol 2020;11:326. https://doi.org/10.3389/fmicb.2020.00326.Search in Google Scholar PubMed PubMed Central

41. Landén, NX, Li, D, Ståhle, M. Transition from inflammation to proliferation: a critical step during wound healing. Cell Mol Life Sci 2016;73:3861–85. https://doi.org/10.1007/s00018-016-2268-0.Search in Google Scholar PubMed PubMed Central

42. Shaw, TJ, Martin, P. Wound repair: a showcase for cell plasticity and migration. Curr Opin Cell Biol 2016;42:29–37. https://doi.org/10.1016/j.ceb.2016.04.001.Search in Google Scholar PubMed

43. Krausz, A, Friedman, AJ. Nitric oxide as a surgical adjuvant. Future Sci OA 2015;1:FSO56. https://doi.org/10.4155/fso.15.56.Search in Google Scholar PubMed PubMed Central

44. DiPietro, LA. Angiogenesis and wound repair: when enough is enough. J Leukoc Biol 2016;100:979–84. https://doi.org/10.1189/jlb.4MR0316-102R.Search in Google Scholar PubMed PubMed Central

45. Potekaev, NN, Borzykh, OB, Medvedev, GV, Pushkin, DV, Petrova, MM, Petrov, AV, et al.. The role of extracellular matrix in skin wound healing. J Clin Med 2021;10:5947. https://doi.org/10.3390/jcm10245947.Search in Google Scholar PubMed PubMed Central

46. Xue, M, Jackson, CJ. Extracellular matrix reorganization during wound healing and its impact on abnormal scarring. Adv Wound Care 2015;4:119–36. https://doi.org/10.1089/wound.2013.0485.Search in Google Scholar PubMed PubMed Central

47. Mathew-Steiner, SS, Roy, S, Sen, CK. Collagen in wound healing. Bioengineering (Basel) 2021;8:63. https://doi.org/10.3390/BIOENGINEERING8050063.Search in Google Scholar

48. Hu, MS, Maan, ZN, Wu, JC, Rennert, RC, Hong, WX, Lai, TS, et al.. Tissue engineering and regenerative repair in wound healing. Ann Biomed Eng 2014;42:1494–507. https://doi.org/10.1007/s10439-014-1010-z.Search in Google Scholar PubMed PubMed Central

49. You, HJ, Han, SK. Cell therapy for wound healing. J Kor Med Sci 2014;29:311–19. https://doi.org/10.3346/jkms.2014.29.3.311.Search in Google Scholar PubMed PubMed Central

50. Kim, Y, Kim, I, Shin, K. A new era of stem cell and developmental biology: from blastoids to synthetic embryos and beyond. Exp Mol Med 2023;55:2127–37. https://doi.org/10.1038/s12276-023-01097-8.Search in Google Scholar PubMed PubMed Central

51. Kosaric, N, Kiwanuka, H, Gurtner, GC. Stem cell therapies for wound healing. Expet Opin Biol Ther 2019;19:575–85. https://doi.org/10.1080/14712598.2019.1596257.Search in Google Scholar PubMed

52. Poliwoda, S, Noor, N, Downs, E, Schaaf, A, Cantwell, A, Ganti, L, et al.. Stem cells: a comprehensive review of origins and emerging clinical roles in medical practice. Orthop Rev 2022;14:37498. https://doi.org/10.52965/001C.37498.Search in Google Scholar PubMed PubMed Central

53. Duscher, D, Barrera, J, Wong, VW, Maan, ZN, Whittam, AJ, Januszyk, M, et al.. Stem cells in wound healing: the future of regenerative medicine? A Mini-Review. Gerontology 2016;62:216–25. https://doi.org/10.1159/000381877.Search in Google Scholar

54. Zhao, Y, Wang, M, Liang, F, Li, J. Recent strategies for enhancing the therapeutic efficacy of stem cells in wound healing. Stem Cell Res Ther 2021;12:588. https://doi.org/10.1186/s13287-021-02657-3.Search in Google Scholar PubMed PubMed Central

55. Zhou, J, Shi, Y. Mesenchymal stem/stromal cells (MSCs): origin, immune regulation, and clinical applications. Cell Mol Immunol 2023;20:555–57. https://doi.org/10.1038/s41423-023-01034-9.Search in Google Scholar PubMed PubMed Central

56. Yolanda, MM, Maria, AV, Amaia, FG, Marcos, PB, Silvia, PL, Dolores, E, et al.. Adult stem cell therapy in chronic wound healing. J Stem Cell Res Ther 2014;4:1–6. https://doi.org/10.4172/2157-7633.1000162.Search in Google Scholar

57. Donnelly, H, Salmeron-Sanchez, M, Dalby, MJ. Designing stem cell niches for differentiation and self-renewal. J R Soc Interface 2018;15:20180388. https://doi.org/10.1098/rsif.2018.0388.Search in Google Scholar PubMed PubMed Central

58. Teo, AKK, Vallier, L. Emerging use of stem cells in regenerative medicine. Biochem J 2010;428:11–23. https://doi.org/10.1042/BJ20100102.Search in Google Scholar PubMed

59. Xia, H, Li, X, Gao, W, Fu, X, Fang, RH, Zhang, L, et al.. Tissue repair and regeneration with endogenous stem cells. Nat Rev Mater 2018;3:174–93. https://doi.org/10.1038/s41578-018-0027-6.Search in Google Scholar

60. Squillaro, T, Peluso, G, Galderisi, U. Clinical trials with mesenchymal stem cells: an update. Cell Transplant 2016;25:829–48. https://doi.org/10.3727/096368915X689622.Search in Google Scholar PubMed

61. Ding, DC, Shyu, WC, Lin, SZ. Mesenchymal stem cells. Cell Transplant 2011;20:5–14. https://doi.org/10.3727/096368910X.Search in Google Scholar PubMed

62. Jo, H, Brito, S, Kwak, BM, Park, S, Lee, MG, Bin, BH. Applications of mesenchymal stem cells in skin regeneration and rejuvenation. Int J Mol Sci 2021;22:2410. https://doi.org/10.3390/ijms22052410.Search in Google Scholar PubMed PubMed Central

63. Viswanathan, S, Shi, Y, Galipeau, J, Krampera, M, Leblanc, K, Martin, I, et al.. Mesenchymal stem versus stromal cells: international society for cell & gene therapy (ISCT®) mesenchymal stromal cell committee position statement on nomenclature. Cytotherapy 2019;21:1019–24. https://doi.org/10.1016/j.jcyt.2019.08.002.Search in Google Scholar PubMed

64. Beeravolu, N, McKee, C, Alamri, A, Mikhael, S, Brown, C, Perez-Cruet, M, et al.. Isolation and characterization of mesenchymal stromal cells from human umbilical cord and fetal placenta. J Vis Exp 2017;2017:55224. https://doi.org/10.3791/55224.Search in Google Scholar PubMed PubMed Central

65. Hmadcha, A, Martin-Montalvo, A, Gauthier, BR, Soria, B, Capilla-Gonzalez, V. Therapeutic potential of mesenchymal stem cells for cancer therapy. Front Bioeng Biotechnol 2020;8:43. https://doi.org/10.3389/fbioe.2020.00043.Search in Google Scholar PubMed PubMed Central

66. Giacomini, C, Granéli, C, Hicks, R, Dazzi, F. The critical role of apoptosis in mesenchymal stromal cell therapeutics and implications in homeostasis and normal tissue repair. Cell Mol Immunol 2023;20:570–82. https://doi.org/10.1038/s41423-023-01018-9.Search in Google Scholar PubMed PubMed Central

67. Davies, BM, Snelling, SJB, Quek, L, Hakimi, O, Ye, H, Carr, A, et al.. Identifying the optimum source of mesenchymal stem cells for use in knee surgery. J Orthop Res 2017;35:1868–75. https://doi.org/10.1002/jor.23501.Search in Google Scholar PubMed

68. Vasiliadis, AV, Galanis, N. Human bone marrow-derived mesenchymal stem cells from different bone sources: a panorama. Stem Cell Invest 2020;7:15. https://doi.org/10.21037/SCI-2020-013.Search in Google Scholar PubMed PubMed Central

69. Jia, Q, Zhao, H, Wang, Y, Cen, Y, Zhang, Z. Mechanisms and applications of adipose-derived stem cell-extracellular vesicles in the inflammation of wound healing. Front Immunol 2023;14:1214757. https://doi.org/10.3389/fimmu.2023.1214757.Search in Google Scholar PubMed PubMed Central

70. Wang, M, Zhao, J, Li, J, Meng, M, Zhu, M. Insights into the role of adipose-derived stem cells and secretome: potential biology and clinical applications in hypertrophic scarring. Stem Cell Res Ther 2024;15:137. https://doi.org/10.1186/s13287-024-03749-6.Search in Google Scholar PubMed PubMed Central

71. Al-Ghadban, S, Bunnell, BA. Adipose tissue-derived stem cells: immunomodulatory effects and therapeutic potential. Physiology (Bethesda) 2020;35:125–33. https://doi.org/10.1152/physiol.00021.2019.Search in Google Scholar PubMed

72. Bunnell, BA. Adipose tissue-derived mesenchymal stem cells. Cells 2021;10:3433. https://doi.org/10.3390/CELLS10123433.Search in Google Scholar PubMed PubMed Central

73. Surowiecka, A, Strużyna, J. Adipose-derived stem cells for facial rejuvenation. J Pers Med 2022;12:117. https://doi.org/10.3390/JPM12010117.Search in Google Scholar PubMed PubMed Central

74. Schneider, I, Calcagni, M, Buschmann, J. Adipose-derived stem cells applied in skin diseases, wound healing and skin defects: a review. Cytotherapy 2023;25:105–19. https://doi.org/10.1016/j.jcyt.2022.08.005.Search in Google Scholar PubMed

75. Díaz-García, D, Filipová, A, Garza-Veloz, I, Martinez-Fierro, ML. A beginner’s introduction to skin stem cells and wound healing. Int J Mol Sci 2021;22:11030. https://doi.org/10.3390/ijms222011030.Search in Google Scholar PubMed PubMed Central

76. Chu, GY, Chen, YF, Chen, HY, Chan, MH, Gau, CS, Weng, SM. Stem cell therapy on skin: mechanisms, recent advances and drug reviewing issues. J Food Drug Anal 2018;26:14–20. https://doi.org/10.1016/j.jfda.2017.10.004.Search in Google Scholar PubMed PubMed Central

77. Koczkowska, M, Kostecka, A, Zawrzykraj, M, Myszczyński, K, Skoniecka, A, Deptuła, M, et al.. Identifying differentiation markers between dermal fibroblasts and adipose-derived mesenchymal stromal cells (AD-MSCs) in human visceral and subcutaneous tissues using single-cell transcriptomics. Stem Cell Res Ther 2025;16:64. https://doi.org/10.1186/s13287-025-04185-w.Search in Google Scholar PubMed PubMed Central

78. Ng, AP, Alexander, WS. Haematopoietic stem cells: past, present and future. Cell Death Discov 2017;3:17002. https://doi.org/10.1038/cddiscovery.2017.2.Search in Google Scholar PubMed PubMed Central

79. Moon, SY, Park, YB, Kim, DS, Oh, SK, Kim, DW. Generation, culture, and differentiation of human embryonic stem cells for therapeutic applications. Mol Ther 2005;13:5–14. https://doi.org/10.1016/j.ymthe.2005.09.008.Search in Google Scholar PubMed

80. The safety and tolerability of sub-retinal transplantation of SCNT-hES-RPE cells in patients with advanced dry AMD (ClinicalTrials.gov). https://clinicaltrials.gov/study/NCT03305029 [Accessed 22 Aug 2024].Search in Google Scholar

81. Safety and efficacy study of OpRegen for treatment of advanced dry-form age-related macular degeneration (ClinicalTrials.gov). https://clinicaltrials.gov/study/NCT02286089 [Accessed 22 Aug 2024].Search in Google Scholar

82. Tsugata, T, Nikoh, N, Kin, T, Saitoh, I, Noguchi, Y, Ueki, H, et al.. Potential factors for the differentiation of ESCs/iPSCs into insulin-producing cells. Cell Med 2014;7:83–93. https://doi.org/10.3727/215517914X685178.Search in Google Scholar PubMed PubMed Central

83. Suaudeau, J. From embryonic stem cells to iPS - an ethical perspective. Cell Prolif 2011;44:70–84. https://doi.org/10.1111/j.1365-2184.2010.00723.x.Search in Google Scholar PubMed PubMed Central

84. Hentze, H, Soong, PL, Wang, ST, Phillips, BW, Putti, TC, Dunn, NR. Teratoma formation by human embryonic stem cells: evaluation of essential parameters for future safety studies. Stem Cell Res 2009;2:198–210. https://doi.org/10.1016/j.scr.2009.02.002.Search in Google Scholar PubMed

85. Sachs, PC, Mollica, PA, Bruno, RD. Tissue specific microenvironments: a key tool for tissue engineering and regenerative medicine. J Biol Eng 2017;11:34. https://doi.org/10.1186/s13036-017-0077-0.Search in Google Scholar PubMed PubMed Central

86. Aboul-Soud, MAM, Alzahrani, AJ, Mahmoud, A. Induced pluripotent stem cells (iPSCs)–roles in regenerative therapies, disease modelling and drug screening. Cells 2021;10:2319. https://doi.org/10.3390/CELLS10092319.Search in Google Scholar PubMed PubMed Central

87. Jiang, Z, Han, Y, Cao, X. Induced pluripotent stem cell (iPSCs) and their application in immunotherapy. Cell Mol Immunol 2014;11:17–24. https://doi.org/10.1038/cmi.2013.62.Search in Google Scholar PubMed PubMed Central

88. Singh, VK, Kalsan, M, Kumar, N, Saini, A, Chandra, R. Induced pluripotent stem cells: applications in regenerative medicine, disease modeling, and drug discovery. Front Cell Dev Biol 2015;3:2. https://doi.org/10.3389/FCELL.2015.00002.Search in Google Scholar

89. Sun, T, Zhou, C, Lu, F, Dong, Z, Gao, J, Li, B. Adipose-derived stem cells in immune-related skin disease: a review of current research and underlying mechanisms. Stem Cell Res Ther 2024;15:37. https://doi.org/10.1186/s13287-023-03561-8.Search in Google Scholar PubMed PubMed Central

90. Gumede, DB, Abrahamse, H, Houreld, NN. Targeting Wnt/β-catenin signaling and its interplay with TGF-β and Notch signaling pathways for the treatment of chronic wounds. Cell Commun Signal 2024;22:244. https://doi.org/10.1186/s12964-024-01623-910.1186/s12964-024-01623-9.Search in Google Scholar

91. Koike, Y, Yozaki, M, Utani, A, Murota, H. Fibroblast growth factor 2 accelerates the epithelial-mesenchymal transition in keratinocytes during wound healing process. Sci Rep 2020;10:18545. https://doi.org/10.1038/s41598-020-75584-7.Search in Google Scholar PubMed PubMed Central

92. Phinney, DG. Functional heterogeneity of mesenchymal stem cells: implications for cell therapy. J Cell Biochem 2012;113:2806–12. https://doi.org/10.1002/jcb.24166.Search in Google Scholar PubMed

93. Aggarwal, S, Pittenger, MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005;105:1815–22. https://doi.org/10.1182/blood-2004-04-1559.Search in Google Scholar PubMed

94. Ren, G, Zhang, L, Zhao, X, Xu, G, Zhang, Y, Roberts, AI, et al.. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell 2008;2:141–50. https://doi.org/10.1016/j.stem.2007.11.014.Search in Google Scholar PubMed

95. Caplan, AI, Dennis, JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem 2006;98:1076–84. https://doi.org/10.1002/jcb.20886.Search in Google Scholar PubMed

96. Ayavoo, T, Murugesan, K, Gnanasekaran, A. Roles and mechanisms of stem cell in wound healing. Stem Cell Invest 2021;8:4. https://doi.org/10.21037/SCI-2020-027.Search in Google Scholar

97. Mei, SHJ, Haitsma, JJ, Dos Santos, CC, Deng, Y, Lai, PF, Slutsky, AS, et al.. Mesenchymal stem cells reduce inflammation while enhancing bacterial clearance and improving survival in sepsis. Am J Respir Crit Care Med 2012;182:1047–57. https://doi.org/101164/rccm201001-0010OC.10.1164/rccm.201001-0010OCSearch in Google Scholar PubMed

98. Hu, MS-M, Rennert, RC, McArdle, A, Chung, MT, Walmsley, GG, Longaker, MT, et al.. The role of stem cells during scarless skin wound healing. Adv Wound Care 2014;3:304–14. https://doi.org/10.1089/wound.2013.0471.Search in Google Scholar PubMed PubMed Central

99. Rustad, KC, Wong, VW, Sorkin, M, Glotzbach, JP, Major, MR, Rajadas, J, et al.. Enhancement of mesenchymal stem cell angiogenic capacity and stemness by a biomimetic hydrogel scaffold. Biomaterials 2012;33:80–90. https://doi.org/10.1016/j.biomaterials.2011.09.041.Search in Google Scholar PubMed PubMed Central

100. Farabi, B, Roster, K, Hirani, R, Tepper, K, Atak, MF, Safai, B. The efficacy of stem cells in wound healing: a systematic review. Int J Mol Sci 2024;25:3006. https://doi.org/10.3390/IJMS25053006/S1.Search in Google Scholar

101. Sen, CK, Gordillo, GM, Roy, S, Kirsner, R, Lambert, L, Hunt, TK, et al.. Human skin wounds: a major and snowballing threat to public health and the economy. Wound Repair Regen 2009;17:763–71. https://doi.org/10.1111/j.1524-475X.2009.00543.x.Search in Google Scholar PubMed PubMed Central

102. Garg, RK, Rennert, RC, Duscher, D, Sorkin, M, Kosaraju, R, Auerbach, LJ, et al.. Capillary force seeding of hydrogels for adipose-derived stem cell delivery in wounds. Stem Cells Transl Med 2014;3:1079–89. https://doi.org/10.5966/sctm.2014-0007.Search in Google Scholar PubMed PubMed Central

103. Duscher, D, Barrera, J, Wong, VW, Maan, ZN, Whittam, AJ, Januszyk, M, et al.. Stem cells in wound healing: the future of regenerative medicine? A mini-review. Gerontology 2016;62:216–25. https://doi.org/10.1159/000381877.Search in Google Scholar PubMed

104. Mukherjee, S, Yadav, G, Kumar, R. Recent trends in stem cell-based therapies and applications of artificial intelligence in regenerative medicine. World J Stem Cells 2021;13:521–41. https://doi.org/10.4252/wjsc.v13.i6.521.Search in Google Scholar PubMed PubMed Central

105. Nawab, K, Bhere, D, Bommarito, A, Mufti, M, Naeem, A. Stem cell therapies: a way to promising cures. Cureus 2019;11:e5712. https://doi.org/10.7759/CUREUS.5712.Search in Google Scholar

106. Li, C, Zhao, H, Cheng, L, Wang, B. Allogeneic vs. autologous mesenchymal stem/stromal cells in their medication practice. Cell Biosci 2021;11:187. https://doi.org/10.1186/s13578-021-00698-y.Search in Google Scholar PubMed PubMed Central

107. Fallah, N, Rasouli, M, Amini, MR. The current and advanced therapeutic modalities for wound healing management. J Diabetes Metab Disord 2021;20:1883–99. https://doi.org/10.1007/s40200-021-00868-2.Search in Google Scholar PubMed PubMed Central

108. Mirhaj, M, Labbaf, S, Tavakoli, M, Seifalian, AM. Emerging treatment strategies in wound care. Int Wound J 2022;19:1934–54. https://doi.org/10.1111/iwj.13786.Search in Google Scholar PubMed PubMed Central

109. Varaprasad, K, Jayaramudu, T, Kanikireddy, V, Toro, C, Sadiku, ER. Alginate-based composite materials for wound dressing application: a mini review. Carbohydr Polym 2020;236:116025. https://doi.org/10.1016/j.carbpol.2020.116025.Search in Google Scholar PubMed

110. Wilcox, JR, Carter, MJ, Covington, S. Frequency of debridements and time to heal: a retrospective cohort study of 312 744 wounds. JAMA Dermatol 2013;149:1050–8. https://doi.org/10.1001/jamadermatol.2013.4960.Search in Google Scholar PubMed

111. Matusiak, D, Wichtowski, M, Pieszko, K, Kobylarek, D, Murawa, D. Is negative-pressure wound therapy beneficial in modern-day breast surgery? Contemp Oncol/Współcz Onkol 2019;23:69–73. https://doi.org/10.5114/wo.2019.85199.Search in Google Scholar PubMed PubMed Central

112. Skrepnek, GH, Mills, JL, Lavery, LA, Armstrong, DG. Health care service and outcomes among an estimated 6.7 million ambulatory care diabetic foot cases in the U.S. Diabetes Care 2017;40:936–42. https://doi.org/10.2337/dc16-2189.Search in Google Scholar PubMed

113. Margolis, DJ, Gupta, J, Hoffstad, O, Papdopoulos, M, Glick, HA, Thom, SR, et al.. Lack of effectiveness of hyperbaric oxygen therapy for the treatment of diabetic foot ulcer and the prevention of amputation: a cohort study. Diabetes Care 2013;36:1961–6. https://doi.org/10.2337/dc12-2160.Search in Google Scholar PubMed PubMed Central

114. Miron, RJ, Zhang, Y. Autologous liquid platelet rich fibrin: a novel drug delivery system. Acta Biomater 2018;75:35–51. https://doi.org/10.1016/j.actbio.2018.05.021.Search in Google Scholar PubMed

115. Wu, J, Chen, LH, Sun, SY, Li, Y, Ran, XW. Mesenchymal stem cell-derived exosomes: the dawn of diabetic wound healing. World J Diabetes 2022;13:1066–95. https://doi.org/10.4239/wjd.v13.i12.1066.Search in Google Scholar PubMed PubMed Central

116. Kalou, Y, Al-Khani, AM, Haider, KH. Bone marrow mesenchymal stem cells for heart failure treatment: a systematic review and meta-analysis. Heart Lung Circ 2023;32:870–80. https://doi.org/10.1016/j.hlc.2023.01.012.Search in Google Scholar PubMed

117. Liu, S, Zhou, J, Zhang, X, Liu, Y, Chen, J, Hu, B, et al.. Strategies to optimize adult stem cell therapy for tissue regeneration. Int J Mol Sci 2016;17:982. https://doi.org/10.3390/ijms17060982.Search in Google Scholar PubMed PubMed Central

118. Jun, EK, Zhang, Q, Yoon, BS, Moon, JH, Lee, G, Park, G, et al.. Hypoxic conditioned medium from human amniotic fluid-derived mesenchymal stem cells accelerates skin wound healing through TGF-β/SMAD2 and PI3K/Akt pathways. Int J Mol Sci 2014;15:605–28. https://doi.org/10.3390/ijms15010605.Search in Google Scholar PubMed PubMed Central

119. Sart, S, Tsai, AC, Li, Y, Ma, T. Three-dimensional aggregates of mesenchymal stem cells: cellular mechanisms, biological properties, and applications. Tissue Eng Part B Rev 2014;20:365–80. https://doi.org/10.1089/ten.TEB.2013.0537.Search in Google Scholar PubMed PubMed Central

120. Song, SH, Lee, MO, Lee, JS, Jeong, HC, Kim, HG, Kim, WS, et al.. Genetic modification of human adipose-derived stem cells for promoting wound healing. J Dermatol Sci 2012;66:98–107. https://doi.org/10.1016/j.jdermsci.2012.02.010.Search in Google Scholar PubMed

121. Rodriguez, LA, Mohammadipoor, A, Alvarado, L, Kamucheka, RM, Asher, AM, Cancio, LC, et al.. Preconditioning in an inflammatory milieu augments the immunotherapeutic function of mesenchymal stromal cells. Cells 2019;8:462. https://doi.org/10.3390/CELLS8050462.Search in Google Scholar PubMed PubMed Central

122. Pang, L, Ding, J, Liu, XX, Kou, Z, Guo, L, Xu, X, et al.. Microfluidics-based single-cell research for intercellular interaction. Front Cell Dev Biol 2021;9:680307. https://doi.org/10.3389/fcell.2021.680307.Search in Google Scholar PubMed PubMed Central

123. Cheng, YH, Chen, YC, Lin, E, Brien, R, Jung, S, Chen, YT, et al.. Hydro-Seq enables contamination-free high-throughput single-cell RNA-sequencing for circulating tumor cells. Nat Commun 2019;10:2163. https://doi.org/10.1038/s41467-019-10122-2.Search in Google Scholar PubMed PubMed Central

124. Pang, L, Ding, J, Liu, XX, Yuan, H, Ge, Y, Fan, J, et al.. Microstructure-based techniques for single-cell manipulation and analysis. TrAC, Trends Anal Chem 2020;129:115940. https://doi.org/10.1016/j.trac.2020.115940.Search in Google Scholar

125. Gupta, N, Liu, JR, Patel, B, Solomon, DE, Vaidya, B, Gupta, V. Microfluidics-based 3D cell culture models: Utility in novel drug discovery and delivery research. Bioeng Transl Med 2016;1:63–81. https://doi.org/10.1002/btm2.10013.Search in Google Scholar PubMed PubMed Central

126. Jakovija, A, Chtanova, T. Skin immunity in wound healing and cancer. Front Immunol 2023;14:1060258. https://doi.org/10.3389/fimmu.2023.1060258.Search in Google Scholar PubMed PubMed Central

127. Diller, RB, Tabor, AJ. The role of the extracellular matrix (ECM) in wound healing: a review. Biomimetics (Basel) 2022;7:87. https://doi.org/10.3390/biomimetics7030087.Search in Google Scholar PubMed PubMed Central

128. Kolimi, P, Narala, S, Nyavanandi, D, Youssef, AAA, Dudhipala, N. Innovative treatment strategies to accelerate wound healing: trajectory and recent advancements. Cells 2022;11:2439. https://doi.org/10.3390/CELLS11152439.Search in Google Scholar

129. Sharpless, NE, DePinho, RA. How stem cells age and why this makes us grow old. Nat Rev Mol Cell Biol 2007;8:703–13. https://doi.org/10.1038/nrm2241.Search in Google Scholar PubMed

130. Koenen, P, Spanholtz, TA, Maegele, M, Stürmer, E, Brockamp, T, Neugebauer, E, et al.. Acute and chronic wound fluids inversely influence adipose-derived stem cell function: molecular insights into impaired wound healing. Int Wound J 2015;12:10–16. https://doi.org/10.1111/iwj.12039.Search in Google Scholar PubMed PubMed Central

131. Hoang, DM, Pham, PT, Bach, TQ, Ngo, ATL, Nguyen, QT, Phan, TTK, et al.. Stem cell-based therapy for human diseases. Signal Transduct Target Ther 2022;7:272. https://doi.org/10.1038/S41392-022-01134-4.Search in Google Scholar

132. Jahani, M, Rezazadeh, D, Mohammadi, P, Abdolmaleki, A, Norooznezhad, A, Mansouri, K. Regenerative medicine and angiogenesis; challenges and opportunities. Adv Pharmaceut Bull 2020;10:490–501. https://doi.org/10.34172/apb.2020.061.Search in Google Scholar PubMed PubMed Central

133. Bedel, A, Beliveau, F, Lamrissi-Garcia, I, Rousseau, B, Moranvillier, I, Rucheton, B, et al.. Preventing pluripotent cell teratoma in regenerative medicine applied to hematology disorders. Stem Cells Transl Med 2016;6:382–93. https://doi.org/10.5966/sctm.2016-0201.Search in Google Scholar PubMed PubMed Central

134. Amariglio, N, Hirshberg, A, Scheithauer, BW, Cohen, Y, Loewenthal, R, Trakhtenbrot, L, et al.. Donor-derived brain tumor following neural stem cell transplantation in an ataxia telangiectasia patient. PLoS Med 2009;6:e1000029. https://doi.org/10.1371/journal.pmed.1000029.Search in Google Scholar PubMed PubMed Central

135. Yael, D. Addressing the high costs of stem cell therapies. Stem Cell Res Regen Med 2024;7:255–6. https://doi.org/10.37532/SRRM.2024.7(5).255-256.Search in Google Scholar

136. Mousaei Ghasroldasht, M, Seok, J, Park, HS, Liakath Ali, FB, Al-Hendy, A. Stem cell therapy: from idea to clinical practice. Int J Mol Sci 2022;23:2850. https://doi.org/10.3390/IJMS23052850.Search in Google Scholar

137. Boopalan, D, Pandian, R, Kesavan, G. Prospects for stem cell-based regenerative therapies in India. Stem J 2021;3:11–21. https://doi.org/10.3233/STJ-210002.Search in Google Scholar

138. Fisher, SA, Doree, C, Mathur, A, Taggart, DP, Martin-Rendon, E. Stem cell therapy for chronic ischaemic heart disease and congestive heart failure. Cochrane Database Syst Rev 2016;12:CD007888. https://doi.org/10.1002/14651858.CD007888.pub3.Search in Google Scholar PubMed PubMed Central

139. Chen, S, Du, K, Zou, C. Current progress in stem cell therapy for type 1 diabetes mellitus. Stem Cell Res Ther 2020;11:275. https://doi.org/10.1186/s13287-020-01793-6.Search in Google Scholar PubMed PubMed Central

140. Xue, R, Meng, Q, Dong, J, Li, J, Yao, Q, Zhu, Y, et al.. Clinical performance of stem cell therapy in patients with acute-on-chronic liver failure: a systematic review and meta-analysis. J Transl Med 2018;16:126. https://doi.org/10.1186/s12967-018-1464-0.Search in Google Scholar PubMed PubMed Central

141. Zhao, D, Cui, D, Wang, B, Tian, F, Guo, L, Yang, L, et al.. Treatment of early stage osteonecrosis of the femoral head with autologous implantation of bone marrow-derived and cultured mesenchymal stem cells. Bone 2012;50:325–30. https://doi.org/10.1016/j.bone.2011.11.002.Search in Google Scholar PubMed

142. Widuchowski, W, Widuchowski, J, Trzaska, T. Articular cartilage defects: study of 25,124 knee arthroscopies. Knee 2007;14:177–82. https://doi.org/10.1016/j.knee.2007.02.001.Search in Google Scholar PubMed

143. Sivandzade, F, Cucullo, L. Regenerative stem cell therapy for neurodegenerative diseases: an overview. Int J Mol Sci 2021;22:2153. https://doi.org/10.3390/ijms22042153.Search in Google Scholar PubMed PubMed Central

144. Patel, SA, King, CC, Lim, PK, Habiba, U, Dave, M, Porecha, R, et al.. Personalizing stem cell research and therapy: the arduous road ahead or missed opportunity? Curr Pharmacogenomics Person Med 2010;8:25–36. https://doi.org/10.2174/1875692111008010025.Search in Google Scholar PubMed PubMed Central

145. Huerta, CT, Ortiz, YY, Li, Y, Ribieras, AJ, Voza, F, Le, N, et al.. Novel gene-modified mesenchymal stem cell therapy reverses impaired wound healing in ischemic limbs. Ann Surg 2023;278:383–95.10.1097/SLA.0000000000005949Search in Google Scholar PubMed PubMed Central

146. Hong, IS. Enhancing stem cell-based therapeutic potential by combining various bioengineering technologies. Front Cell Dev Biol 2022;10:901661. https://doi.org/10.3389/fcell.2022.901661.Search in Google Scholar PubMed PubMed Central

147. Li, Q, Wang, D, Jiang, Z, Li, R, Xue, T, Lin, C, et al.. Advances of hydrogel combined with stem cells in promoting chronic wound healing. Front Chem 2022;10:1038839. https://doi.org/10.3389/fchem.2022.1038839.Search in Google Scholar PubMed PubMed Central

148. Isasi, R, Bentzen, HB, Fabbri, M, Fuhr, A, Glover, JC, Mah, N, et al.. Dynamic governance: a new era for consent for stem cell research. Stem Cell Rep 2024;19:1233–41. https://doi.org/10.1016/j.stemcr.2024.07.006.Search in Google Scholar PubMed PubMed Central

149. Volarevic, V, Markovic, BS, Gazdic, M, Volarevic, A, Jovicic, N, Arsenijevic, N, et al.. Ethical and safety issues of stem cell-based therapy. Int J Med Sci 2018;15:36–45. https://doi.org/10.7150/ijms.21666.Search in Google Scholar PubMed PubMed Central

150. King, NM, Perrin, J. Ethical issues in stem cell research and therapy. Stem Cell Res Ther 2014;5:85. https://doi.org/10.1186/scrt474.Search in Google Scholar PubMed PubMed Central

151. 2024 ISSCR annual meeting — international society for stem cell research. https://www.isscr.org/upcoming-programs/isscr-2024 [Accessed 21 Jul 2024].Search in Google Scholar

152. Iltis, AS, Koster, G, Reeves, E, Matthews, KRW. Ethical, legal, regulatory, and policy issues concerning embryoids: a systematic review of the literature. Stem Cell Res Ther 2023;14:209. https://doi.org/10.1186/s13287-023-03448-8.Search in Google Scholar PubMed PubMed Central

Received: 2025-01-16
Accepted: 2025-04-27
Published Online: 2025-05-23

© 2025 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 16.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/znc-2025-0020/html
Scroll to top button