The role of labeled cell therapy with and without scaffold in early excision burn wounds in a rat animal model

Document Type : Original Article

Authors

1 urn and Wound Healing Research Center, Shiraz University of Medical Science, Shiraz, Iran

2 Nour Danesh Institute of Higher Education, Isfahan, Iran

3 Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran

4 Center of Comparative and Experimental Medicine, Shiraz University of Medical Sciences, Shiraz, Iran

Abstract

Objective(s): One of the essential problems in burn therapy is performing the permanent replacement of skin in full and deep thickness injuries. Human Wharton’s Jelly mesenchymal stem cells (HWJMSCs) have a unique combination of prenatal and postnatal properties. Decellularized human amniotic membrane (DHAM) can be used as a scaffold for HWJMSCs-therapy. We aimed to evaluate the quantity and quality of healing in the early excision burn wound dressing with 3-dimensional and 2- dimensional cell cultures.
Materials and Methods: Amniotic and umbilical cords were isolated from the mothers who were candidates for cesarean section. HAM was decellularized using the mechanical and enzymatic method. HWJMSCs were isolated and cultured; cell surface markers were examined for authentication of MSCs and labeled using a viral vector containing the cGFP gene. Burns were created using brass bar in 32 adult male Albino rats and randomly divided into four groups (DHAM+HWJMSCs, injection of HWJMSCs, HWJMSCs was spread on the wound, and DHAM alone). Rats were sacrificed on the 7th and 14th days for pathological examination of the wound. Comparisons between the study groups were made by one-way analysis of variance.
Results: Wound healing process in DHAM+HWJMSCs was much more progressed during the first week in comparison to other groups, and exhibited significant differences in re-epithelialization, formation of granulation tissue, and hemorrhage (P<0.05).
Conclusion: The utility of the amniotic scaffold seeded by the human mesenchymal stem cells is recommended for accelerating the healing process.

Keywords


1. Bloemsma GC, Dokter J, Boxma H, Oen IM. Mortality and causes of death in a burn centre. Burns 2008; 34:1103-1107.
2. Boyce ST, Warden GD. Principles and practices for treatment of cutaneous wounds with cultured skin substitutes. Am J Surg 2002; 183:445-456.
3. Theoret C. Tissue engineering in wound repair: the three “R”s-repair, replace, regenerate. Vet Surg 2009; 38:905-913.
4. Buinewicz B, Rosen B. Acellular cadaveric dermis (AlloDerm): a new alternative for abdominal hernia repair. Ann Plast Surg 2004; 52:188-194.
5. Orgill DP, Straus FH, Lee RC. The use of collagen-gag membranes in reconstructive surgery. Ann N Y Acad Sci 1999; 888:233-248.
6. Hafemann B, Ensslen S, Erdmann C, Niedballa R, Zühlke A, Ghofrani K, et al. Use of a collagen/elastin-membrane for the tissue engineering of dermis. Burns 1999; 25:373-384.
7. Mazlyzam AL, Aminuddin BS, Fuzina NH, Norhayati MM, Fauziah O, Isa MR, et al. Reconstruction of living bilayer human skin equivalent utilizing human fibrin as a scaffold. Burns 2007; 33:355-363.
8. Niknejad H, Peirovi H, Jorjani M, Ahmadiani A, Ghanavi J, Seifalian AM. Properties of the amniotic membrane for potential use in tissue engineering. Eur Cell Mater 2008; 15:88-99.
9. Kesting MR, Wolff KD, Hohlweg-Majert B, Steinstraesser L. The role of allogenic amniotic membrane in burn treatment. J Burn Care Res 2008; 29:907-916.
10. Troyer DL, Weiss ML. Wharton’s jelly-derived cells are a primitive stromal cell population. Stem Cells 2008; 26:591-599.
11. Tsai PC, Fu TW, Chen YM, Ko TL, Chen TH, Shih YH, et al. The therapeutic potential of human umbilical mesenchymal stem cells from wharton’s jelly in the treatment of rat liver fibrosis. Liver Transpl 2009; 15:484-495.
12. Du T, Zou X, Cheng J, Wu S, Zhong L, Ju G, et al. Human wharton’s jelly-derived mesenchymal stromal cells reduce renal fibrosis through induction of native and foreign hepatocyte growth factor synthesis in injured tubular epithelial cells. Stem Cell Res Ther 2013; 4:1-13.
13. Zhang Y, Hao H, Liu J, Fu X, Han W. Repair and regeneration of skin injury by transplanting microparticles mixed with wharton’s jelly and MSCs from the human umbilical cord. Int J Low Extrem Wounds 2012; 11:264-270.
14. Hashemi S, Rafati A. Comparison between human cord blood serum and platelet-rich plasma supplementation for human wharton’s jelly stem cells and dermal fibroblasts culture. Int J Med Res Health Sci 2016; 5:191-196.
15. Pourfath MR, Behzad-Behbahani A, Hashemi SS, Derakhsahnfar A, Taheri MN, Salehi S. Monitoring wound healing of burn in rat model using human wharton’s jelly mesenchymal stem cells containing cGFP integrated by lentiviral vectors. Iran J Basic Med Sci 2018; 21:70-76.
16. Hashemi SS, Mohammadi AA, Kabiri H, Hashempoor MR, Mahmudi M, Amini M, et al. The healing effect of wharton’s jelly stem cells seeded on biological scaffold in chronic skin ulcers: A randomized clinical trial. J Cosmet Dermatol 2019; 18:1961-1967.
17. Sobhanian P, Khorram M, Hashemi S-S, Mohammadi A. Development of nanofibrous collagen-grafted poly (vinyl alcohol)/gelatin/alginate scaffolds as potential skin substitute. Int J Biol Macromol 2019; 130:977-987.
18. Hashemi S-S, Mahmoodi M, Rafati AR, Manafi F, Mehrabani D. The role of human adult peripheral and umbilical cord blood platelet-rich plasma on proliferation and migration of human skin fibroblasts. World J Plast Surg 2017; 6:198-205.
19. Derakhshanfar A, Moayedi J, Hashemi S-S, Vahedi M, Valizadeh A. Comparative study on the effects of heated brass bar and scald methods in experimental skin burn in rat. Comp Clin Path 2019; 28:1381-1385.
20. Meyer TN, Silva ALd. A standard burn model using rats. Acta Cir Bras 1999; 14:1-4.
21. Wong D, Makowska IJ, Weary DM. Rat aversion to isoflurane versus carbon dioxide. Biol Lett 2013; 9:1-4.
22. Mohtasham Amiri Z, Tanideh N, Seddighi A, Mokhtari M, Amini M, Shakouri Partovi A, et al. The effect of lithospermum officinale, silver sulfadiazine and alpha ointments in healing of burn wound injuries in rat. World J Plast Surg 2017; 6:313-318.
23. Gong C, Wu Q, Wang Y, Zhang D, Luo F, Zhao X, et al. A biodegradable hydrogel system containing curcumin encapsulated in micelles for cutaneous wound healing. Biomaterials 2013; 34:6377-6387.
24. Bharti D, Shivakumar SB, Park J-K, Ullah I, Subbarao RB, Park J-S, et al. Comparative analysis of human wharton’s jelly mesenchymal stem cells derived from different parts of the same umbilical cord. Cell Tissue Res 2018; 372:51-65.
25. Rowan MP, Cancio LC, Elster EA, Burmeister DM, Rose LF, Natesan S, et al. Burn wound healing and treatment: review and advancements. Crit Care 2015; 19:1-12.
26. Liu P, Deng Z, Han S, Liu T, Wen N, Lu W, et al. Tissue-engineered skin containing mesenchymal stem cells improves burn wounds. Artif Organs 2008; 32:925-931.
27. Ma Y, Xu Y, Xiao Z, Yang W, Zhang C, Song E, et al. Reconstruction of chemically burned rat corneal surface by bone marrow-derived human mesenchymal stem cells. Stem Cells 2006; 24:315-321.
28. Ye J, Yao K, Kim JC. Mesenchymal stem cell transplantation in a rabbit corneal alkali burn model: engraftment and involvement in wound healing. Eye 2006; 20:482-490.
29. Jayakumar R, Prabaharan M, Sudheesh Kumar PT, Nair SV, Tamura H. Biomaterials based on chitin and chitosan in wound dressing applications. Biotechnol Adv 2011; 29:322-337.
30. Boucard N, Viton C, Agay D, Mari E, Roger T, Chancerelle Y, et al. The use of physical hydrogels of chitosan for skin regeneration following third-degree burns. Biomaterials 2007; 28:3478-3488.
31. Xu H, Ma L, Shi H, Gao C, Han C. Chitosan–hyaluronic acid hybrid film as a novel wound dressing: in vitro and in vivo studies. Polym Adv Technol 2007; 18:869-875.
32. Oyama N, Bhogal B, Carrington P, Gratian M, Black M. Human placental amnion is a novel substrate for detecting autoantibodies in autoimmune bullous diseases by immunoblotting. Br J Dermatol 2003; 148:939-944.
33. Nakamura T, Endo K-I, Cooper LJ, Fullwood NJ, Tanifuji N, Tsuzuki M, et al. The successful culture and autologous transplantation of rabbit oral mucosal epithelial cells on amniotic membrane. Invest Ophthalmol Vis Sci 2003; 44:106-116.
34. Kumar T, Shanmugasundaram N, Babu M. Biocompatible collagen scaffolds from a human amniotic membrane: physicochemical and in vitro culture characteristics. J Biomater Sci Polym Ed 2003; 14:689-706.
35. Solomon A, Wajngarten M, Alviano F, Anteby I, Elchalal U, Pe’er J, et al. Suppression of inflammatory and fibrotic responses in allergic inflammation by the amniotic membrane stromal matrix. Clin Exp Allergy 2005; 35:941-948.
36. Yang L, Shirakata Y, Tokumaru S, Xiuju D, Tohyama M, Hanakawa Y, et al. Living skin equivalents constructed using human amnions as a matrix. J Dermatol Sci 2009; 56:188-195.
37. Kim SS, Song CK, Shon SK, Lee KY, Kim CH, Lee MJ, et al. Effects of human amniotic membrane grafts combined with marrow mesenchymal stem cells on healing of full-thickness skin defects in rabbits. Cell Tissue Res 2009; 336:59-66.
38. Arno AI, Amini-Nik S, Blit PH, Al-Shehab M, Belo C, Herer E, et al. Human wharton’s jelly mesenchymal stem cells promote skin wound healing through paracrine signaling. Stem Cell Res Ther 2014; 5:1-13.
39. Loeffelbein DJ, Rohleder NH, Eddicks M, Baumann CM, Stoeckelhuber M, Wolff K-D, et al. Evaluation of human amniotic membrane as a wound dressing for split-thickness skin-graft donor sites. Biomed Res Int 2014; 2014:1-12.
40. Sanjurjo-Rodríguez C, Díaz-Prado S, Hermida-Gómez T, Fuentes-Boquete I, Blanco FJ. Mesenchymal stem cells from human amniotic membrane.  Perinatal Stem Cells: Springer; 2014. p. 191-198.
41. Sasaki M, Abe R, Fujita Y, Ando S, Inokuma D, Shimizu H. Mesenchymal stem cells are recruited into wounded skin and contribute to wound repair by transdifferentiation into multiple skin cell type. J Immunol 2008; 180:2581-2587.
42. Argôlo Neto N, Del Carlo R, Monteiro B, Nardi N, Chagastelles P, De Brito A, et al. Role of autologous mesenchymal stem cells associated with platelet‐rich plasma on healing of cutaneous wounds in diabetic mice. Clin Exp Dermatol 2012; 37:544-553.
43. Nambu M, Kishimoto S, Nakamura S, Mizuno H, Yanagibayashi S, Yamamoto N, et al. Accelerated wound healing in healing-impaired db/db mice by autologous adipose tissue-derived stromal cells combined with atelocollagen matrix. Ann Plast Surg 2009; 62:317-321.
44. Javazon EH, Keswani SG, Badillo AT, Crombleholme TM, Zoltick PW, Radu AP, et al. Enhanced epithelial gap closure and increased angiogenesis in wounds of diabetic mice treated with adult murine bone marrow stromal progenitor cells. Wound Repair Regen 2007; 15:350-359.