Vol 60, No 2 (2022)
Original paper
Published online: 2022-05-30

open access

Page views 5649
Article views/downloads 1202
Get Citation

Connect on Social Media

Connect on Social Media

Application of adipose mesenchymal stem cell-derived exosomes-loaded β-chitin nanofiber hydrogel for wound healing

Ying Liu12, Yunen Liu34, Yan Zhao5, Mi Wu4, Shun Mao4, Peifang Cong2, Rufei Zou5, Mingxiao Hou34, Hongxu Jin2, Yongli Bao1
Pubmed: 35645038
Folia Histochem Cytobiol 2022;60(2):167-178.

Abstract

Introduction. Clarifying the role and mechanism of exosome gel in wound repair can provide a new effective strategy for wound treatment. Materials and methods. The cellular responses of adipose mesenchymal stem cell-derived exosomes (AMSC-exos) and the wound healing ability of AMSC-exos-loaded β-chitin nanofiber (β-ChNF) hydrogel were studied in vitro in mouse fibroblasts cells (L929) and in vivo in rat skin injury model. The transcriptome and proteome of rat skin were studied with the use of sequenator and LC-MS/MS, respectively. Results. 80 and 160 μg/mL AMSC-exos could promote the proliferation and migration of mouse fibroblastic cells. Furthermore, AMSC-exos-loaded β-ChNF hydrogel resulted in a significant acceleration rate of wound closure, notably, acceleration of re-epithelialization, and increased collagen expression based on the rat full-thickness skin injury model. The transcriptomics and proteomics studies revealed the changes of the expression of 18 genes, 516 transcripts and 250 proteins. The metabolic pathways, tight junction, NF-κB signaling pathways were enriched in Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway. Complement factor D (CFD) and downstream Aldolase A (Aldoa) and Actn2 proteins in rats treated with AMSC-exos-loaded β-ChNF hydrogel were noticed and further confirmed by ELISA and Western blot. Conclusion. These findings suggested that AMSC-exos-loaded β-ChNF hydrogel could promote wound healing with the mechanism which is related to the effect of AMSC-exos on CFD and downstream proteins.

Article available in PDF format

View PDF Download PDF file

References

  1. Zhang B, Wang M, Gong A, et al. HucMSC-Exosome Mediated-Wnt4 Signaling Is Required for Cutaneous Wound Healing. Stem Cells. 2015; 33(7): 2158–2168.
  2. Li J, Chen L, Xu J, et al. Effects of Periploca forrestii Schltr on wound healing by Src meditated Mek/Erk and PI3K/Akt signals. J Ethnopharmacol. 2019; 237: 116–127.
  3. Yu M, Liu W, Li J, et al. Exosomes derived from atorvastatin-pretreated MSC accelerate diabetic wound repair by enhancing angiogenesis via AKT/eNOS pathway. Stem Cell Res Ther. 2020; 11(1): 350.
  4. Oh M, Lee J, Kim YuJ, et al. Exosomes derived from human induced pluripotent stem cells ameliorate the aging of skin fibroblasts. Int J Mol Sci. 2018; 19(6).
  5. Cooper DR, Wang C, Patel R, et al. Human adipose-derived stem cell conditioned media and exosomes containing promote human dermal fibroblast migration and ischemic wound healing. Adv Wound Care (New Rochelle). 2018; 7(9): 299–308.
  6. Zhang K, Zhao X, Chen X, et al. Enhanced therapeutic effects of mesenchymal stem cell-derived exosomes with an injectable hydrogel for hindlimb ischemia treatment. ACS Appl Mater Interfaces. 2018; 10(36): 30081–30091.
  7. Wang C, Wang M, Xu T, et al. Engineering bioactive self-healing antibacterial exosomes hydrogel for promoting chronic diabetic wound healing and complete skin regeneration. Theranostics. 2019; 9(1): 65–76.
  8. Shi Q, Qian Z, Liu D, et al. GMSC-Derived Exosomes Combined with a Chitosan/Silk Hydrogel Sponge Accelerates Wound Healing in a Diabetic Rat Skin Defect Model. Front Physiol. 2017; 8: 904.
  9. Nooshabadi VT, Khanmohamadi M, Valipour E, et al. Impact of exosome-loaded chitosan hydrogel in wound repair and layered dermal reconstitution in mice animal model. J Biomed Mater Res A. 2020; 108(11): 2138–2149.
  10. Shafei S, Khanmohammadi M, Heidari R, et al. Exosome loaded alginate hydrogel promotes tissue regeneration in full-thickness skin wounds: An in vivo study. J Biomed Mater Res A. 2020; 108(3): 545–556.
  11. Wan ACA, Tai BCU. CHITIN - a promising biomaterial for tissue engineering and stem cell technologies. Biotechnol Adv. 2013; 31(8): 1776–1785.
  12. Choi EW, Seo MK, Woo EY, et al. Exosomes from human adipose-derived stem cells promote proliferation and migration of skin fibroblasts. Exp Dermatol. 2018; 27(10): 1170–1172.
  13. Zhao G, Liu F, Liu Z, et al. MSC-derived exosomes attenuate cell death through suppressing AIF nucleus translocation and enhance cutaneous wound healing. Stem Cell Res Ther. 2020; 11(1): 174.
  14. Borges GÁ, Elias ST, da Silva SM, et al. In vitro evaluation of wound healing and antimicrobial potential of ozone therapy. J Craniomaxillofac Surg. 2017; 45(3): 364–370.
  15. Chen L, Jiang P, Li J, et al. Periplocin promotes wound healing through the activation of Src/ERK and PI3K/Akt pathways mediated by Na/K-ATPase. Phytomedicine. 2019; 57: 72–83.
  16. Ma T, Fu B, Yang X, et al. Adipose mesenchymal stem cell-derived exosomes promote cell proliferation, migration, and inhibit cell apoptosis via Wnt/β-catenin signaling in cutaneous wound healing. J Cell Biochem. 2019; 120(6): 10847–10854.
  17. Jiang Z, Li L, Li H, et al. Preparation, biocompatibility, and wound healing effects of O-carboxymethyl chitosan nonwoven fabrics in partial-thickness burn model. Carbohydr Polym. 2022; 280: 119032.
  18. Chen L, Jiang P, Li J, et al. Periplocin promotes wound healing through the activation of Src/ERK and PI3K/Akt pathways mediated by Na/K-ATPase. Phytomedicine. 2019; 57: 72–83.
  19. Zhang W, Bai X, Zhao B, et al. Cell-free therapy based on adipose tissue stem cell-derived exosomes promotes wound healing via the PI3K/Akt signaling pathway. Exp Cell Res. 2018; 370(2): 333–342.
  20. Lei Z, Singh G, Min Z, et al. Bone marrow-derived mesenchymal stem cells laden novel thermo-sensitive hydrogel for the management of severe skin wound healing. Mater Sci Eng C Mater Biol Appl. 2018; 90: 159–167.
  21. Shabbir A, Cox A, Rodriguez-Menocal L, et al. Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro. Stem Cells Dev. 2015; 24(14): 1635–1647.
  22. Hu X, Zhou X, Li Y, et al. Application of stem cells and chitosan in the repair of spinal cord injury. Int J Dev Neurosci. 2019; 76: 80–85.
  23. Tao SC, Guo SC, Li M, et al. Chitosan wound dressings incorporating exosomes derived from microrna-126-overexpressing synovium mesenchymal stem cells provide sustained release of exosomes and heal full-thickness skin defects in a diabetic rat model. Stem Cells Transl Med. 2017; 6(3): 736–747.
  24. Rao F, Zhang D, Fang T, et al. Exosomes from human gingiva-derived mesenchymal stem cells combined with biodegradable chitin conduits promote rat sciatic nerve regeneration. Stem Cells Int. 2019; 2019: 2546367.
  25. Azuma K, Koizumi R, Izawa H, et al. Preparation and biomedical applications of chitin and chitosan nanofibers. J Biomed Nanotechnol. 2014; 10(10): 2891–2920.
  26. Khayambashi P, Iyer J, Pillai S, et al. Hydrogel encapsulation of mesenchymal stem cells and their derived exosomes for tissue engineering. Int J Mol Sci. 2021; 22(2): 684.
  27. Ahmad SI, Ahmad R, Khan MS, et al. Chitin and its derivatives: Structural properties and biomedical applications. Int J Biol Macromol. 2020; 164: 526–539.
  28. Jones M, Kujundzic M, John S, et al. Crab vs. Mushroom: a review of crustacean and fungal chitin in wound treatment. Mar Drugs. 2020; 18(1).
  29. Bailey-Steinitz LJ, Shih YH, Radeke MJ, et al. An in vitro model of chronic wounding and its implication for age-related macular degeneration. PLoS One. 2020; 15(7): e0236298.
  30. McCullough RL, McMullen MR, Sheehan MM, et al. Complement Factor D protects mice from ethanol-induced inflammation and liver injury. Am J Physiol Gastrointest Liver Physiol. 2018; 315(1): G66–G79.
  31. Ezure T, Sugahara M, Amano S. Senescent dermal fibroblasts negatively influence fibroblast extracellular matrix-related gene expression partly via secretion of complement factor D. Biofactors. 2019; 45(4): 556–562.
  32. Qi H, Wei J, Gao Y, et al. Reg4 and complement factor D prevent the overgrowth of E. coli in the mouse gut. Commun Biol. 2020; 3(1): 483.
  33. Song NJ, Kim S, Jang BH, et al. Small molecule-induced complement factor d (adipsin) promotes lipid accumulation and adipocyte differentiation. PLoS One. 2016; 11(9): e0162228.
  34. Wang C, Xu J, Yuan D, et al. Exosomes carrying ALDOA and ALDH3A1 from irradiated lung cancer cells enhance migration and invasion of recipients by accelerating glycolysis. Mol Cell Biochem. 2020; 469(1-2): 77–87.
  35. Chang YC, Chan YC, Chang WM, et al. Feedback regulation of ALDOA activates the HIF-1α/MMP9 axis to promote lung cancer progression. Cancer Lett. 2017; 403: 28–36.
  36. Lincet H, Icard P. How do glycolytic enzymes favour cancer cell proliferation by nonmetabolic functions? Oncogene. 2015; 34(29): 3751–3759.
  37. Saito Y, Takasawa A, Takasawa K, et al. Aldolase A promotes epithelial-mesenchymal transition to increase malignant potentials of cervical adenocarcinoma. Cancer Sci. 2020; 111(8): 3071–3081.



Folia Histochemica et Cytobiologica