Skip to main content

Advertisement

Log in

Antioxidant-antibacterial containing bi-layer scaffolds as potential candidates for management of oxidative stress and infections in wound healing

  • Tissue Engineering Constructs and Cell Substrates
  • Original Research
  • Published:
Journal of Materials Science: Materials in Medicine Aims and scope Submit manuscript

Abstract

Tissue engineering techniques are continuously evolving towards providing better microenvironment along with therapeutic potential to address the skin tissue defects. Factors such as microbial infections, presence of excessive free radicals and depletion in antioxidant based scavenging systems pose serious challenges by prolonging inflammation and delaying the repair process. Incorporation of bioactive molecules in polymer based biomimetic scaffolds may present new vistas for handling chronic wounds. In this study, chitosan/collagen scaffolds incorporating 0.5, 1 and 2% (w/w) silymarin (CS-CO-SM) were synthesized and studied for their biocompatibility, in vitro release kinetics and anti-oxidant activity. The release kinetics of silymarin from the CS-CO-SM scaffold showed an initial burst followed by sustained release. The scaffolds were biocompatible and supported the recovery of COS-7 cells from UV induced oxidative stress. Further the CS-CO-SM(2) scaffolds were used to fabricate a bi-layer scaffold by layer upon layer arrangement with CS-Ag3 (3% Ag, w/w). The Ag was incorporated to impart antimicrobial property to the scaffold. The in vivo studies on bi-layer scaffolds were carried out in Wistar rat models at 3, 7 and 10 days post injury and the skin excisions were studied for wound contraction, histology (H&E staining), and lipid peroxidation. The bi-layer scaffold accelerated the process of wound healing with no inflammatory cells, proliferation of fibroblast, neovascularization and collagen deposition. By day 10 post transplantation of the scaffold, the skin had a structure similar to normal skin with complete re-epithelization. This bi-layer scaffold with antioxidant and antimicrobial properties promotes wound healing and is proposed as a potential tissue engineering material for managing chronic wounds.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9

Similar content being viewed by others

References

  1. Pellegrini G, Ranno R, Stracuzzi G, Bondanza S, Guerra L, Zambruno G. et al. The control of epidermal stem cells (holoclones) in the treatment of massive full-thickness burns with autologous keratinocytes cultured on fibrin1. Transplantation. 1999;68:868–79.

    Article  CAS  Google Scholar 

  2. Kim MS, Kim JH, Min BH, Chun HJ, Han DK, Lee HB. Polymeric scaffolds for regenerative medicine. Polym Rev. 2011;51:23–52.

    Article  CAS  Google Scholar 

  3. Muzzarelli RA, Greco F, Busilacchi A, Sollazzo V, Gigante A. Chitosan, hyaluronan and chondroitin sulfate in tissue engineering for cartilage regeneration: a review. Carbohydr Polym. 2012;89:723–39.

    Article  CAS  Google Scholar 

  4. Singh D, Singh D, Han SS. 3D printing of scaffold for cells delivery: advances in skin tissue engineering. Polymers. 2016;8:19.

    Article  Google Scholar 

  5. Watt FM, Fujiwara H. Cell-extracellular matrix interactions in normal and diseased skin. Cold Spring Harb Perspect Biol. 2011;3:a005124.

    Article  Google Scholar 

  6. Knight T, Basu J, Rivera EA, Spencer T, Jain D, Payne R. Fabrication of a multi-layer three-dimensional scaffold with controlled porous micro-architecture for application in small intestine tissue engineering. Cell Adhesion Migration. 2013;7:267–74.

    Article  Google Scholar 

  7. Maheshwari SU, Samuel VK, Nagiah N. Fabrication and evaluation of (PVA/HAp/PCL) bilayer composites as potential scaffolds for bone tissue regeneration application. Ceram Int. 2014;40:8469–77.

    Article  Google Scholar 

  8. Mohandas A, Nimal T, Das V, Shankarappa SA, Biswas R, Jayakumar R. Drug loaded bi-layered sponge for wound management in hyperfibrinolytic conditions. J Mater Chem B. 2015;3:5795–805.

    Article  CAS  Google Scholar 

  9. Kumbhar JV, Jadhav SH, Bodas DS, Barhanpurkar-Naik A, Wani MR, Paknikar KM. et al. In vitro and in vivo studies of a novel bacterial cellulose-based acellular bilayer nanocomposite scaffold for the repair of osteochondral defects. Int J Nanomed. 2017;12:6437.

    Article  Google Scholar 

  10. Deng Z, Qian Y, Yu Y, Liu G, Hu J, Zhang G. et al. Engineering intracellular delivery nanocarriers and nanoreactors from oxidation-responsive polymersomes via synchronized bilayer cross-linking and permeabilizing inside live cells. J Am Chem Soc. 2016;138:10452.

    Article  Google Scholar 

  11. Wolcott R, Rhoads D, Bennett M, Wolcott B, Gogokhia L, Costerton J. et al. Chronic wounds and the medical biofilm paradigm. J Wound Care. 2010;19:45–53.

    Article  CAS  Google Scholar 

  12. Guo Sa, DiPietro LA. Factors affecting wound healing. J Dent Res. 2010;89:219–29.

    Article  CAS  Google Scholar 

  13. Schreml S, Szeimies R, Prantl L, Karrer S, Landthaler M, Babilas P. Oxygen in acute and chronic wound healing. Br J Dermatol. 2010;163:257–68.

    Article  CAS  Google Scholar 

  14. Bryan N, Ahswin H, Smart N, Bayon Y, Wohlert S, Hunt JA. Reactive oxygen species (ROS)–a family of fate deciding molecules pivotal in constructive inflammation and wound healing. Eur Cell Mater. 2012;24:e65

    Google Scholar 

  15. Li X, Nan K, Li L, Zhang Z, Chen H. In vivo evaluation of curcumin nanoformulation loaded methoxy poly (ethylene glycol)-graft-chitosan composite film for wound healing application. Carbohydr Polym. 2012;88:84–90.

    Article  CAS  Google Scholar 

  16. Lee Y-H, Chang J-J, Chien C-T, Yang M-C, Chien H-F. Antioxidant sol-gel improves cutaneous wound healing in streptozotocin-induced diabetic rats. Exp Diabetes Res. 2012;2012:1–11.

    Google Scholar 

  17. Tabandeh MR, Oryan A, Mohhammad-Alipour A, Tabatabaei-Naieni A. Silibinin regulates matrix metalloproteinase 3 (stromelysine1) gene expression, hexoseamines and collagen production during rat skin wound healing. Phytother Res. 2013;27:1149–53.

    Article  CAS  Google Scholar 

  18. Soto C, Pérez J, García V, Uría E, Vadillo M, Raya L. Effect of silymarin on kidneys of rats suffering from alloxan-induced diabetes mellitus. Phytomedicine. 2010;17:1090–4.

    Article  CAS  Google Scholar 

  19. Gülçin I. Antioxidant activity of food constituents: an overview. Arch Toxicol. 2012;86:345–91.

    Article  Google Scholar 

  20. Guzman M, Dille J, Godet S. Synthesis and antibacterial activity of silver nanoparticles against gram-positive and gram-negative bacteria. Nanomed: Nanotechnol, Biol Med. 2012;8:37–45.

    Article  CAS  Google Scholar 

  21. Li W-R, Xie X-B, Shi Q-S, Zeng H-Y, You-Sheng O-Y, Chen Y-B. Antibacterial activity and mechanism of silver nanoparticles on Escherichia coli. Appl Microbiol Biotechnol. 2010;85:1115–22.

    Article  CAS  Google Scholar 

  22. Naik K, Kowshik M. The silver lining: towards the responsible and limited usage of silver. J Appl Microbiol. 2017;123:1068–87.

    Article  CAS  Google Scholar 

  23. Shaik MM, Kowshik M. Novel melt-down neutralization method for synthesis of chitosan–silver scaffolds for tissue engineering applications. Polym Bull. 2016;73:841–58.

    Article  CAS  Google Scholar 

  24. Yang G, Zhao Y, Feng N, Zhang Y, Liu Y, Dang B. Improved dissolution and bioavailability of silymarin delivered by a solid dispersion prepared using supercritical fluids. Asian J Pharm Sci. 2015;10:194.

    Article  Google Scholar 

  25. Sionkowska A, Wisniewski M, Skopinska J, Kennedy C, Wess T. Molecular interactions in collagen and chitosan blends. Biomaterials. 2004;25:795–801.

    Article  CAS  Google Scholar 

  26. Pooja D, Babu Bikkina DJ, Kulhari H, Nikhila N, Chinde S, Raghavendra YM. et al. Fabrication, characterization and bioevaluation of silibinin loaded chitosan nanoparticles. Int J Biol Macromol. 2014;69:267–73.

    Article  CAS  Google Scholar 

  27. Han K-S, Song JE, Tripathy N, Kim H, Moon BM, Park CH. et al. Effect of pore sizes of silk scaffolds for cartilage tissue engineering. Macromol Res. 2015;23:1091–7.

    Article  CAS  Google Scholar 

  28. Chou S-F, Carson D, Woodrow KA. Current strategies for sustaining drug release from electrospun nanofibers. J Control Release. 2015;220:584–91.

    Article  CAS  Google Scholar 

  29. Lin C-C, Metters AT. Hydrogels in controlled release formulations: network design and mathematical modeling. Adv Drug Deliv Rev. 2006;58:1379–408.

    Article  CAS  Google Scholar 

  30. Pundir S, Badola A, Sharma D. Sustained release matrix technology and recent advance in matrix drug delivery system: a review. Int J Drug Res Technol. 2017;3:8.

    Google Scholar 

  31. Kishen A, Shrestha S, Shrestha A, Cheng C, Goh C. Characterizing the collagen stabilizing effect of crosslinked chitosan nanoparticles against collagenase degradation. Dent Mater. 2016;32:968–77.

    Article  CAS  Google Scholar 

  32. Sharma S, Bano S, Ghosh AS, Mandal M, Kim H-W, Dey T. et al. Silk fibroin nanoparticles support in vitro sustained antibiotic release and osteogenesis on titanium surface. Nanomed: Nanotechnol, Biol Med. 2016;12:1193–204.

    Article  CAS  Google Scholar 

  33. Leena R, Vairamani M, Selvamurugan N. Alginate/Gelatin scaffolds incorporated with Silibinin-loaded Chitosan nanoparticles for bone formation in vitro. Colloids Surf B: Biointerfaces. 2017;158:308–18.

    Article  CAS  Google Scholar 

  34. Lee K, Seo C-R, Ku J-M, Lee H, Yoon H, Lee J. et al. 3D-printed alginate/phenamil composite scaffolds constituted with microsized core–shell struts for hard tissue regeneration. RSC Adv. 2015;5:29335–45.

    Article  CAS  Google Scholar 

  35. Škottová N, Večeřa R, Urbánek K, Váňa P, Walterová D, Cvak L. Effects of polyphenolic fraction of silymarin on lipoprotein profile in rats fed cholesterol-rich diets. Pharmacol Res. 2003;47:17–26.

    Article  Google Scholar 

  36. Morgan C, Nigam Y. Naturally derived factors and their role in the promotion of angiogenesis for the healing of chronic wounds. Angiogenesis. 2013;16:493–502.

    Article  CAS  Google Scholar 

  37. Khanna S, Biswas S, Shang Y, Collard E, Azad A, Kauh C. et al. Macrophage dysfunction impairs resolution of inflammation in the wounds of diabetic mice. PLoS ONE. 2010;5:e9539.

    Article  Google Scholar 

  38. Jain J, Arora S, Rajwade JM, Omray P, Khandelwal S, Paknikar KM. Silver nanoparticles in therapeutics: development of an antimicrobial gel formulation for topical use. Mol Pharm. 2009;6:1388–401.

    Article  CAS  Google Scholar 

  39. You C, Li Q, Wang X. et al. Silver nanoparticle loaded collagen/chitosan scaffolds promote wound healing via regulating fibroblast migration and macrophage activation. Sci Rep. 2017;7:10489.

    Article  Google Scholar 

  40. Nencini C, Giorgi G, Micheli L. Protective effect of silymarin on oxidative stress in rat brain. Phytomedicine. 2007;14:129–35.

    Article  CAS  Google Scholar 

  41. Sherif IO, Al-Gayyar MM. Antioxidant, anti-inflammatory and hepatoprotective effects of silymarin on hepatic dysfunction induced by sodium nitrite. Eur Cytokine Netw. 2013;24:114–21.

    CAS  Google Scholar 

  42. Nielsen F, Mikkelsen BB, Nielsen JB, Andersen HR, Grandjean P.Plasma malondialdehyde as biomarker for oxidative stress: reference interval and effects of life-style factors. Clin Chem. 1997;43:1209–14.

    CAS  Google Scholar 

  43. Feng Z, Hu W, Marnett LJ, Tang M-s. Malondialdehyde, a major endogenous lipid peroxidation product, sensitizes human cells to UV-and BPDE-induced killing and mutagenesis through inhibition of nucleotide excision repair. Mutat Res/Fundam Mol Mech Mutagen. 2006;601:125–36.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge Mr. Joseph RD Fernandes and Dr. Arnab Banerjee for the kind support in immunohistochemical staining. The authors also want to acknowledge the central facility, BITS Pilani K K Birla Goa Campus for the help in characterization of the scaffolds, Dr. Chandrashekhar S. Mote for the histological evaluation of skin grafts and Agharkar Research Institute, Pune for the animal studies experiments.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Meenal Kowshik.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shaik, M.M., Dapkekar, A., Rajwade, J.M. et al. Antioxidant-antibacterial containing bi-layer scaffolds as potential candidates for management of oxidative stress and infections in wound healing. J Mater Sci: Mater Med 30, 13 (2019). https://doi.org/10.1007/s10856-018-6212-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s10856-018-6212-8

Navigation