We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Fabrication, characterization and wound-healing properties of core–shell SF@chitosan/ZnO/Astragalus arbusculinus gum nanofibers

    Zahra Amiri

    Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 74877-94149, Iran

    ,
    Amir Mahdi Molavi

    Department of Materials Research, Iranian Academic Center for Education, Culture & Research (ACECR), Khorasan Razavi Branch, Mashhad, 9177-948974, Iran

    ,
    Amir Amani

    Natural Products & Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 74877-94149, Iran

    ,
    Kurosh Hamzanlui Moqadam

    North Khorasan University of Medical Sciences, Bojnurd, 74877-94149, Iran

    ,
    Mehran Vatanchian

    Department of Anatomical Sciences School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 74877-94149, Iran

    ,
    Seyyed Ahmad Hashemi

    Vector-borne Diseases Research Center, North Khorasan University of Medical Sciences, Bojnurd, 74877-94149, Iran

    &
    Fatemeh Oroojalian

    *Author for correspondence:

    E-mail Address: f.oroojalian@gmail.com

    Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 74877-94149, Iran

    Natural Products & Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 74877-94149, Iran

    Published Online:https://doi.org/10.2217/nnm-2023-0311

    Aim: Silk fibroin/chitosan/ZnO/Astragalus arbusculinus (Ast) gum fibrous scaffolds along with adipose-derived mesenchymal stem cells (ADSCs) were investigated for accelerating diabetic wound healing. Methods: Scaffolds with a core–shell structure and different compositions were synthesized using the electrospinning method. Biological in vitro investigations included antibacterial testing, cell viability analysis and cell attachment evaluation. In vivo experiments, including the chicken chorioallantoic membrane (CAM) test, were conducted to assess wound-healing efficacy and histopathological changes. Results: The incorporation of Ast to the silk fibroin@ chitosan/ZnO scaffold improved wound healing in diabetic mice. In addition, seeding of ADSCs on the scaffold accelerated wound healing. Conclusion: These findings suggest that the designed scaffold can be useful for skin regeneration applications.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Ebrahim N, Dessouky AA, Mostafa O et al. Adipose mesenchymal stem cells combined with platelet-rich plasma accelerate diabetic wound healing by modulating the Notch pathway. Stem Cell Res. Ther. 12(1), 392 (2021). •• Demonstrates that diabetic wound healing is accelerated by adipose mesenchymal stem cells.
    • 2. Patel S, Srivastava S, Singh MR, Singh D. Mechanistic insight into diabetic wounds: pathogenesis, molecular targets and treatment strategies to pace wound healing. Biomed. Pharmacother. 112, 108615 (2019). •• Shows a mechanistic understanding of diabetic wounds.
    • 3. Wu J, Chen LH, Sun SY, Li Y, Ran XW. Mesenchymal stem cell-derived exosomes: the dawn of diabetic wound healing. World J. Diabetes 13(12), 1066–1095 (2022).
    • 4. Li C, Vepari C, Jin H-J, Kim HJ, Kaplan DL. Electrospun silk-BMP-2 scaffolds for bone tissue engineering. Biomaterials 27(16), 3115–3124 (2006).
    • 5. Abdollahiyan P, Oroojalian F, Mokhtarzadeh A. The triad of nanotechnology, cell signalling, and scaffold implantation for the successful repair of damaged organs: an overview on soft-tissue engineering. J. Control. Rel. 332, 460–492 (2021).
    • 6. Abdollahiyan P, Baradaran B, De La Guardia M, Oroojalian F, Mokhtarzadeh A. Cutting-edge progress and challenges in stimuli responsive hydrogel microenvironment for success in tissue engineering today. J. Control. Rel. 328, 514–531 (2020).
    • 7. Abdollahiyan P, Oroojalian F, Hejazi M, De La Guardia M, Mokhtarzadeh A. Nanotechnology, and scaffold implantation for the effective repair of injured organs: an overview on hard tissue engineering. J. Control. Rel. 333, 391–417 (2021).
    • 8. Babitha S, Rachita L, Karthikeyan K et al. Electrospun protein nanofibers in healthcare: a review. Int. J. Pharmaceut. 523(1), 52–90 (2017).
    • 9. Bagheri M, Validi M, Gholipour A, Makvandi P, Sharifi E. Chitosan nanofiber biocomposites for potential wound healing applications: antioxidant activity with synergic antibacterial effect. Bioeng. Translat. Med. 7(1), e10254 (2022).
    • 10. Farokhi M, Mottaghitalab F, Fatahi Y et al. Silk fibroin scaffolds for common cartilage injuries: possibilities for future clinical applications. Eur. Polymer J. 115, 251–267 (2019).
    • 11. Sukmana BI, Margiana R, Almajidi YQ et al. Supporting wound healing by mesenchymal stem cells (MSCs) therapy in combination with scaffold, hydrogel, and matrix; state of the art. Pathol. Res. Pract. 248, 154575 (2023). • Potential of stem cells, including mesenchymal stem cells (MSCs), in regenerative medicine.
    • 12. Khosravi N, Pishavar E, Baradaran B, Oroojalian F, Mokhtarzadeh A. Stem cell membrane, stem cell-derived exosomes and hybrid stem cell camouflaged nanoparticles: a promising biomimetic nanoplatforms for cancer theranostics. J. Control. Rel. 348, 706–722 (2022).
    • 13. Ullah I, Subbarao RB, Rho GJ. Human mesenchymal stem cells – current trends and future prospective. Biosci. Rep. 35(2), e00191 (2015).
    • 14. Jung S, Kim JH, Yim C, Lee M, Kang HJ, Choi D. Therapeutic effects of a mesenchymal stem cell-based insulin-like growth factor-1/enhanced green fluorescent protein dual gene sorting system in a myocardial infarction rat model. Mol. Med. Rep. 18(6), 5563–5571 (2018).
    • 15. Klar AS, Zimoch J, Biedermann T. Skin tissue engineering: application of adipose-derived stem cells. BioMed Res. Int. 2017, 9747010 (2017).
    • 16. Mazini L, Rochette L, Amine M, Malka G. Regenerative capacity of adipose derived stem cells (ADSCs), comparison with mesenchymal stem cells (MSCs). Int. J. Mol. Sci. 20(10), 1–30 (2019).
    • 17. Alamdari SG, Alibakhshi A, de la Guardia M et al. Evaluation of chemical modification effects on DNA plasmid transfection efficiency of single-walled carbon nanotube–succinate–polyethylenimine conjugates as non-viral gene carriers. Adv. Healthc. Mater. 11, 2200526 (2022).
    • 18. Altman AM, Yan Y, Matthias N et al. IFATS collection: human adipose-derived stem cells seeded on a silk fibroin-chitosan scaffold enhance wound repair in a murine soft tissue injury model. Stem Cells 27(1), 250–258 (2009).
    • 19. Wu YY, Jiao YP, Xiao LL et al. Experimental study on effects of adipose-derived stem cell-seeded silk fibroin chitosan film on wound healing of a diabetic rat model. Ann. Plastic Surg. 80(5), 572–580 (2018). •• Shows the role of chitosan, fibroin and adipose-derived stem cells in wound healing.
    • 20. Karimi MA, Dadmehr M, Hosseini M, Korouzhdehi B, Oroojalian F. Sensitive detection of methylated DNA and methyltransferase activity based on the lighting up of FAM-labeled DNA quenched fluorescence by gold nanoparticles. RSC Adv. 9(21), 12063–12069 (2019).
    • 21. Omidi M, Malakoutian MA, Choolaei M, Oroojalian F, Haghiralsadat F, Yazdian F. A label-free detection of biomolecules using micromechanical biosensors. Chin. Phys. Lett. 30(6), 068701 (2013).
    • 22. Le VAT, Trinh TX, Chien PN et al. Evaluation of the performance of a ZnO-nanoparticle-coated hydrocolloid patch in wound healing. Polymers 14(5), 919 (2022).
    • 23. Liu Y, Zhang X, Yang L et al. Proteomics and transcriptomics explore the effect of mixture of herbal extract on diabetic wound healing process. Phytomedicine 116, 154892 (2023).
    • 24. Ding X, Tang Q, Xu Z et al. Challenges and innovations in treating chronic and acute wound infections: from basic science to clinical practice. Burns Trauma 10, tkac014 (2022).
    • 25. Ahmadi E, Rezadoost H, Moridi Farimani M. Isolation, characterization, and antioxidant activity of neutral carbohydrates from Astragalus arbusculinus gum. S. Afr. J. Botany 146, 669–675 (2022).
    • 26. Shojaii A, Motevalian M, Rahnama N. Evaluation of anti-inflammatory and analgesic activities and the phytochemical study of Astragalus arbusculinus gum in animal models. J. Basic Clin. Physiol. Pharmacol. 26(4), 369–374 (2015). • Demonstrates the significance of Astragalus arbusculinus in medicine.
    • 27. Chaala M, Sebba FZ, Fuster MG et al. Accelerated simple preparation of curcumin-loaded silk fibroin/hyaluronic acid hydrogels for biomedical applications. Polymers 15(3), 504 (2023).
    • 28. Doostan M, Doostan M, Mohammadi P, Khoshnevisan K, Maleki H. Wound healing promotion by flaxseed extract-loaded polyvinyl alcohol/chitosan nanofibrous scaffolds. Int. J. Biol. Macromol. 228, 506–516 (2023).
    • 29. Pourpirali R, Mahmoudnezhad A, Oroojalian F, Zarghami N, Pilehvar Y. Prolonged proliferation and delayed senescence of the adipose-derived stem cells grown on the electrospun composite nanofiber co-encapsulated with TiO2 nanoparticles and metformin-loaded mesoporous silica nanoparticles. Int. J. Pharmaceut. 604, 120733 (2021).
    • 30. Shitole AA, Raut P, Giram P et al. Poly(vinylpyrrolidone)-iodine engineered poly (ε-caprolactone) nanofibers as potential wound dressing materials. Mater. Sci. Eng. C 110, 110731 (2020).
    • 31. Yue F, Zhang J, Xu J, Niu T, Lü X, Liu M. Effects of monosaccharide composition on quantitative analysis of total sugar content by phenol-sulfuric acid method. Front. Nutr. 9, 963318 (2022).
    • 32. Mirmajidi T, Chogan F, Rezayan AH, Sharifi AM. In vitro and in vivo evaluation of a nanofiber wound dressing loaded with melatonin. Int. J. Pharmaceut. 596, 120213 (2021).
    • 33. Bunnell BA, Flaat M, Gagliardi C, Patel B, Ripoll C. Adipose-derived stem cells: isolation, expansion and differentiation. Methods (San Diego, Calif.) 45(2), 115–120 (2008).
    • 34. Hashem Nia A, Behnam B, Taghavi S et al. Evaluation of chemical modification effects on DNA plasmid transfection efficiency of singlewalled carbon nanotube–succinate–polyethylenimine conjugates as non-viral gene carriers. MedChemComm 8(2), 364–375 (2017).
    • 35. Mirzadegan E, Golshahi H, Saffarian Z et al. The remarkable effect of menstrual blood stem cells seeded on bilayer scaffold composed of amniotic membrane and silk fibroin aiming to promote wound healing in diabetic mice. Int. Immunopharmacol. 102, 108404 (2022).
    • 36. Burggren W, Rojas Antich M. Angiogenesis in the avian embryo chorioallantoic membrane: a perspective on research trends and a case study on toxicant vascular effects. J. Cardiovasc. Develop. Dis. 7(4), 56 (2020).
    • 37. Li J, Zhang T, Pan M et al. Nanofiber/hydrogel core–shell scaffolds with three-dimensional multilayer patterned structure for accelerating diabetic wound healing. J. Nanobiotechnol. 20(1), 28 (2022). •• Intriguing method for creating a multipurpose hydrogel to aid in wound healing.
    • 38. Zarekhalili Z, Bahrami SH, Ranjbar-Mohammadi M, Milan PB. Fabrication and characterization of PVA/Gum tragacanth/PCL hybrid nanofibrous scaffolds for skin substitutes. Int. J. Biol. Macromol. 94, 679–690 (2017).
    • 39. Lovett M, Eng G, Kluge JA, Cannizzaro C, Vunjak-Novakovic G, Kaplan DL. Tubular silk scaffolds for small diameter vascular grafts. Organogenesis 6(4), 217–224 (2010).
    • 40. Pham DT, Saelim N, Tiyaboonchai W. Crosslinked fibroin nanoparticles using EDC or PEI for drug delivery: physicochemical properties, crystallinity and structure. J. Mater. Sci. 53(20), 14087–14103 (2018).
    • 41. Nagpal M, Singh S, Mishra D. Superporous hybrid hydrogels based on polyacrylamide and chitosan: characterization and in vitro drug release. Int. J. Pharmaceut. Invest. 3, 88–94 (2013).
    • 42. Kumirska J, Czerwicka M, Kaczyński Z et al. Application of spectroscopic methods for structural analysis of chitin and chitosan. Marine Drugs 8(5), 1567–1636 (2010).
    • 43. Lustriane C, Dwivany FM, Suendo V, Reza M. Effect of chitosan and chitosan-nanoparticles on post harvest quality of banana fruits. J. Plant Biotechnol. 45(1), 36–44 (2018).
    • 44. Kurusu RS, Demarquette NR. Surface modification to control the water wettability of electrospun mats. Int. Mater. Rev. 64(5), 249–287 (2019).
    • 45. Azari P, Hosseini S, Murphy BP, Martinez-Chapa SO. Electrospun biopolyesters: hydrophobic scaffolds with favorable biological response. J. Public Health Int. 1, 5–9 (2018).
    • 46. Stewart SA, Domínguez-Robles J, Donnelly RF, Larrañeta E. Implantable polymeric drug delivery devices: classification, manufacture, materials, and clinical applications. Polymers 10(12), 1379 (2018).
    • 47. Ali A, Shahid MA, Hossain MD, Islam MN. Antibacterial bi-layered polyvinyl alcohol (PVA)-chitosan blend nanofibrous mat loaded with Azadirachta indica (neem) extract. Int. J. Biol. Macromol. 138, 13–20 (2019).
    • 48. Revathi T, Thambidurai S. Cytotoxic, antioxidant and antibacterial activities of copper oxide incorporated chitosan-neem seed biocomposites. Int. J. Biol. Macromol. 139, 867–878 (2019).
    • 49. Augustine R, Dominic EA, Reju I, Kaimal B, Kalarikkal N, Thomas S. Investigation of angiogenesis and its mechanism using zinc oxide nanoparticle-loaded electrospun tissue engineering scaffolds. RSC Adv. 4(93), 51528–51536 (2014).
    • 50. Xu X, Wang X, Qin C, Khan AUR, Zhang W, Mo X. Silk fibroin/poly-(L-lactide-co-caprolactone) nanofiber scaffolds loaded with Huangbai liniment to accelerate diabetic wound healing. Colloids Surf. B. Biointerf. 199, 111557 (2021).
    • 51. Sabra S, Ragab DM, Agwa MM, Rohani S. Recent advances in electrospun nanofibers for some biomedical applications. Eur. J. Pharmaceut. Sci. 144, 105224 (2020).
    • 52. Elsherbini AM, Sabra SA, Rashed SA, Abdelmonsif DA, Haroun M, Shalaby TI. Electrospun polyvinyl alcohol/Withania somnifera extract nanofibers incorporating tadalafil-loaded nanoparticles for diabetic ulcers. Nanomedicine (Lond.) 18(20), 1361–1382 (2023).
    • 53. Jamnongkan T, Kaewpirom S, Wattanakornsiri A, Mongkholrattanasit R. Effect of ZnO concentration on the diameter of electrospun fibers from poly(vinyl alcohol) composited with ZnO nanoparticles. Key Engin. Mater. 759, 81–85 (2018).
    • 54. Mariani E, Lisignoli G, Borzì RM, Pulsatelli L. Biomaterials: foreign bodies or tuners for the immune response? Int. J. Mol. Sci. 20(3), 636 (2019).
    • 55. Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J. Cell Sci. 123(24), 4195–4200 (2010).
    • 56. Zhao S, Yan W, Shi M, Wang Z, Wang J, Wang S. Improving permeability and antifouling performance of polyethersulfone ultrafiltration membrane by incorporation of ZnO-DMF dispersion containing nano-ZnO and polyvinylpyrrolidone. J. Membr. Sci. 478, 105–116 (2015).
    • 57. Zhang L, Liu X, Li G, Wang P, Yang Y. Tailoring degradation rates of silk fibroin scaffolds for tissue engineering. J. Biomed. Mater. Res. Part A 107(1), 104–113 (2019).
    • 58. Du M, Zhao W, Ma R et al. Visible-light-driven photocatalytic inactivation of S. aureus in aqueous environment by hydrophilic zinc oxide (ZnO) nanoparticles based on the interfacial electron transfer in S. aureus/ZnO composites. J. Hazard. Mater. 418, 126013 (2021). •• Demonstrates zinc oxide's antimicrobial activity.
    • 59. Zhao B, Zhang X, Han W, Cheng J, Qin Y. Wound healing effect of an Astragalus membranaceus polysaccharide and its mechanism. Mol. Med. Rep. 15(6), 4077–4083 (2017).
    • 60. Guo J, Hu H, Gorecka J et al. Adipose-derived mesenchymal stem cells accelerate diabetic wound healing in a similar fashion as bone marrow-derived cells. Am. J. Physiol. Cell Physiol. 315(6), C885–C896 (2018). •• Confirms mesenchymal stem cells produced from adipose tissue and bone marrow both speed up the healing of diabetic wounds.
    • 61. Tyeb S, Shiekh PA, Verma V, Kumar A. Adipose-derived stem cells (ADSCs) loaded gelatin-sericin-laminin cryogels for tissue regeneration in diabetic wounds. Biomacromolecules 21(2), 294–304 (2020).
    • 62. Navone SE, Pascucci L, Dossena M et al. Decellularized silk fibroin scaffold primed with adipose mesenchymal stromal cells improves wound healing in diabetic mice. Stem Cell Res. Ther. 5, 1–15 (2014).