Journal Home > Volume 16 , Issue 1

Wound ulceration caused by diabetes is a typical chronic wound wherein healing the local tissue is difficult due to lack of blood vessels and tissue necrosis caused by the long-term accumulation of free radicals. Near-field electrospinning (NFES) is an innovative technology used to produce micro-nano-scaled, controllable sequencing fibers. In this study, we constructed a novel wound dressing based on the NFES polycaprolactone (PCL) fiber network and modified gelatin with methacrylic anhydride (GelMA) hydrogel to promote angiogenesis and the re-epithelialization of diabetic wounds. An angiogenic and antioxidant drug named deferoxamine (DFO) was encapsulated in a GelMA hydrogel to achieve a slow-release effect that is more suitable for chronic wounds. The cell adhesion experiment showed that the cells could attach to the fibers in the dressing group having a network of PCL fibers on the surface and grow along the direction of the fibers, which in turn, effectively regulates cell behavior from the physical structure. Additionally, the large pore size (~ 500 μm) of the network allowed the cells to penetrate the pores and enter the surface of the hydrogel without being blocked out. Besides, the composite dressing had a notable effect on angiogenesis. Furthermore, antioxidation experiments confirmed that the DFO-loaded hydrogel exhibited antioxidant activity. Experimental animal models of diabetes showed that rats treated with the PCL-GelMA-DFO (PGD) hydrogel had faster ability of hemostasis, scab formation, and wound healing. In conclusion, the PGD hydrogel effectively promoted the repair of chronic wounds.


menu
Abstract
Full text
Outline
Electronic supplementary material
About this article

Near-field electrospun PCL fibers/GelMA hydrogel composite dressing with controlled deferoxamine-release ability and retiform surface for diabetic wound healing

Show Author's information Huiling Zhong1,2,§Jun Huang3,§Moucheng Luo2Yifei Fang1,2Xinchen Zeng4Jun Wu1,2( )Jianhang Du1,5( )
Medical Research Center, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, China
Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
Department of Liver Surgery, The Third People’s Hospital of Shenzhen, Shenzhen 518112, China
NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou 510080, China

§ Huiling Zhong and Jun Huang contributed equally to this work.

Abstract

Wound ulceration caused by diabetes is a typical chronic wound wherein healing the local tissue is difficult due to lack of blood vessels and tissue necrosis caused by the long-term accumulation of free radicals. Near-field electrospinning (NFES) is an innovative technology used to produce micro-nano-scaled, controllable sequencing fibers. In this study, we constructed a novel wound dressing based on the NFES polycaprolactone (PCL) fiber network and modified gelatin with methacrylic anhydride (GelMA) hydrogel to promote angiogenesis and the re-epithelialization of diabetic wounds. An angiogenic and antioxidant drug named deferoxamine (DFO) was encapsulated in a GelMA hydrogel to achieve a slow-release effect that is more suitable for chronic wounds. The cell adhesion experiment showed that the cells could attach to the fibers in the dressing group having a network of PCL fibers on the surface and grow along the direction of the fibers, which in turn, effectively regulates cell behavior from the physical structure. Additionally, the large pore size (~ 500 μm) of the network allowed the cells to penetrate the pores and enter the surface of the hydrogel without being blocked out. Besides, the composite dressing had a notable effect on angiogenesis. Furthermore, antioxidation experiments confirmed that the DFO-loaded hydrogel exhibited antioxidant activity. Experimental animal models of diabetes showed that rats treated with the PCL-GelMA-DFO (PGD) hydrogel had faster ability of hemostasis, scab formation, and wound healing. In conclusion, the PGD hydrogel effectively promoted the repair of chronic wounds.

Keywords: hydrogel, diabetic wound healing, near field electrospinning, large pore

References(47)

[1]

Xu, Z. J.; Han, S. Y.; Gu, Z. P.; Wu, J. Advances and impact of antioxidant hydrogel in chronic wound healing. Adv. Healthcare Mater. 2020, 9, 1901502.

[2]

Gregg, E. W.; Li, Y. F.; Wang, J.; Burrows, N. R.; Ali, M. K.; Rolka, D.; Williams, D. E.; Geiss, L. Changes in diabetes-related complications in the United States, 1990–2010. N. Engl. J. Med. 2014, 370, 1514–1523.

[3]

Jeffcoate, W. J.; Vileikyte, L.; Boyko, E. J.; Armstrong, D. G.; Boulton, A. J. M. Current challenges and opportunities in the prevention and management of diabetic foot ulcers. Diabetes Care 2018, 41, 645–652.

[4]

Su, X. H.; Xian, C. H.; Gao, M.; Liu, G. T.; Wu, J. Edible materials in tissue regeneration. Macromol. Biosci. 2021, 21, 2100114.

[5]

Zhou, Y.; Liu, G. T.; Huang, H.; Wu, J. Advances and impact of arginine-based materials in wound healing. J. Mater. Chem. B 2021, 9, 6738–6750.

[6]

Matoori, S.; Veves, A.; Mooney, D. J. Advanced bandages for diabetic wound healing. Sci. Trans. Med. 2021, 13, eabe4839.

[7]

Hu, B.; Gao, M. Z.; Boakye-Yiadom, K. O.; Ho, W.; Yu, W.; Xu, X. Y.; Zhang, X. Q. An intrinsically bioactive hydrogel with on-demand drug release behaviors for diabetic wound healing. Bioact. Mater. 2021, 6, 4592–4606.

[8]

Liu, J.; Chen, Z. Q.; Wang, J.; Li, R. H.; Li, T. T.; Chang, M. Y.; Yan, F.; Wang, Y. F. Encapsulation of curcumin nanoparticles with MMP9-responsive and thermos-sensitive hydrogel improves diabetic wound healing. ACS Appl. Mater. Interfaces 2018, 10, 16315–16326.

[9]

Qian, Z. Y.; Wang, H. P.; Bai, Y. T.; Wang, Y. Q.; Tao, L.; Wei, Y.; Fan, Y. B.; Guo, X. M.; Liu, H. F. Improving chronic diabetic wound healing through an injectable and self-healing hydrogel with platelet-rich plasma release. ACS Appl. Mater. Interfaces 2020, 12, 55659–55674.

[10]

Hu, Y. Q.; Tao, R. Y.; Chen, L.; Xiong, Y.; Xue, H.; Hu, L. C.; Yan, C. C.; Xie, X. D.; Lin, Z.; Panayi, A. C. et al. Exosomes derived from pioglitazone-pretreated MSCs accelerate diabetic wound healing through enhancing angiogenesis. J. Nanobiotechnol. 2021, 19, 150.

[11]

Xu, Y.; Chen, X.; Qian, Y.; Tang, H. Z.; Song, J. J.; Qu, X. H.; Yue, B.; Yuan, W. E. Melatonin-based and biomimetic scaffold as muscle-ECM implant for guiding myogenic differentiation of volumetric muscle loss. Adv. Funct. Mater. 2020, 30, 2002378.

[12]
Chen, X.; Han, S. Y.; Wu, W. H.; Wu, Z. H.; Yuan, Y.; Wu, J.; Liu, C. S. Harnessing 4D printing bioscaffolds for advanced orthopedics. Small, in press, https://doi.org/10.1002/smll.202106824.
[13]

Yoon, D. S.; Lee, Y.; Ryu, H. A.; Jang, Y.; Lee, K. M.; Choi, Y.; Choi, W. J.; Lee, M.; Park, K. M.; Park, K. D. et al. Cell recruiting chemokine-loaded sprayable gelatin hydrogel dressings for diabetic wound healing. Acta Biomater. 2016, 38, 59–68.

[14]

Saito, T.; Tabata, Y. Hypoxia-induced angiogenesis is increased by the controlled release of deferoxiamine from gelatin hydrogels. Acta Biomater. 2014, 10, 3641–3649.

[15]

Bao, Z.; Xian, C.; Yuan, Q.; Liu, G.; Wu, J. Natural polymer-based hydrogels with enhanced mechanical performances: Preparation, structure, and property. Adv. Healthcare Mater. 2019, 8, 1900670.

[16]

Hunt, J. A.; Chen, R.; Van Veen, T.; Bryan, N. Hydrogels for tissue engineering and regenerative medicine. J. Mater. Chem. B 2014, 2, 5319–5338.

[17]

Duscher, D.; Neofytou, E.; Wong, V. W.; Maan, Z. N.; Rennert, R. C.; Inayathullah, M.; Januszyk, M.; Rodrigues, M.; Malkovskiy, A. V.; Whitmore, A. J. et al. Transdermal deferoxamine prevents pressure-induced diabetic ulcers. Proc. Natl. Acad. Sci. USA 2015, 112, 94–99.

[18]

Luo, M. C.; Zhang, X. Y.; Wu, J.; Zhao, J. M. Modifications of polysaccharide-based biomaterials under structure−property relationship for biomedical applications. Carbohydr. Polym. 2021, 266, 118097.

[19]

Ding, J. X.; Zhang, J.; Li, J. N.; Li, D.; Xiao, C. S.; Xiao, H. H.; Yang, H. H.; Zhuang, X. L.; Chen, X. S. Electrospun polymer biomaterials. Prog. Polym. Sci. 2019, 90, 1–34.

[20]

Zhang, X.; Li, Y.; Ma, Z. J.; He, D.; Li, H. Y. Modulating degradation of sodium alginate/bioglass hydrogel for improving tissue infiltration and promoting wound healing. Bioact. Mater. 2021, 6, 3692–3704.

[21]

Wu, J. L.; Hong, Y. Enhancing cell infiltration of electrospun fibrous scaffolds in tissue regeneration. Bioact. Mater. 2016, 1, 56–64.

[22]

Feng, X. R.; Li, J. N.; Zhang, X.; Liu, T. J.; Ding, J. X.; Chen, X. S. Electrospun polymer micro/nanofibers as pharmaceutical repositories for healthcare. J. Controlled Release 2019, 302, 19–41.

[23]

Kumbar, S. G.; Nukavarapu, S. P.; James, R.; Nair, L. S.; Laurencin, C. T. Electrospun poly (lactic acid-co-glycolic acid) scaffolds for skin tissue engineering. Biomaterials 2008, 29, 4100–4107.

[24]

Zhu, X. L.; Cui, W. G.; Li, X. H.; Jin, Y. Electrospun fibrous mats with high porosity as potential scaffolds for skin tissue engineering. Biomacromolecules 2008, 9, 1795–1801.

[25]

Zhong, H. L.; Huang, J.; Wu, J.; Du, J. H. Electrospinning nanofibers to 1D, 2D, and 3D scaffolds and their biomedical applications. Nano Res. 2021, 15, 787–804.

[26]

Zhang, Q. K.; Oh, J. H.; Park, C. H.; Baek, J. H.; Ryoo, H. M.; Woo, K. M. Effects of dimethyloxalylglycine-embedded poly(ε-caprolactone) fiber meshes on wound healing in diabetic rats. ACS Appl. Mater. Interfaces 2017, 9, 7950–7963.

[27]

Fattahi, P.; Dover, J. T.; Brown, J. L. 3D near-field electrospinning of biomaterial microfibers with potential for blended microfiber-cell-loaded gel composite structures. Adv. Healthcare Mater. 2017, 6, 1700456.

[28]

Rothrauff, B. B.; Lauro, B. B.; Yang, G.; Debski, R. E.; Musahl, V.; Tuan, R. S. Braided and stacked electrospun nanofibrous scaffolds for tendon and ligament tissue engineering. Tissue Eng. Part A 2017, 23, 378–389.

[29]

Semenza, G. L. Pharmacologic targeting of hypoxia-inducible factors. Annu. Rev. Pharmacol. Toxicol. 2019, 59, 379–403.

[30]

Chen, H.; Jia, P.; Kang, H.; Zhang, H. B.; Liu, Y.; Yang, P. L.; Yan, Y. F.; Zuo, G. L.; Guo, L.; Jiang, M. et al. Upregulating HIF-1α by hydrogel nanofibrous scaffolds for rapidly recruiting angiogenesis relative cells in diabetic wound. Adv. Healthcare Mater. 2016, 5, 907–918.

[31]

Zhu, F.; Zi, L.; Yang, P.; Wei, Y.; Zhong, R.; Wang, Y.; You, C.; Li, Y. W.; Tian, M.; Gu, Z. P. Efficient iron and ROS nanoscavengers for brain protection after intracerebral hemorrhage. ACS Appl. Mater. Interfaces 2021, 13, 9729–9738.

[32]

Gao, S. Q.; Chang, C.; Li, J. J.; Li, Y.; Niu, X. Q.; Zhang, D. P.; Li, L. J.; Gao, J. Q. Co-delivery of deferoxamine and hydroxysafflor yellow A to accelerate diabetic wound healing via enhanced angiogenesis. Drug Delivery 2018, 25, 1779–1789.

[33]

Xu, Z. J.; Liang, B.; Tian, J. Z.; Wu, J. Anti-inflammation biomaterial platforms for chronic wound healing. Biomater. Sci. 2021, 9, 4388–4409.

[34]

Xu, Z. J.; Liu, G. T.; Huang, J.; Wu, J. Novel glucose-responsive antioxidant hybrid hydrogel for enhanced diabetic wound repair. ACS Appl. Mater. Interfaces 2022, 14, 7680–7689.

[35]

Lu, N. H.; Chen, W.; Peng, Y. Y. Effects of glutathione, trolox and desferrioxamine on hemoglobin-induced protein oxidative damage: Anti-oxidant or pro-oxidant? Eur. J. Pharmacol. 2011, 659, 95–101.

[36]

Baysal, E.; Monteiro, H. P.; Sullivan, S. G.; Stern, A. Desferrioxamine protects human red blood cells from hemin-induced hemolysis. Free Radical Biol. Med. 1990, 9, 5–10.

[37]

Hu, C.; Long, L. Y.; Cao, J.; Zhang, S. M.; Wang, Y. B. Dual-crosslinked mussel-inspired smart hydrogels with enhanced antibacterial and angiogenic properties for chronic infected diabetic wound treatment via pH-responsive quick cargo release. Chem. Eng. J. 2021, 411, 128564.

[38]

Lv, F.; Wang, J.; Xu, P.; Han, Y. M.; Ma, H. S.; Xu, H.; Chen, S. J.; Chang, J.; Ke, Q. F.; Liu, M. Y. et al. A conducive bioceramic/polymer composite biomaterial for diabetic wound healing. Acta Biomater. 2017, 60, 128–143.

[39]

Liu, J. H.; Yan, L. W.; Yang, W.; Lan, Y.; Zhu, Q. Y.; Xu, H. J.; Zheng, C. B.; Guo, R. Controlled-release neurotensin-loaded silk fibroin dressings improve wound healing in diabetic rat model. Bioact. Mater. 2019, 4, 151–159.

[40]

Morgado, P. I.; Aguiar-Ricardo, A.; Correia, I. J. Asymmetric membranes as ideal wound dressings: An overview on production methods, structure, properties and performance relationship. J. Membr. Sci. 2015, 490, 139–151.

[41]

Qiao, Z. W.; Lv, X. L.; He, S. H.; Bai, S. M.; Liu, X. C.; Hou, L. X.; He, J. J.; Tong, D. M.; Ruan, R. J.; Zhang, J. et al. A mussel-inspired supramolecular hydrogel with robust tissue anchor for rapid hemostasis of arterial and visceral bleedings. Bioact. Mater. 2021, 6, 2829–2840.

[42]

Li, Z. T.; Huang, X. H.; Lin, L. C.; Jiao, Y. P.; Zhou, C. R.; Liu, Z. H. Polyphenol and Cu2+ surface-modified chitin sponge synergizes with antibacterial, antioxidant and pro-vascularization activities for effective scarless regeneration of burned skin. Chem. Eng. J. 2021, 419, 129488.

[43]

Lo, W. C.; Chen, C. C.; Fuh, Y. K. 3D stacked near-field electrospun nanoporous PVDF-TrFE nanofibers as self-powered smart sensing in gait big data analytics. Adv. Mater. Technol. 2021, 6, 2000779.

[44]

Huang, Y.; You, X. Y.; Tang, Z. Q.; Tong, K. Y.; Guo, P.; Zhao, N. Interface engineering of flexible piezoresistive sensors via near-field electrospinning processed spacer layers. Small Methods 2021, 5, 2000842.

[45]

Kong, L. Z.; Wu, Z.; Zhao, H. K.; Cui, H. M.; Shen, J.; Chang, J.; Li, H. Y.; He, Y. H. Bioactive injectable hydrogels containing desferrioxamine and bioglass for diabetic wound healing. ACS Appl. Mater. Interfaces 2018, 10, 30103–30114.

[46]

Zieliński, P. S.; Gudeti, P. K. R.; Rikmanspoel, T.; Włodarczyk-Biegun, M. K. 3D printing of bio-instructive materials: Toward directing the cell. Bioact. Mater. 2023, 19, 292–327.

[47]

Li, R. T.; Liu, K.; Huang, X.; Li, D.; Ding, J. X.; Liu, B.; Chen, X. S. Bioactive materials promote wound healing through modulation of cell behaviors. Adv. Sci. 2022, 9, 2105152.

File
12274_2022_4813_MOESM1_ESM.pdf (660.2 KB)
Publication history
Copyright
Acknowledgements

Publication history

Received: 14 June 2022
Revised: 19 July 2022
Accepted: 25 July 2022
Published: 03 September 2022
Issue date: January 2023

Copyright

© Tsinghua University Press 2022

Acknowledgements

Acknowledgements

We sincerely acknowledge the funding from the Science and Technology Planning Project of Shenzhen Municipality (Nos. JCYJ20180306174831458 and JCYJ20190807155801657), the National Natural Science Foundation of China (Nos. 51973243 and 52173150), Guangdong Innovative and Entrepreneurial Research Team Program (No. 2016ZT06S029), and Key international (regional) cooperative research projects of the National Natural Science Foundation of China (No. 5181001045). The International Science and technology cooperation project of Shenzhen Municipality (No. GJHZ20200731095008026), Public welfare project of Futian District Health Bureau (No. FTWS2021001).

Return