AccScience Publishing / IJB / Volume 9 / Issue 3 / DOI: 10.18063/ijb.703
RESEARCH ARTICLE

Integrating zinc/silicon dual ions with 3D-printed GelMA hydrogel promotes in situ hair follicle regeneration

Fanliang Zhang1† Zhaowenbin Zhang2† Xianlan Duan1 Wei Song1 Zhao Li1 Bin Yao1 Yi Kong1 Xing Huang3 Xiaobing Fu1 Jiang Chang2,4* Sha Huang1*
Show Less
1 Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, Beijing, 100853, China
2 Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
3 Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
4 State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
Submitted: 25 November 2022 | Accepted: 13 January 2023 | Published: 8 March 2023
© 2023 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Correction

Correction is available for this article: view correction

Abstract

The regeneration of hair follicles lost from injury or disease represents a major challenge in cutaneous regenerative medicine. In this study, we investigated the synergetic effects between zinc and silicon ions on dermal cells and screened the optimal concentration of ions for medical applications. We integrated zinc/silicon dual ions into gelatin methacryloyl (GelMA) to bioprint a scaffold and determined that its mechanical properties are suitable for biological treatment. Then, the scaffold was employed to treat mouse excisional model in order to promote in situ hair follicle regeneration. Our findings showed that GelMA-zinc/silicon-printed hydrogel can significantly activate hair follicle stem cells and enhance neovascularization. The beneficial effects of the scaffold were further confirmed by the growth of hairs in the center of wounds and the improvement in perfusion recovery. Taken together, the present study is the first to combine GelMA with zinc/silicon dual ions to bioprint in situ for treating excisional wound, and this approach may regulate hair follicle regeneration not only directly by impacting stem cells but also indirectly through promoting angiogenesis.

Keywords
Zinc and silicon ions
3D bioprinting
GelMA
Hair follicle regeneration
Angiogenesis
References

1. Gurtner GC, Werner S, Barrandon Y, et al., 2008, Wound repair and regeneration. Nature, 453(7193):314–321. https://doi.org/10.1038/nature07039 

2. Takeo M, Lee W, Ito M, 2015, Wound healing and skin regeneration. CSH Perspect Med, 5(1):a023267. https://doi.org/10.1101/cshperspect.a023267 

3. Chuong CM, Randall VA, Widelitz RB, et al., 2012, Physiological regeneration of skin appendages and implications for regenerative medicine. Physiology, 27(2):61–72. https://doi.org/10.1152/physiol.00028.2011 

4. Ito M, Yang Z, Andl T, et al., 2007, Wnt-dependent de novo hair follicle regeneration in adult mouse skin after wounding. Nature, 447(7142):316–320. https://doi.org/10.1038/nature05766 

5. Lim CH, Sun Q, Ratti K, et al., 2018, Hedgehog stimulates hair follicle neogenesis by creating inductive dermis during murine skin wound healing. Nat Commun, 9(1):4903. https://doi.org/10.1038/s41467-018-07142-9 

6. Xing M, Jiang Y, Bi W, et al., 2021, Strontium ions protect hearts against myocardial ischemia/reperfusion injury. Sci Adv, 7(3):eabe0726. https://doi.org/10.1126/sciadv.abe0726 

7. Huang T, Zhang T, Jiang X, et al., 2021, Iron oxide nanoparticles augment the intercellular mitochondrial transfer-mediated therapy. Sci Adv, 7(40):eabj0534. https://doi.org/10.1126/sciadv.abj0534

8. Zhang Z, Li W, Liu Y, et al., 2021, Design of a biofluid-absorbing bioactive sandwich-structured Zn-Si bioceramic composite wound dressing for hair follicle regeneration and skin burn wound healing. Bioact Mater, 6(7):1910–1920. https://doi.org/10.1016/j.bioactmat.2020.12.006

9. Wang X, Gao L, Han Y, et al., 2018, Silicon-enhanced adipogenesis and angiogenesis for vascularized adipose tissue engineering. Adv Sci, 5(11):1800776. https://doi.org/10.1002/advs.201800776 

10. Li H, He J, Yu H, et al., 2016, Bioglass promotes wound healing by affecting gap junction connexin 43 mediated endothelial cell behavior. Biomaterials, 84:64–75. https://doi.org/10.1016/j.biomaterials.2016.01.033 

11. Sun Y, You Y, Jiang W, et al., 2020, 3D bioprinting dual-factor releasing and gradient-structured constructs ready to implant for anisotropic cartilage regeneration. Sci Adv, 6(37):eaay1422. https://doi.org/10.1126/sciadv.aay1422 

12. Kong L, Wu Z, Zhao H, et al., 2018, Bioactive injectable hydrogels containing desferrioxamine and bioglass for diabetic wound healing. ACS Appl Mater Interfaces, 10(36):30103–30114. https://doi.org/10.1021/acsami.8b09191 

13. Yue K, Santiago GT, Alvarez MM, et al., 2015, Synthesis, properties, and biomedical applications of gelatin methacryloyl (GelMA) hydrogels. Biomaterials, 73:254–271. https://doi.org/10.1016/j.biomaterials.2015.08.045 

14. Duan X, Yuan X, Yao B, et al., 2022, The role of CTHRC1 in promotion of cutaneous wound healing. Signal Transduct Target Ther, 7(1):183. https://doi.org/10.1038/s41392-022-01008-9 

15. Yuan X, Duan X, Enhejirigala, et al., 2023, Reciprocal interaction between vascular niche and sweat gland promotes sweat gland regeneration. Bioact Mater, 21: 340–357. https://doi.org/10.1016/j.bioactmat.2022.08.021 

16. Hu T, Cui X, Zhu M, et al., 2020, 3D-printable supramolecular hydrogels with shear-thinning property: Fabricating strength tunable bioink via dual crosslinking. Bioact Mater, 5(4):808–818. https://doi.org/10.1016/j.bioactmat.2020.06.001 

17. Zhou F, Hong Y, Liang R, et al., 2020, Rapid printing of bio-inspired 3D tissue constructs for skin regeneration. Biomaterials, 258:120287. https://doi.org/10.1016/j.biomaterials.2020.120287 

18. Chen M, Wu Y, Chen B, et al., 2022, Fast, strong, and reversible adhesives with dynamic covalent bonds for potential use in wound dressing. Proc Natl Acad Sci U S A, 119(29):e2203074119. https://doi.org/10.1073/pnas.2203074119 

19. Xia S, Weng T, Jin R, et al., 2022, Curcumin-incorporated 3D bioprinting gelatin methacryloyl hydrogel reduces reactive oxygen species-induced adipose-derived stem cell apoptosis and improves implanting survival in diabetic wounds. Burns Trauma, 10:tkac001. https://doi.org/10.1093/burnst/tkac001 

20. Galiano RD, Michaels J, Dobryansky M, et al., 2004, Quantitative and reproducible murine model of excisional wound healing. Wound Repair Regen, 12(4):485–492. https://doi.org/10.1111/j.1067-1927.2004.12404.x 

21. Yao B, Wang R, Wang Y, et al., 2020, Biochemical and structural cues of 3D-printed matrix synergistically direct MSC differentiation for functional sweat gland regeneration. Sci Adv, 6(10):eaaz1094. https://doi.org/10.1126/sciadv.aaz1094 

22. Gianni-Barrera R, Butschkau A, Uccelli A, et al., 2018, PDGF-BB regulates splitting angiogenesis in skeletal muscle by limiting VEGF-induced endothelial proliferation. Angiogenesis, 21(4):883–900. https://doi.org/10.1007/s10456-018-9634-5 

23. Yano K, Brown LF, Detmar M, 2001, Control of hair growth and follicle size by VEGF-mediated angiogenesis. J Clin Invest, 107(4):409–417. https://doi.org/10.1172/jci11317 

24. Fredriksson I, Larsson M, Strömberg T, 2009, Measurement depth and volume in laser Doppler flowmetry. Microvasc Res, 78(1):4–13. https://doi.org/10.1016/j.mvr.2009.02.008 

25. Abbasi S, Sinha S, Labit E, et al., 2020, Distinct regulatory programs control the latent regenerative potential of dermal fibroblasts during wound healing. Cell Stem Cell, 27(3): 396–412. https://doi.org/10.1016/j.stem.2020.07.008 

26. Brewer CM, Nelson BR, Wakenight P, et al., 2021, Adaptations in Hippo-Yap signaling and myofibroblast fate underlie scar-free ear appendage wound healing in spiny mice. Dev Cell, 56(19):2722–2740. https://doi.org/10.1016/j.devcel.2021.09.008 

27. Fernandes KJ, McKenzie IA, Mill P, et al., 2004, A dermal niche for multipotent adult skin-derived precursor cells. Nat Cell Biol, 6(11):1082–1093. https://doi.org/10.1038/ncb1181 

28. Grellier M, Bordenave L, Amedee J, 2009, Cell-to-cell communication between osteogenic and endothelial lineages: implications for tissue engineering. Trends Biotechnol, 27(10):562–571. https://doi.org/10.1016/j.tibtech.2009.07.001 

29. Griffin DR, Archang MM, Kuan CH, et al., 2020, Activating an adaptive immune response from a hydrogel scaffold imparts regenerative wound healing. Nat Mater, 20(4):560–569. https://doi.org/10.1038/s41563-020-00844-w

30. DiPietro LA, 2013, Angiogenesis and scar formation in healing wounds. Curr Opin Rheumatol, 25(1):87–91. https://doi.org/10.1097/BOR.0b013e32835b13b6 

31. Shabbir A, Cox A, Rodriguez-Menocal L, et al., 2015, Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro. Stem Cells Dev, 24(14):1635– 1647. https://doi.org/10.1089/scd.2014.0316 

32. Rezza A, Sennett R, Tanguy M, et al., 2015, PDGF signalling in the dermis and in dermal condensates is dispensable for hair follicle induction and formation. Exp Dermatol, 24(6):468–470. https://doi.org/10.1111/exd.12672 

33. Richardson GD, Bazzi H, Fantauzzo KA, et al., 2009, KGF and EGF signalling block hair follicle induction and promote interfollicular epidermal fate in developing mouse skin. Development, 136(13):2153–2164. https://doi.org/10.1242/dev.031427 

34. Veith AP, Henderson K, Spencer A, et al., 2019, Therapeutic strategies for enhancing angiogenesis in wound healing. Adv Drug Deliv Rev, 146:97–125. https://doi.org/10.1016/j.addr.2018.09.010 

35. Roche B, Vanden-Bossche A, Normand M, et al., 2013, Validated laser Doppler protocol for measurement of mouse bone blood perfusion—Response to age or ovariectomy differs with genetic background. Bone, 55(2):418–426. https://doi.org/10.1016/j.bone.2013.03.022
 
36. Choi YS, Zhang Y, Xu M, et al., 2013, Distinct functions for Wnt/β-catenin in hair follicle stem cell proliferation and survival and interfollicular epidermal homeostasis. Cell Stem Cell, 13(6):720–733. https://doi.org/10.1016/j.stem.2013.10.003 

37. Plikus MV, Mayer JA, de la Cruz D, et al., 2008, Cyclic dermal BMP signalling regulates stem cell activation during hair regeneration. Nature, 451(7176):340–344. https://doi.org/10.1038/nature06457 

38. Zhang L, Wang WH, Jin JY, et al., 2019, Induction of hair follicle neogenesis with cultured mouse dermal papilla cells in de novo regenerated skin tissues. J Tissue Eng Regen Med, 13(9):1641–1650. https://doi.org/10.1002/term.2918 

39. Harn HI, Wang SP, Lai YC, et al., 2021, Symmetry breaking of tissue mechanics in wound induced hair follicle regeneration of laboratory and spiny mice. Nat Commun, 12(1):2595. https://doi.org/10.1038/s41467-021-22822-9 

40. Zhai W, Lu H, Chen L, et al., 2012, Silicate bioceramics induce angiogenesis during bone regeneration. Acta Biomater, 8(1):341–349. https://doi.org/10.1016/j.actbio.2011.09.008 

41. Xue Y, Lim CH, Plikus MV, et al., 2022, Wound-induced hair neogenesis model. J Invest Dermatol, 142(10):2565– 2569. https://doi.org/10.1016/j.jid.2022.07.013 

42. Gay D, Kwon O, Zhang Z, et al., 2013, Fgf9 from dermal gammadelta T cells induces hair follicle neogenesis after wounding. Nat Med, 19(7):916–923. https://doi.org/10.1038/nm.3181 

43. Chueh SC, Lin SJ, Chen CC, et al., 2013, Therapeutic strategy for hair regeneration: Hair cycle activation, niche environment modulation, wound-induced follicle neogenesis, and stem cell engineering. Expert Opin Biol Ther, 13(3):377–391. https://doi.org/10.1517/14712598.2013.739601 

44. Li KN, Tumbar T, 2021, Hair follicle stem cells as a skin-organizing signaling center during adult homeostasis. Embo J, 40(11):e107135. https://doi.org/10.15252/embj.2020107135 

45. Guerra A, Belinha J, Jorge RN, 2018, Modelling skin wound healing angiogenesis: A review. J Theor Biol, 459:1–17. https://doi.org/10.1016/j.jtbi.2018.09.020 

46. Bin BH, Bhin J, Takaishi M, et al., 2017, Requirement of zinc transporter ZIP10 for epidermal development: Implication of the ZIP10-p63 axis in epithelial homeostasis. Proc Natl Acad Sci U S A, 114(46):12243–12248. https://doi.org/10.1073/pnas.1710726114 

47. Ma Y, Lin M, Huang G, et al., 2018, 3D Spatiotemporal mechanical microenvironment: A hydrogel-based platform for guiding stem cell fate. Adv Mater, 30(49):e1705911. https://doi.org/10.1002/adma.201705911

Share
Back to top
International Journal of Bioprinting, Electronic ISSN: 2424-8002 Print ISSN: 2424-7723, Published by AccScience Publishing