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

Self-assembling peptide RADA16: a promising scaffold for tissue engineering and regenerative medicine

    Xin Yao‡

    Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China

    Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China

    ‡Contributed equally to this work

    Search for more papers by this author

    ,
    Yicun Hu‡

    Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China

    Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China

    ‡Contributed equally to this work

    Search for more papers by this author

    ,
    Maoqiang Lin

    Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China

    Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China

    ,
    Kaichen Peng

    Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China

    Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China

    ,
    Peng Wang

    Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China

    Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China

    ,
    Yanbing Gao

    Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China

    Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China

    ,
    Xidan Gao

    Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, Shaanxi, China

    ,
    Taowen Guo

    Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China

    Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China

    ,
    Xiaobo Zhang

    *Author for correspondence:

    E-mail Address: 845808289@qq.com

    Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710000, Shaanxi, China

    &
    Haiyu Zhou

    **Author for correspondence: Tel.: +86 158 2733 4208;

    E-mail Address: zhouhy@lzu.edu.cn

    Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China

    Key Laboratory of Bone & Joint Disease Research of Gansu Provincial, Lanzhou 730030, Gansu, China

    Published Online:https://doi.org/10.2217/nnm-2023-0161

    RADA16 is a peptide-based biomaterial whose acidic aqueous solution spontaneously forms an extracellular matrix-like 3D structure within seconds upon contact with physiological pH body fluids. Meanwhile, its good biocompatibility, low immunogenicity, nontoxic degradation products and ease of modification make it an ideal scaffold for tissue engineering. RADA16 is a good delivery vehicle for cells, drugs and factors. Its shear thinning and thixotropic properties allow it to fill tissue voids by injection and not to swell. However, the weaker mechanical properties and poor hydrophilicity are troubling limitations of RADA16. To compensate for this limitation, various functional groups and polymers have been designed to modify RADA16, thus contributing to its scope and progress in the field of tissue engineering.

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

    References

    • 1. Langer R, Vacanti JP. Tissue engineering. Science 260(5110), 920–926 (1993).
    • 2. Williams DJ, Sebastine IM. Tissue engineering and regenerative medicine: manufacturing challenges. IEE Proc. Nanobiotechnol. 152(6), 207–210 (2005).
    • 3. Gao C, Seuntjens J, Kaufman GN et al. Mesenchymal stem cell transplantation to promote bone healing. J. Orthopaedic Res. 30(8), 1183–1189 (2012).
    • 4. Eugenis I, Wu D, Rando TA. Cells, scaffolds, and bioactive factors: engineering strategies for improving regeneration following volumetric muscle loss. Biomaterials 278, 121173 (2021).
    • 5. Grover CN, Cameron RE, Best SM. Investigating the morphological, mechanical and degradation properties of scaffolds comprising collagen, gelatin and elastin for use in soft tissue engineering. J. Mech. Behav. Biomed. Mater. 10, 62–74 (2012).
    • 6. Loh QL, Choong C. Three-dimensional scaffolds for tissue engineering applications: role of porosity and pore size. Tissue Eng. Part B. Rev. 19(6), 485–502 (2013).
    • 7. Wubneh A, Tsekoura EK, Ayranci C, Uludağ H. Current state of fabrication technologies and materials for bone tissue engineering. Acta Biomater. 80, 1–30 (2018).
    • 8. Bowles RD, Setton LA. Biomaterials for intervertebral disc regeneration and repair. Biomaterials 129, 54–67 (2017).
    • 9. Chaudhari AA, Vig K, Baganizi DR et al. Future prospects for scaffolding methods and biomaterials in skin tissue engineering: a review. Int. J. Mol. Sci. 17(12), 1974 (2016).
    • 10. Zhang S, Lockshin C, Herbert A, Winter E, Rich A. Zuotin, a putative Z-DNA binding protein in Saccharomyces cerevisiae. EMBO J. 11(10), 3787–3796 (1992).
    • 11. Edwards-Gayle CJC, Hamley IW. Self-assembly of bioactive peptides, peptide conjugates, and peptide mimetic materials. Organic Biomolec. Chem. 15(28), 5867–5876 (2017).
    • 12. Lee S, Trinh THT, Yoo M et al. Self-assembling peptides and their application in the treatment of diseases. Int. J. Mol. Sci. 20(23), 5850 (2019).
    • 13. Dong X, Guo X, Liu G, Fan A, Wang Z, Zhao Y. When self-assembly meets topology: an enhanced micelle stability. Chem. Comm. (Cambridge, England) 53(27), 3822–3825 (2017).
    • 14. Blau WJ, Fleming AJ. Materials science. Designer nanotubes by molecular self-assembly. Science 304(5676), 1457–1458 (2004).
    • 15. Zhang S. Self-assembling peptides: from a discovery in a yeast protein to diverse uses and beyond. Protein Sci. 29(11), 2281–2303 (2020).
    • 16. Siddiqui Z, Sarkar B, Kim KK et al. Angiogenic hydrogels for dental pulp revascularization. Acta Biomaterialia 126, 109–118 (2021).
    • 17. Hong JY, Kim SH, Seo Y et al. Self-assembling peptide gels promote angiogenesis and functional recovery after spinal cord injury in rats. J. Tissue Eng. 13, 20417314221086491 (2022).
    • 18. De Souza Araújo IJ, Ferreira JA, Daghrery A et al. Self-assembling peptide-laden electrospun scaffolds for guided mineralized tissue regeneration. Dent. Mater. 38(11), 1749–1762 (2022).
    • 19. Wu T, Wu Y, Cao Z et al. Cell-free and cytokine-free self-assembling peptide hydrogel-polycaprolactone composite scaffolds for segmental bone defects. Biomater. Sci. 11(3), 840–853 (2023).
    • 20. Huan Y, Kong Q, Tang Q et al. Antimicrobial peptides/ciprofloxacin-loaded O-carboxymethyl chitosan/self-assembling peptides hydrogel dressing with sustained-release effect for enhanced anti-bacterial infection and wound healing. Carbohydr. Polym. 280, 119033 (2022).
    • 21. Zhou Y, Liu S, Zhao M et al. Injectable extracellular vesicle-released self-assembling peptide nanofiber hydrogel as an enhanced cell-free therapy for tissue regeneration. J. Control. Rel. 316, 93–104 (2019).
    • 22. Galler KM, Aulisa L, Regan KR, D'Souza RN, Hartgerink JD. Self-assembling multidomain peptide hydrogels: designed susceptibility to enzymatic cleavage allows enhanced cell migration and spreading. J. Am. Chem. Soc. 132(9), 3217–3223 (2010).
    • 23. Marchini A, Favoino C, Gelain F. Multi-functionalized self-assembling peptides as reproducible 3D cell culture systems enabling differentiation and survival of various human neural stem cell lines. Front. Neurosci. 14, 413 (2020).
    • 24. Pugliese R, Gelain F. Peptidic biomaterials: from self-assembling to regenerative medicine. Trends Biotechnol. 35(2), 145–158 (2017).
    • 25. Guo W, Ma Y, Hu L et al. Modification strategies for ionic complementary self-assembling peptides: taking RADA16-I as an example. Polymers 14(23), 5221 (2022).
    • 26. Mendes AC, Baran ET, Reis RL, Azevedo HS. Self-assembly in nature: using the principles of nature to create complex nanobiomaterials. Wiley Interdisciplinary Reviews. Nanomed. Nanobiotechnol. 5(6), 582–612 (2013).
    • 27. Zhang S. Emerging biological materials through molecular self-assembly. Biotechnol. Adv. 20(5–6), 321–339 (2002).
    • 28. Jonker AM, Löwik DWPM, Van Hest JCM. Peptide- and protein-based hydrogels. Chem. Mater. 24(5), 759–773 (2012).
    • 29. Cormier AR, Ruiz-Orta C, Alamo RG, Paravastu AK. Solid state self-assembly mechanism of RADA16-I designer peptide. Biomacromolecules 13(6), 1794–1804 (2012).
    • 30. Altman M, Lee P, Rich A, Zhang S. Conformational behavior of ionic self-complementary peptides. Protein Sci. 9(6), 1095–1105 (2000).
    • 31. Holmes TC. Novel peptide-based biomaterial scaffolds for tissue engineering. Trends Biotechnol. 20(1), 16–21 (2002).
    • 32. Li R, Pang Z, He H et al. Drug depot-anchoring hydrogel: a self-assembling scaffold for localized drug release and enhanced stem cell differentiation. J. Control. Rel. 261, 234–245 (2017). •• A study of drug release from RADA16 hydrogels and stem cell differentiation. Innovatively anchored PLGA microspheres to RADA16 hydrogels, which is a covalent binding mode. A very effective dual-delivery system model (microspheres encapsulating drug and hydrogel-delivering cells) is provided in terms of drug release and stem cell differentiation.
    • 33. Tang C, Shao X, Sun B, Huang W, Zhao X. The effect of self-assembling peptide RADA16-I on the growth of human leukemia cells in vitro and in nude mice. Int. J. Mol. Sci. 10(5), 2136–2145 (2009).
    • 34. Song H, Zhang L, Zhao X. Hemostatic efficacy of biological self-assembling peptide nanofibers in a rat kidney model. Macromol. Biosci. 10(1), 33–39 (2010).
    • 35. Zhao Y, Yokoi H, Tanaka M, Kinoshita T, Tan T. Self-assembled pH-responsive hydrogels composed of the RATEA16 peptide. Biomacromolecules 9(6), 1511–1518 (2008).
    • 36. Wu D, Zhang S, Zhao Y, Ao N, Ramakrishna S, He L. The effects of motif net charge and amphiphilicity on the self-assembly of functionally designer RADA16-I peptides. Biomed. Mater. 13(3), 035011 (2018).
    • 37. Loo Y, Zhang S, Hauser CA. From short peptides to nanofibers to macromolecular assemblies in biomedicine. Biotechnol. Adv. 30(3), 593–603 (2012).
    • 38. Cormier AR, Pang X, Zimmerman MI, Zhou HX, Paravastu AK. Molecular structure of RADA16-I designer self-assembling peptide nanofibers. ACS Nano 7(9), 7562–7572 (2013).
    • 39. Yokoi H, Kinoshita T, Zhang S. Dynamic reassembly of peptide RADA16 nanofiber scaffold. Proc. Natl Acad. Sci. USA 102(24), 8414–8419 (2005).
    • 40. Wang TW, Chang KC, Chen LH, Liao SY, Yeh CW, Chuang YJ. Effects of an injectable functionalized self-assembling nanopeptide hydrogel on angiogenesis and neurogenesis for regeneration of the central nervous system. Nanoscale 9(42), 16281–16292 (2017).
    • 41. Cormier AR, Lopez-Majada JM, Alamo RG, Paravastu AK. Distinct solid and solution state self-assembly pathways of RADA16-I designer peptide. J. Peptide Sci. 19(8), 477–484 (2013).
    • 42. Sankar S, O'Neill K, Bagot D'Arc M et al. Clinical use of the self-assembling peptide RADA16: a review of current and future trends in biomedicine. Front. Bioeng. Biotechnol. 9, 679525 (2021).
    • 43. Koutsopoulos S. Self-assembling peptide nanofiber hydrogels in tissue engineering and regenerative medicine: progress, design guidelines, and applications. J. Biomed. Mater. Res. A 104(4), 1002–1016 (2016).
    • 44. Ye Z, Zhang H, Luo H et al. Temperature and pH effects on biophysical and morphological properties of self-assembling peptide RADA16-I. J. Peptide Sci. 14(2), 152–162 (2008).
    • 45. He B, Ou Y, Chen S et al. Designer bFGF-incorporated D-form self-assembly peptide nanofiber scaffolds to promote bone repair. Mater. Sci. Eng. C. Mater. Biol. Appl. 74, 451–458 (2017).
    • 46. Zhou A, Chen S, He B, Zhao W, Chen X, Jiang D. Controlled release of TGF-beta 1 from RADA self-assembling peptide hydrogel scaffolds. Drug Design Develop. Therapy 10, 3043–3051 (2016).
    • 47. Wu M, Ye Z, Zhu H, Zhao X. Self-assembling peptide nanofibrous hydrogel on immediate hemostasis and accelerative osteosis. Biomacromolecules 16(10), 3112–3118 (2015).
    • 48. Marchesan S, Easton CD, Styan KE et al. Chirality effects at each amino acid position on tripeptide self-assembly into hydrogel biomaterials. Nanoscale 6(10), 5172–5180 (2014).
    • 49. Chen S, Zhou A, He B, Zhao W, Chen X, Jiang D. Designer D-form self-assembling peptide scaffolds promote the proliferation and migration of rat bone marrow-derived mesenchymal stem cells. Int. J. Mol. Med. 40(3), 679–688 (2017).
    • 50. Friedman M. Chemistry, nutrition, and microbiology of D-amino acids. J. Agri. Food Chem. 47(9), 3457–3479 (1999).
    • 51. Nagai Y, Unsworth LD, Koutsopoulos S, Zhang S. Slow release of molecules in self-assembling peptide nanofiber scaffold. J. Control. Rel. 115(1), 18–25 (2006).
    • 52. Wei W, Meng C, Wang Y et al. The interaction between self-assembling peptides and emodin and the controlled release of emodin from in situ hydrogel. Artific. Cells Nanomed. Biotechnol. 47(1), 3961–3975 (2019).
    • 53. Wei W, Tang J, Hu L et al. Experimental anti-tumor effect of emodin in suspension – in situ hydrogels formed with self-assembling peptide. Drug Delivery 28(1), 1810–1821 (2021).
    • 54. Gelain F, Unsworth LD, Zhang S. Slow and sustained release of active cytokines from self-assembling peptide scaffolds. J. Control. Rel. 145(3), 231–239 (2010).
    • 55. Tian H, Guo A, Li K, Tao B, Lei D, Deng Z. Effects of a novel self-assembling peptide scaffold on bone regeneration and controlled release of two growth factors. J. Biomed. Mater. Res. A 110(4), 943–953 (2022).
    • 56. Huang LC, Wang HC, Chen LH et al. Bioinspired self-assembling peptide hydrogel with proteoglycan-assisted growth factor delivery for therapeutic angiogenesis. Theranostics 9(23), 7072–7087 (2019). • A study of functionalized RADA16 hydrogels in angiogenesis. The experiment is designed with two novel functionalized RADA16 hydrogels. This study confirms that these two functionalized RADA16 hydrogels can induce proper angiogenesis under tissue ischemia to provide adequate vascular supply to the injury site.
    • 57. Luo H, Xu C, Liu Z et al. Neural differentiation of bone marrow mesenchymal stem cells with human brain-derived neurotrophic factor gene-modified in functionalized self-assembling peptide hydrogel in vitro. J. Cell. Biochem. 120(3), 2828–2835 (2019).
    • 58. Liu H, Xu X, Tu Y et al. Engineering microenvironment for endogenous neural regeneration after spinal cord injury by reassembling extracellular matrix. ACS Appl. Mater. Interfaces 12(15), 17207–17219 (2020).
    • 59. Liu X, Ren H, Peng A et al. The effect of RADA16-I and CDNF on neurogenesis and neuroprotection in brain ischemia-reperfusion injury. Int. J. Mol. Sci. 23(3), 1436 (2022).
    • 60. Wang Y, Wang J, Gao R et al. Biomimetic glycopeptide hydrogel coated PCL/nHA scaffold for enhanced cranial bone regeneration via macrophage M2 polarization-induced osteo-immunomodulation. Biomaterials 285, 121538 (2022). •• Presents the first noncovalent composite of RADA16 with polycaprolactone/nanohydroxyapatite (PCL/nHA) scaffolds for bone regeneration studies. Also, 3D printing of RADA16 was used innovatively in this study.
    • 61. Kim JH, Jung Y, Kim BS, Kim SH. Stem cell recruitment and angiogenesis of neuropeptide substance P coupled with self-assembling peptide nanofiber in a mouse hind limb ischemia model. Biomaterials 34(6), 1657–1668 (2013).
    • 62. Yang H, Hong N, Liu H, Wang J, Li Y, Wu S. Differentiated adipose-derived stem cell cocultures for bone regeneration in RADA16-I in vitro. J. Cell. Physiol. 233(12), 9458–9472 (2018).
    • 63. Gao XR, Xu HJ, Wang LF, Liu CB, Yu F. Mesenchymal stem cell transplantation carried in SVVYGLR modified self-assembling peptide promoted cardiac repair and angiogenesis after myocardial infarction. Biochem. Biophys. Res. Comm. 491(1), 112–118 (2017).
    • 64. Cai H, Wu FY, Wang QL et al. Self-assembling peptide modified with QHREDGS as a novel delivery system for mesenchymal stem cell transplantation after myocardial infarction. FASEB J. 33(7), 8306–8320 (2019).
    • 65. Ichihara Y, Kaneko M, Yamahara K et al. Self-assembling peptide hydrogel enables instant epicardial coating of the heart with mesenchymal stromal cells for the treatment of heart failure. Biomaterials 154, 12–23 (2018).
    • 66. Chen K, Sahoo S, He P, Ng KS, Toh SL, Goh JC. A hybrid silk/RADA-based fibrous scaffold with triple hierarchy for ligament regeneration. Tissue Eng. Part A 18(13-14), 1399–1409 (2012).
    • 67. Song H, Cai GH, Liang J, Ao DS, Wang H, Yang ZH. Three-dimensional culture and clinical drug responses of a highly metastatic human ovarian cancer HO-8910PM cells in nanofibrous microenvironments of three hydrogel biomaterials. J. Nanobiotechnol. 18(1), 90 (2020).
    • 68. Gelain F, Luo Z, Zhang S. Self-assembling peptide EAK16 and RADA16 nanofiber scaffold hydrogel. Chem. Rev. 120(24), 13434–13460 (2020).
    • 69. Zhang F, Shi GS, Ren LF, Hu FQ, Li SL, Xie ZJ. Designer self-assembling peptide scaffold stimulates pre-osteoblast attachment, spreading and proliferation. J. Mater. Sci. Mater. Med. 20(7), 1475–1481 (2009).
    • 70. Gelain F, Bottai D, Vescovi A, Zhang S. Designer self-assembling peptide nanofiber scaffolds for adult mouse neural stem cell 3-dimensional cultures. PLOS ONE 1(1), e119 (2006).
    • 71. Sun L, Zhao X. A self-assembling peptide RADA16-I integrated with spider fibroin uncrystalline motifs. Int. J. Nanomed. 7, 571–580 (2012).
    • 72. Wei S, Chen F, Geng Z, Cui R, Zhao Y, Liu C. Self-assembling RATEA16 peptide nanofiber designed for rapid hemostasis. J. Mater. Chem. B 8(9), 1897–1905 (2020).
    • 73. Yang X, Zhang Y, Huang C, Lu L, Chen J, Weng Y. Biomimetic hydrogel scaffolds with copper peptide-functionalized RADA16 nanofiber improve wound healing in diabetes. Macromol. Biosci. 22(8), e2200019 (2022).
    • 74. Wu X, He L, Li W et al. Functional self-assembling peptide nanofiber hydrogel for peripheral nerve regeneration. Regen. Biomater. 4(1), 21–30 (2017).
    • 75. Pan H, Hao S, Zheng Q et al. Bone induction by biomimetic PLGA copolymer loaded with a novel synthetic RADA16-P24 peptide in vivo. Mater. Sci. Eng. C. Mater. Biol. Appl. 33(6), 3336–3345 (2013).
    • 76. Li Y, Zhang J, Chen L, Li H, Wang J. Repair of critical-sized rat cranial defects with RADA16-W9 self-assembled peptide hydrogel. Biochem. Biophys. Res. Comm. 652, 68–75 (2023).
    • 77. Taraballi F, Natalello A, Campione M et al. Glycine-spacers influence functional motifs exposure and self-assembling propensity of functionalized substrates tailored for neural stem cell cultures. Front. Neuroeng. 3, 1 (2010).
    • 78. Feng F, Song X, Tan Z et al. Cooperative assembly of a designer peptide and silk fibroin into hybrid nanofiber gels for neural regeneration after spinal cord injury. Sci. Adv. 9(25), eadg0234 (2023). •• Innovatively prepares a composite hydrogel by mixing RADA16 hydrogel with other hydrogels (SF). The study shows that the composite hydrogel has great potential as a long-term in vivo scaffold for the treatment of spinal cord injury. This study provides new ideas for RADA16 improvement strategies.
    • 79. Xiao L, Xie P, Ma J et al. A bioinspired injectable, adhesive and self-healing hydrogel with dual hybrid network for neural regeneration after spinal cord injury. Adv. Mater. doi:10.1002/adma.202304896 (2023).
    • 80. Fu XB. Repair cell first, then regenerate the tissues and organs. Military Med. Res. 8(1), 2 (2021).
    • 81. Galliot B, Crescenzi M, Jacinto A, Tajbakhsh S. Trends in tissue repair and regeneration. Development 144(3), 357–364 (2017).
    • 82. Ghandy N, Ebrahimzadeh-Bideskan A, Gorji A, Negah SS. Co-transplantation of novel Nano-SDF scaffold with human neural stem cells attenuates inflammatory responses and apoptosis in traumatic brain injury. Int. Immunopharmacol. 115, 109709 (2023).
    • 83. Yang G, Huang T, Wang Y et al. Sustained release of antimicrobial peptide from self-assembling hydrogel enhanced osteogenesis. J. Biomater. Sci. Polymer Ed. 29(15), 1812–1824 (2018).
    • 84. Mi K, Wang G, Liu Z, Feng Z, Huang B, Zhao X. Influence of a self-assembling peptide, RADA16, compared with collagen I and Matrigel on the malignant phenotype of human breast-cancer cells in 3D cultures and in vivo. Macromol. Biosci. 9(5), 437–443 (2009).
    • 85. Meng H, Chen L, Ye Z, Wang S, Zhao X. The effect of a self-assembling peptide nanofiber scaffold (peptide) when used as a wound dressing for the treatment of deep second degree burns in rats. J. Biomed. Mater. Res. Part B Appl. Biomater. 89(2), 379–391 (2009).
    • 86. Im H, Kim SH, Kim SH, Jung Y. Skin regeneration with a scaffold of predefined shape and bioactive peptide hydrogels. Tissue Eng. Part A 24(19–20), 1518–1530 (2018).
    • 87. Sun Y, Li W, Wu X et al. Functional self-assembling peptide nanofiber hydrogels designed for nerve degeneration. ACS Appl. Mater. Interf. 8(3), 2348–2359 (2016).
    • 88. Pan Q, Li W, Yuan X et al. Chondrogenic effect of cell-based scaffold of self-assembling peptides/PLGA-PLL loading the hTGFβ3 plasmid DNA. J. Mater. Sci. Mater. Med. 27(1), 19 (2016).
    • 89. Tao H, Wu Y, Li H et al. BMP7-based functionalized self-assembling peptides for nucleus pulposus tissue engineering. ACS Appl. Mater. Interf. 7(31), 17076–17087 (2015). • Investigates the role of three functionalized RADA16 hydrogels in myeloid regeneration. Significant potential for nanoparticle regeneration is identified through studies of cellular activity, extracellular matrix secretion, gene expression and protein expression rates. This study not only innovatively designs three functionalized hydrogels, but also expands the research of RADA16 hydrogels in tissue regeneration.
    • 90. Giri S, Nieber K, Acikgöz A, Pavlica S, Keller M, Bader A. Telomerase activity and hepatic functions of rat embryonic liver progenitor cell in nanoscaffold-coated model bioreactor. Mol. Cell. Biochem. 336(1-2), 137–149 (2010).
    • 91. Nyame TT, Chiang HA, Leavitt T, Ozambela M, Orgill DP. Tissue-engineered skin substitutes. Plastic Reconstruct. Surg. 136(6), 1379–1388 (2015).
    • 92. Sorg H, Tilkorn DJ, Hager S, Hauser J, Mirastschijski U. Skin wound healing: an update on the current knowledge and concepts. Eur. Surg. Res. 58(1–2), 81–94 (2017).
    • 93. Op'T Veld RC, Walboomers XF, Jansen JA, Wagener F. Design considerations for hydrogel wound dressings: strategic and molecular advances. Tissue Eng. B. Rev. 26(3), 230–248 (2020).
    • 94. Bradshaw M, Ho D, Fear MW, Gelain F, Wood FM, Iyer KS. Designer self-assembling hydrogel scaffolds can impact skin cell proliferation and migration. Sci. Rep. 4, 6903 (2014).
    • 95. Wang X, Wang J, Guo L et al. Self-assembling peptide hydrogel scaffolds support stem cell-based hair follicle regeneration. Nanomed. Nanotechnol. Biol. Med. 12(7), 2115–2125 (2016).
    • 96. Xue J, Sun N, Liu Y et al. Erratum: self-assembled nano-peptide hydrogels with human umbilical cord mesenchymal stem cell spheroids accelerate diabetic skin wound healing by inhibiting inflammation and promoting angiogenesis [Corrigendum]. Int. J. Nanomed. 17, 3057–3058 (2022).
    • 97. Feng T, Wu H, Ma W et al. An injectable thermosensitive hydrogel with a self-assembled peptide coupled with an antimicrobial peptide for enhanced wound healing. J. Mater. Chem. B 10(32), 6143–6157 (2022).
    • 98. Prokosch V, Liu H, Leibinger M, Fischer D. Regeneration of the optic nerve – will it one day be reality? Die Ophthalmologie 119(9), 919–928 (2022).
    • 99. Nocera G, Jacob C. Mechanisms of Schwann cell plasticity involved in peripheral nerve repair after injury. Cell. Molec. Life Sci. 77(20), 3977–3989 (2020).
    • 100. Sullivan R, Dailey T, Duncan K, Abel N, Borlongan CV. Peripheral nerve injury: stem cell therapy and peripheral nerve transfer. Int. J. Mol. Sci. 17(12), 2101 (2016).
    • 101. Kim HA, Mindos T, Parkinson DB. Plastic fantastic: Schwann cells and repair of the peripheral nervous system. Stem Cell. Transl. Med. 2(8), 553–557 (2013).
    • 102. Nune M, Subramanian A, Krishnan UM, Kaimal SS, Sethuraman S. Self-assembling peptide nanostructures on aligned poly(lactide-co-glycolide) nanofibers for the functional regeneration of sciatic nerve. Nanomedicine (Lond.) 12(3), 219–235 (2017).
    • 103. Nune M, Krishnan UM, Sethuraman S. PLGA nanofibers blended with designer self-assembling peptides for peripheral neural regeneration. Mater. Sci. Eng. C. Mater. Biol. Appl. 62, 329–337 (2016).
    • 104. Yoshimatsu M, Nakamura R, Kishimoto Y et al. Recurrent laryngeal nerve regeneration using a self-assembling peptide hydrogel. Laryngoscope 130(10), 2420–2427 (2020).
    • 105. Frick C, Müller M, Wank U et al. Biofunctionalized peptide-based hydrogels provide permissive scaffolds to attract neurite outgrowth from spiral ganglion neurons. Colloid. Surf. B. Biointerfaces 149, 105–114 (2017).
    • 106. Zhang N, Luo Y, He L, Zhou L, Wu W. A self-assembly peptide nanofibrous scaffold reduces inflammatory response and promotes functional recovery in a mouse model of intracerebral hemorrhage. Nanomed. Nanotechnol. Biol. Med. 12(5), 1205–1217 (2016).
    • 107. Cheng TY, Chen MH, Chang WH, Huang MY, Wang TW. Neural stem cells encapsulated in a functionalized self-assembling peptide hydrogel for brain tissue engineering. Biomaterials 34(8), 2005–2016 (2013).
    • 108. Francis NL, Bennett NK, Halikere A, Pang ZP, Moghe PV. Self-assembling peptide nanofiber scaffolds for 3D reprogramming and transplantation of human pluripotent stem cell-derived neurons. ACS Biomater. Sci. Eng. 2(6), 1030–1038 (2016).
    • 109. Shi W, Huang CJ, Xu XD et al. Transplantation of RADA16-BDNF peptide scaffold with human umbilical cord mesenchymal stem cells forced with CXCR4 and activated astrocytes for repair of traumatic brain injury. Acta Biomaterialia 45, 247–261 (2016).
    • 110. Ni N, Hu Y, Ren H et al. Self-assembling peptide nanofiber scaffolds enhance dopaminergic differentiation of mouse pluripotent stem cells in 3-dimensional culture. PLOS ONE 8(12), e84504 (2013).
    • 111. Francis NL, Zhao N, Calvelli HR et al. Peptide-based scaffolds for the culture and transplantation of human dopaminergic neurons. Tissue Eng. Part A 26(3–4), 193–205 (2020).
    • 112. Li J, Ji Z, Wang Y et al. Human adipose-derived stem cells combined with nano-hydrogel promote functional recovery after spinal cord injury in rats. Biology 11(5), 781 (2022).
    • 113. Wang J, Zheng J, Zheng Q et al. FGL-functionalized self-assembling nanofiber hydrogel as a scaffold for spinal cord-derived neural stem cells. Mater. Sci. Eng. C. Mater. Biol. Appl. 46, 140–147 (2015).
    • 114. Cigognini D, Satta A, Colleoni B et al. Evaluation of early and late effects into the acute spinal cord injury of an injectable functionalized self-assembling scaffold. PLOS ONE 6(5), e19782 (2011).
    • 115. Gelain F, Panseri S, Antonini S et al. Transplantation of nanostructured composite scaffolds results in the regeneration of chronically injured spinal cords. ACS Nano 5(1), 227–236 (2011).
    • 116. Sun X, Liu H, Tan Z et al. Remodeling microenvironment for endogenous repair through precise modulation of chondroitin sulfate proteoglycans following spinal cord injury. Small 19(6), e2205012 (2023).
    • 117. Kim HD, Amirthalingam S, Kim SL, Lee SS, Rangasamy J, Hwang NS. Biomimetic materials and fabrication approaches for bone tissue engineering. Adv. Healthcare Mater. 6(23), 201700612 (2017).
    • 118. Nakahara H, Misawa H, Yoshida A et al. Bone repair using a hybrid scaffold of self-assembling peptide PuraMatrix and polyetheretherketone cage in rats. Cell Transplant. 19(6), 791–797 (2010).
    • 119. Zhao W, He B, Zhou A et al. D-RADA16-RGD-reinforced nano-hydroxyapatite/polyamide 66 ternary biomaterial for bone formation. Tissue Eng. Reg. Med. 16(2), 177–189 (2019).
    • 120. Matsugami D, Murakami T, Yoshida W et al. Treatment with functionalized designer self-assembling peptide hydrogels promotes healing of experimental periodontal defects. J. Periodont. Res. 56(1), 162–172 (2021).
    • 121. Tuan RS, Chen AF, Klatt BA. Cartilage regeneration. J. Am. Acad. Orthopaed. Surg. 21(5), 303–311 (2013).
    • 122. Florine EM, Miller RE, Liebesny PH et al. Delivering heparin-binding insulin-like growth factor 1 with self-assembling peptide hydrogels. Tissue Eng. Part A 21(3-4), 637–646 (2015).
    • 123. Yu P, Duan L, Yan Z, Li J, Cai DZ. RADA-16-based self-assembled peptide nanofiber scaffolds loaded with TGF-β1 enhance the chondrogenic differentiation potential of BMSCs in vitro. Curr. Stem Cell Res. Ther. doi:10.2174/1574888x18666230316112847 (2023).
    • 124. Wang B, Wu Y, Shao Z et al. Functionalized self-assembling peptide nanofiber hydrogel as a scaffold for rabbit nucleus pulposus cells. J. Biomed. Mater. Res. A 100(3), 646–653 (2012).
    • 125. Chinese National Natural Science Foundation (NSF) report. https://kd.nsfc.gov.cn/finalDetails?id=1a08fc8e91fe0277c52c3ef5fa16a4ec
    • 126. Tao H, Zhang Y, Wang CF et al. Biological evaluation of human degenerated nucleus pulposus cells in functionalized self-assembling peptide nanofiber hydrogel scaffold. Tissue Eng. Part A 20(11–12), 1621–1631 (2014).
    • 127. Li X, Cheng S, Wu Y et al. Functional self-assembled peptide scaffold inhibits tumor necrosis factor-alpha-induced inflammation and apoptosis in nucleus pulposus cells by suppressing nuclear factor-κB signaling. J. Biomed. Mater. Res. Part A 106(4), 1082–1091 (2018).
    • 128. Li XC, Wu YH, Bai XD et al. BMP7-based functionalized self-assembling peptides protect nucleus pulposus-derived stem cells from apoptosis in vitro. Tissue Eng. Part A 22(19–20), 1218–1228 (2016).
    • 129. Wang YL, Yu SN, Shen HR et al. Thymosin β4 released from functionalized self-assembling peptide activates epicardium and enhances repair of infarcted myocardium. Theranostics 11(9), 4262–4280 (2021).
    • 130. Guo HD, Cui GH, Yang JJ et al. Sustained delivery of VEGF from designer self-assembling peptides improves cardiac function after myocardial infarction. Biochem. Biophys. Res. Comm. 424(1), 105–111 (2012).
    • 131. Guo HD, Cui GH, Wang HJ, Tan YZ. Transplantation of marrow-derived cardiac stem cells carried in designer self-assembling peptide nanofibers improves cardiac function after myocardial infarction. Biochem. Biophys. Res. Comm. 399(1), 42–48 (2010).
    • 132. Tokunaga M, Liu ML, Nagai T et al. Implantation of cardiac progenitor cells using self-assembling peptide improves cardiac function after myocardial infarction. J. Mol. Cell. Cardiol. 49(6), 972–983 (2010).
    • 133. Gong R, Jiang Z, Zagidullin N, Liu T, Cai B. Regulation of cardiomyocyte fate plasticity: a key strategy for cardiac regeneration. Signal Transduct. Target. Ther. 6(1), 31 (2021).
    • 134. Cavalcanti BN, Zeitlin BD, Nör JE. A hydrogel scaffold that maintains viability and supports differentiation of dental pulp stem cells. Dental Mater. 29(1), 97–102 (2013).
    • 135. Rosa V, Zhang Z, Grande RH, Nör JE. Dental pulp tissue engineering in full-length human root canals. J. Dental Res. 92(11), 970–975 (2013).
    • 136. Mu X, Shi L, Pan S, He L, Niu Y, Wang X. A customized self-assembling peptide hydrogel-wrapped stem cell factor targeting pulp regeneration rich in vascular-like structures. ACS Omega 5(27), 16568–16574 (2020).
    • 137. Takeuchi T, Bizenjima T, Ishii Y et al. Enhanced healing of surgical periodontal defects in rats following application of a self-assembling peptide nanofibre hydrogel. J. Clin. Periodontol. 43(3), 279–288 (2016).
    • 138. Jasty S, Suriyanarayanan S, Krishnakumar S. Influence of self-assembling peptide nanofibre scaffolds on retinal differentiation potential of stem/progenitor cells derived from ciliary pigment epithelial cells. J. Tissue Eng. Reg. Med. 11(2), 509–518 (2017).
    • 139. Akiyama N, Yamamoto-Fukuda T, Takahashi H, Koji T. In situ tissue engineering with synthetic self-assembling peptide nanofiber scaffolds, PuraMatrix, for mucosal regeneration in the rat middle-ear. Int. J. Nanomed. 8, 2629–2640 (2013).
    • 140. Wu H, Zhou T, Tian L, Xia Z, Xu F. Self-assembling RADA16-I peptide hydrogel scaffold loaded with tamoxifen for breast reconstruction. BioMed Res. Int. 2017, 3656193 (2017).
    • 141. Araki T, Mitsuyama K, Yamasaki H et al. Therapeutic potential of a self-assembling peptide hydrogel to treat colonic injuries associated with inflammatory bowel disease. J. Crohn's Colitis 15(9), 1517–1527 (2021).
    • 142. Wang T, Zhong X, Wang S, Lv F, Zhao X. Molecular mechanisms of RADA16-1 peptide on fast stop bleeding in rat models. Int. J. Mol. Sci. 13(11), 15279–15290 (2012).
    • 143. Taghavi L, Aramvash A, Seyedkarimi MS, Malek Sabet N. Evaluation of the hemocompatibility of RADA 16-I peptide. J. Biomater. Appl. 32(8), 1024–1031 (2018).
    • 144. Saini A, Serrano K, Koss K, Unsworth LD. Evaluation of the hemocompatibility and rapid hemostasis of (RADA)4 peptide-based hydrogels. Acta Biomaterialia 31, 71–79 (2016).
    • 145. Verbraeken B, Lammens M, Van Rompaey V et al. Efficacy and histopathological effects of self-assembling peptides RADA16 and IEIK13 in neurosurgical hemostasis. Nanomed. Nanotechnol. Biol. Med. 40, 102485 (2022).
    • 146. Stenson KM, Loftus IM, Chetter I et al. A multi-centre, single-arm clinical study to confirm safety and performance of PuraStat®, for the management of bleeding in elective carotid artery surgery. Clin. Appl. Thrombosis/Hemostasis 28, 10760296221144307 (2022).
    • 147. Yoshida M, Goto N, Kawaguchi M et al. Initial clinical trial of a novel hemostat, TDM-621, in the endoscopic treatments of the gastric tumors. J. Gastroenterol. Hepatol. 29(Suppl. 4), 77–79 (2014).
    • 148. Uraoka T, Ochiai Y, Fujimoto A et al. A novel fully synthetic and self-assembled peptide solution for endoscopic submucosal dissection-induced ulcer in the stomach. Gastroint. Endosc. 83(6), 1259–1264 (2016).
    • 149. Ortenzi M, Haji A. Safety and feasibility of PuraStat(®) in laparoscopic colorectal surgery (feasibility study). Min. Invasive Ther. Allied Technol. 30(6), 363–368 (2021).
    • 150. Wong E, Ho J, Smith M, Sritharan N, Riffat F, Smith MC. Use of Purastat, a novel haemostatic matrix based on self-assembling peptides in the prevention of nasopharyngeal adhesion formation. Int. J. Surg. Case Rep. 70, 227–229 (2020).
    • 151. Gangner Y, Bagot D'Arc M, Delin C. The use of self-assembling peptides (PuraStat) for hemostasis in cervical endocrine surgery. A real-life case series of 353 patients. Int. J. Surg. Case Rep. 94, 107072 (2022).
    • 152. Nagai Y, Unsworth LD, Koutsopoulos S, Zhang S. Slow release of molecules in self-assembling peptide nanofiber scaffold. J. Control. Rel. 115(1), 18–25 (2006).
    • 153. Roth AD, Lama P, Dunn S, Hong S, Lee MY. Polymer coating on a micropillar chip for robust attachment of PuraMatrix peptide hydrogel for 3D hepatic cell culture. Mater. Sci. Eng. C. Mater. Biol. Appl. 90, 634–644 (2018).
    • 154. Owczarz M, Casalini T, Motta AC, Morbidelli M, Arosio P. Contribution of electrostatics in the fibril stability of a model ionic-complementary peptide. Biomacromolecules 16(12), 3792–3801 (2015).
    • 155. Briuglia ML, Urquhart AJ, Lamprou DA. Sustained and controlled release of lipophilic drugs from a self-assembling amphiphilic peptide hydrogel. Int. J. Pharmaceut. 474(1–2), 103–111 (2014).