Skip to main content
Log in

Development of L-arginine-based poly(ester urethane)urea for enhanced vascular adaptability

  • Article
  • Published:
Science China Technological Sciences Aims and scope Submit manuscript

Abstract

Mismatched biomechanical properties between artificial vascular grafts and native blood vessels can result in intimal hyperplasia, especially for the implantation of small-diameter blood vessels. Ideal biomaterials for vascular repair still remain challenged. Biodegradable poly(ε-caprolactone) (PCL) has been applied for the preparation of electrospun vascular grafts, but more efforts are needed to improve its compliance with tissue growth. Herein, L-arginine-based poly(ester urethane)urea (PEUU) with both elasticity and biodegradability was synthesized so as to enhance the biomechanical properties of vascular grafts by blending electrospinning with PCL in a given mass ratio. It was exhibited that the prepared electrospun PCL/PEUU fibrous membranes were suitable for cell proliferation with normal cell morphology. More importantly, the electrospun membrane with 1/1 mass ratio of PCL/PEUU (PEUU50) showed specific flexibility, exhibiting more suitable mechanical properties matching to the native blood vessels. Specifically, the PEUU50 electrospun membrane demonstrated significantly lower Young’s modulus (9.3±0.8 MPa) and tensile strength (6.0±0.5 MPa), and extreme higher elongation (389%±24%) in wet state than those (16.3±3.0 MPa, 11.4±7.1 MPa and 196%±57%, respectively) of the pristine PCL membrane. Overall, this study demonstrated the great potential of amino acid-based PEUUs for the application in small-diameter vascular grafts.

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.

Similar content being viewed by others

References

  1. Lopera Higuita M, Griffiths L G. Small diameter xenogeneic extracellular matrix scaffolds for vascular applications. Tissue Eng Part B-Rev, 2020, 26: 26–45

    Article  Google Scholar 

  2. Gupta P, Mandal B B. Tissue-engineered vascular grafts: Emerging trends and technologies. Adv Funct Mater, 2021, 31: 2100027

    Article  Google Scholar 

  3. Wolny R, Mintz G S, Pręgowski J, et al. Mechanisms, prevention and treatment of saphenous vein graft disease. Am J Cardiol, 2021, 154: 41–47

    Article  Google Scholar 

  4. Leal B B J, Wakabayashi N, Oyama K, et al. Vascular tissue engineering: Polymers and methodologies for small caliber vascular grafts. Front Cardiovasc Med, 2020, 7: 592361

    Article  Google Scholar 

  5. Li Z, Wu Z, Lu T, et al. Materials and surface modification for tissue engineered vascular scaffolds. J Biomater Sci Polym Ed, 2016, 27: 1534–1552

    Article  Google Scholar 

  6. Hiob M A, She S, Muiznieks L D, et al. Biomaterials and modifications in the development of small-diameter vascular grafts. ACS Biomater Sci Eng, 2017, 3: 712–723

    Article  Google Scholar 

  7. Li J, Cai Z, Cheng J, et al. Characterization of a heparinized decellularized scaffold and its effects on mechanical and structural properties. J Biomater Sci Polym Ed, 2020, 31: 999–1023

    Article  Google Scholar 

  8. Bai S, Zhang X, Zang L, et al. Electrospinning of biomaterials for vascular regeneration. Chem Res Chin Univ, 2021, 37: 394–403

    Article  Google Scholar 

  9. Fan S, Zhang Y, Huang X, et al. Silk materials for medical, electronic and optical applications. Sci China Technol Sci, 2019, 62: 903–918

    Article  Google Scholar 

  10. Fang S, Ellman D G, Andersen D C. Review: Tissue engineering of small-diameter vascular grafts and their in vivo evaluation in large animals and humans. Cells, 2021, 10: 713

    Article  Google Scholar 

  11. Wen M, Zhi D, Wang L, et al. Local delivery of dual microRNAs in trilayered electrospun grafts for vascular regeneration. ACS Appl Mater Interfaces, 2020, 12: 6863–6875

    Article  Google Scholar 

  12. Wang Z, Zheng W, Wu Y, et al. Differences in the performance of PCL-based vascular grafts as abdominal aorta substitutes in healthy and diabetic rats. Biomater Sci, 2016, 4: 1485–1492

    Article  Google Scholar 

  13. Zhu M, Wang Z, Zhang J, et al. Circumferentially aligned fibers guided functional neoartery regeneration in vivo. Biomaterials, 2015, 61: 85–94

    Article  Google Scholar 

  14. Wu Y, Qin Y, Wang Z, et al. The regeneration of macro-porous electrospun poly(ε-caprolactone) vascular graft during long-term in situ implantation. J Biomed Mater Res B Appl Biomater, 2018, 106: 1618–1627

    Article  Google Scholar 

  15. Obiweluozor F O, Emechebe G A, Kim D W, et al. Considerations in the development of small-diameter vascular graft as an alternative for bypass and reconstructive surgeries: A review. Cardiovasc Eng Tech, 2020, 11: 495–521

    Article  Google Scholar 

  16. Zhu M, Wu Y, Li W, et al. Biodegradable and elastomeric vascular grafts enable vascular remodeling. Biomaterials, 2018, 183: 306–318

    Article  Google Scholar 

  17. Woodard L N, Grunlan M A. Hydrolytic degradation and erosion of polyester biomaterials. ACS Macro Lett, 2018, 7: 976–982

    Article  Google Scholar 

  18. van Uden S, Vanerio N, Catto V, et al. A novel hybrid silk-fibroin/polyurethane three-layered vascular graft: Towards in situ tissue-engineered vascular accesses for haemodialysis. Biomed Mater, 2019, 14: 025007

    Article  Google Scholar 

  19. Yu E, Mi H, Zhang J, et al. Development of biomimetic thermoplastic polyurethane/fibroin small-diameter vascular grafts via a novel electrospinning approach. J Biomed Mater Res B Appl Biomater, 2018, 106: 985–996

    Article  Google Scholar 

  20. Gostev A A, Chernonosova V S, Murashov I S, et al. Electrospun polyurethane-based vascular grafts: Physicochemical properties and functioning in vivo. Biomed Mater, 2020, 15: 015010

    Article  Google Scholar 

  21. He M, Chu C C. A new family of functional biodegradable arginine-based polyester urea urethanes: Synthesis, chracterization and biodegradation. Polymer, 2013, 54: 4112–4125

    Article  Google Scholar 

  22. Abel A K, Dreger N Z, Nettleton K, et al. Amino acid-based poly(ester urea)s as a matrix for extended release of entecavir. Biomacromolecules, 2020, 21: 946–954

    Article  Google Scholar 

  23. Policastro G M, Lin F, Smith Callahan L A, et al. OGP functionalized phenylalanine-based poly(ester urea) for enhancing osteoinductive potential of human mesenchymal stem cells. Biomacromolecules, 2015, 16: 1358–1371

    Article  Google Scholar 

  24. Zhu T, Gu H, Zhang H, et al. Covalent grafting of PEG and heparin improves biological performance of electrospun vascular grafts for carotid artery replacement. Acta Biomater, 2021, 119: 211–224

    Article  Google Scholar 

  25. Zhu T, Yu K, Bhutto M A, et al. Synthesis of RGD-peptide modified poly(ester-urethane) urea electrospun nanofibers as a potential application for vascular tissue engineering. Chem Eng J, 2017, 315: 177–190

    Article  Google Scholar 

  26. Yao Y, Ding J, Wang Z, et al. ROS-responsive polyurethane fibrous patches loaded with methylprednisolone (MP) for restoring structures and functions of infarcted myocardium in vivo. Biomaterials, 2020, 232: 119726

    Article  Google Scholar 

  27. Du J, Wang J, Yu H, et al. Electrospun poly(p-dioxanone)/poly(esterurethane)ureas composite nanofibers for potential heart valve tissue reconstruction. Chin J Polym Sci, 2019, 37: 560–569

    Article  Google Scholar 

  28. Zhu Q, Li X, Fan Z, et al. Biomimetic polyurethane/TiO2 nanocomposite scaffolds capable of promoting biomineralization and mesenchymal stem cell proliferation. Mater Sci Eng-C, 2018, 85: 79–87

    Article  Google Scholar 

  29. Song H, Chu C C. Synthesis and characterization of a new family of cationic amino acid-based poly(ester amide)s and their biological properties. J Appl Polym Sci, 2012, 124: 3840–3853

    Article  Google Scholar 

  30. You X, Gu Z, Huang J, et al. Arginine-based poly(ester amide) nanoparticle platform: From structure-property relationship to nucleic acid delivery. Acta Biomater, 2018, 74: 180–191

    Article  Google Scholar 

  31. Kabirian F, Ditkowski B, Zamanian A, et al. Controlled NO-release from 3D-printed small-diameter vascular grafts prevents platelet activation and bacterial infectivity. ACS Biomater Sci Eng, 2019, 5: 2284–2296

    Article  Google Scholar 

  32. He M, Potuck A, Kohn J C, et al. Self-assembled cationic biodegradable nanoparticles from pH-responsive amino-acid-based poly (ester urea urethane)s and their application as a drug delivery vehicle. Biomacromolecules, 2016, 17: 523–537

    Article  Google Scholar 

  33. He M, Sun L, Fu X, et al. Biodegradable amino acid-based poly(ester amine) with tunable immunomodulating properties and their in vitro and in vivo wound healing studies in diabetic rats’ wounds. Acta Biomater, 2019, 84: 114–132

    Article  Google Scholar 

  34. Yin M, Wan S, Ren X, et al. Development of inherently antibacterial, biodegradable, and biologically active chitosan/pseudo-protein hybrid hydrogels as biofunctional wound dressings. ACS Appl Mater Interfaces, 2021, 13: 14688–14699

    Article  Google Scholar 

  35. Li Y, Zhang X, Ying B. On textile biomedical engineering. Sci China Technol Sci, 2019, 62: 945–957

    Article  Google Scholar 

  36. Han J, Xiong L, Jiang X, et al. Bio-functional electrospun nanomaterials: From topology design to biological applications. Prog Polym Sci, 2019, 91: 1–28

    Article  Google Scholar 

  37. Cui C, Wen M, Zhou F, et al. Target regulation of both VECs and VSMCs by dual-loading miRNA-126 and miRNA-145 in the bilayered electrospun membrane for small-diameter vascular regeneration. J Biomed Mater Res B Appl BioMater, 2019, 107: 371–382

    Article  Google Scholar 

  38. Wen M, Zhou F, Cui C, et al. Performance of TMC-g-PEG-VAPG/miRNA-145 complexes in electrospun membranes for target-regulating vascular SMCs. Colloids Surfs B-Biointerfaces, 2019, 182: 110369

    Article  Google Scholar 

  39. Zhang Q, Liu B, Chong J, et al. Combination of hydrophobically modified γ-poly(glutamic acid) and trehalose achieving high cryosurvival of RBCs. Sci China Technol Sci, 2021, 64: 806–816

    Article  Google Scholar 

  40. Liu X, Gao S, Niu Q, et al. Facilitating trehalose entry into hRBCs at 4°C by alkylated ε-poly(L-lysine) for glycerol-free cryopreservation. J Mater Chem B, 2022, 10: 1042–1054

    Article  Google Scholar 

  41. Braiman M S, Briercheck D M, Kriger K M. Modeling vibrational spectra of amino acid side chains in proteins: Effects of protonation state, counterion, and solvent on arginine C-N stretch frequencies. J Phys Chem B, 1999, 103: 4744–4750

    Article  Google Scholar 

  42. Rickel A P, Deng X, Engebretson D, et al. Electrospun nanofiber scaffold for vascular tissue engineering. Mater Sci Eng-C, 2021, 129: 112373

    Article  Google Scholar 

  43. Xie F, Zhang T, Bryant P, et al. Degradation and stabilization of polyurethane elastomers. Prog Polym Sci, 2019, 90: 211–268

    Article  Google Scholar 

  44. Li T, Wang Y, Li S, et al. Mechanically robust, elastic, and healable ionogels for highly sensitive ultra-durable ionic skins. Adv Mater, 2020, 32: 2002706

    Article  Google Scholar 

  45. Jeong Y J, Yao Y, Yim E K F. Current understanding of intimal hyperplasia and effect of compliance in synthetic small diameter vascular grafts. Biomater Sci, 2020, 8: 4383–4395

    Article  Google Scholar 

  46. Stekelenburg M, Rutten M C M, Snoeckx L H E H, et al. Dynamic straining combined with fibrin gel cell seeding improves strength of tissue-engineered small-diameter vascular grafts. Tissue Eng Part A, 2009, 15: 1081–1089

    Article  Google Scholar 

  47. Zheng Y, Ashizawa M, Zhang S, et al. Tuning the mechanical properties of a polymer semiconductor by modulating hydrogen bonding interactions. Chem Mater, 2020, 32: 5700–5714

    Article  Google Scholar 

  48. Jirofti N, Mohebbi-Kalhori D, Samimi A, et al. Small-diameter vascular graft using co-electrospun composite PCL/PU nanofibers. Biomed Mater, 2018, 13: 055014

    Article  Google Scholar 

  49. Oliveira S, Felizardo T, Amorim S, et al. Tubular fibrous scaffolds functionalized with tropoelastin as a small-diameter vascular graft. Biomacromolecules, 2020, 21: 3582–3595

    Article  Google Scholar 

  50. Konig G, McAllister T N, Dusserre N, et al. Mechanical properties of completely autologous human tissue engineered blood vessels compared to human saphenous vein and mammary artery. Biomaterials, 2009, 30: 1542–1550

    Article  Google Scholar 

  51. Jia W, Li M, Weng H, et al. Design and comprehensive assessment of a biomimetic tri-layer tubular scaffold via biodegradable polymers for vascular tissue engineering applications. Mater Sci Eng-C, 2020, 110: 110717

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to XiaoYan Yuan.

Additional information

This work was supported by the National Natural Science Foundation of China (Grant No. 52073204).

Supporting Information

The supporting information is available online at tech.scichina.com and link.springer.com. The supporting materials are published as submitted, without typesetting or editing. The responsibility for scientific accuracy and content remains entirely with the authors.

Supplementary Material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bai, S., Zhang, X., Zang, L. et al. Development of L-arginine-based poly(ester urethane)urea for enhanced vascular adaptability. Sci. China Technol. Sci. 65, 2751–2762 (2022). https://doi.org/10.1007/s11431-022-2038-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11431-022-2038-9

Keywords

Navigation