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

Nanotechnology-based bone regeneration in orthopedics: a review of recent trends

    Wenqing Liang‡

    **Author for correspondence: Tel.: +86 580 261 5027;

    E-mail Address: liangwq@usx.edu.cn

    Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Chao Zhou‡

    Department of Orthopedics, Zhoushan Guanghua hospital, Zhoushan, 316000, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Juqin Bai

    Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China

    ,
    Hongwei Zhang

    Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China

    ,
    Hengguo Long

    Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China

    ,
    Bo Jiang

    Rehabilitation Department, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China

    ,
    Lu Liu

    Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China

    ,
    Linying Xia

    Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China

    ,
    Chanyi Jiang

    Department of Pharmacy, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China

    ,
    Hengjian Zhang

    Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China

    &
    Jiayi Zhao

    *Author for correspondence: Tel.: +86 580 261 5024;

    E-mail Address: zjy2038689@sina.com

    Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China

    Published Online:https://doi.org/10.2217/nnm-2023-0187

    Nanotechnology has revolutionized the field of bone regeneration, offering innovative solutions to address the challenges associated with conventional therapies. This comprehensive review explores the diverse landscape of nanomaterials – including nanoparticles, nanocomposites and nanofibers – tailored for bone tissue engineering. We delve into the intricate design principles, structural mimicry of native bone and the crucial role of biomaterial selection, encompassing bioceramics, polymers, metals and their hybrids. Furthermore, we analyze the interface between cells and nanostructured materials and their pivotal role in engineering and regenerating bone tissue. In the concluding outlook, we highlight emerging frontiers and potential research directions in harnessing nanomaterials for bone regeneration.

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

    References

    • 1. Annamalai RT, Hong X, Schott NG, Tiruchinapally G, Levi B, Stegemann JP. Injectable osteogenic microtissues containing mesenchymal stromal cells conformally fill and repair critical size defects. Biomaterials 208, 32–44 (2019). • Explores the development of injectable microtissues containing mesenchymal stromal cells, focusing on their ability to repair critical-size bone defects efficiently.
    • 2. Yang D, Chen Z, Xu Z, Qin L, Yi W, Long Y. Roles of stem cell exosomes and their microRNA carrier in bone and cartilage regeneration. Curr. Stem Cell Res. Ther. 18(7), 917–925 (2023). •• Provides insights into the mechanisms by which stem cell exosomes contribute to tissue repair, offering valuable information for researchers and clinicians interested in regenerative medicine.
    • 3. Mcdermott AM, Herberg S, Mason DE et al. Recapitulating bone development through engineered mesenchymal condensations and mechanical cues for tissue regeneration. Sci. Transl. Med. 11(495), eaav7756 (2019). • Presents a novel approach to recapitulate bone development by engineering mesenchymal condensations and applying mechanical cues, and explores how these engineered processes can be leveraged for tissue regeneration, particularly in the context of bone.
    • 4. Lee J-W, Han H-S, Han K-J et al. Long-term clinical study and multiscale analysis of in vivo biodegradation mechanism of Mg alloy. Proc. Natl Acad. Sci. USA 113(3), 716–721 (2016). • Long-term clinical study and multiscale analysis of the in vivo biodegradation mechanism of magnesium alloy, contributing to our understanding of magnesium alloy implants’ long-term performance and safety.
    • 5. Bandyopadhyay A, Mitra I, Goodman SB, Kumar M, Bose S. Improving biocompatibility for next generation of metallic implants. Prog. Mater. Sci. 133, 101053 (2022). •• Provides insights into innovative materials and surface modification techniques aimed at reducing adverse reactions and promoting better tissue integration, thus advancing the field of implantology.
    • 6. Marrale J, Morrissey MC, Haddad FS. A literature review of autograft and allograft anterior cruciate ligament reconstruction. Knee Surg. Sports Traumatol. Arthrosc. 15, 690–704 (2007). • Comprehensive overview of autograft and allograft options for anterior cruciate ligament reconstruction. Discusses the advantages and disadvantages of each approach, helping clinicians make informed decisions regarding graft selection.
    • 7. Xu Z, Gao W, Bai H. Silk-based bioinspired structural and functional materials. iScience 25(3), 103940 (2022). •• Explores the development of bioinspired materials based on silk; discusses the structural and functional properties of silk and how these properties can inspire the design of innovative biomaterials for various applications, including tissue engineering and regenerative medicine.
    • 8. Gong T, Xie J, Liao J, Zhang T, Lin S, Lin Y. Nanomaterials and bone regeneration. Bone Res. 3(1), 1–7 (2015). • Provides insights into the role of nanomaterials in bone regeneration; discusses various nanomaterials, their properties and how they can enhance bone tissue engineering and regeneration.
    • 9. Kunrath MF, Shah FA, Dahlin C. Bench-to-bedside: feasibility of nano-engineered and drug-delivery biomaterials for bone-anchored implants and periodontal applications. Mater. Today Bio 18, 100540 (2022).
    • 10. Gresham RC, Bahney CS, Leach JK. Growth factor delivery using extracellular matrix-mimicking substrates for musculoskeletal tissue engineering and repair. Bioact. Mater. 6(7), 1945–1956 (2021).
    • 11. Al-Qudsy L, Hu Y-W, Xu H, Yang P-F. Mineralized collagen fibrils: an essential component in determining the mechanical behavior of cortical bone. ACS Biomater. Sci. Eng. 9(5), 2203–2219 (2023).
    • 12. Liu Y, Luo D, Kou XX et al. Hierarchical intrafibrillar nanocarbonated apatite assembly improves the nanomechanics and cytocompatibility of mineralized collagen. Adv. Funct. Mater. 23(11), 1404–1411 (2013).
    • 13. Liu Y, Luo D, Wang T. Hierarchical structures of bone and bioinspired bone tissue engineering. Small 12(34), 4611–4632 (2016).
    • 14. Feng C, Zhang W, Deng C et al. 3D printing of lotus root-like biomimetic materials for cell delivery and tissue regeneration. Adv. Sci. 4(12), 1700401 (2017).
    • 15. Zhou T, Yan L, Xie C et al. A mussel-inspired persistent ROS-scavenging, electroactive, and osteoinductive scaffold based on electrochemical-driven in situ nanoassembly. Small 15(25), 1805440 (2019).
    • 16. Abdelaziz AG, Nageh H, Abdo SM et al. A review of 3D polymeric scaffolds for bone tissue engineering: principles, fabrication techniques, immunomodulatory roles, and challenges. Bioengineering 10(2), 204 (2023).
    • 17. Oo JA, Brandes RP, Leisegang MS. Long non-coding RNAs: novel regulators of cellular physiology and function. Pflugers Arch. 474(2), 191–204 (2021).
    • 18. Han X, Li B, Zhang S. MIR503HG: a potential diagnostic and therapeutic target in human diseases. Biomed. Pharmacother. 160, 114314 (2023).
    • 19. Guo C-J, Ma X-K, Xing Y-H et al. Distinct processing of lncRNAs contributes to non-conserved functions in stem cells. Cell 181(3), 621–636 (2020).
    • 20. Statello L, Guo C-J, Chen L-L, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev. Mol. Cell Biol. 22(2), 96–118 (2021).
    • 21. Liu H, Xu Y, Yao B, Sui T, Lai L, Li Z. A novel N6-methyladenosine (m6A)-dependent fate decision for the lncRNA THOR. Cell Death Dis. 11(8), 613 (2020).
    • 22. Zhang D, Xue J, Peng F. The regulatory activities of MALAT1 in the development of bone and cartilage diseases. Front. Endocrinol. 13, 1054827 (2022).
    • 23. Yetisgin AA, Cetinel S, Zuvin M, Kosar A, Kutlu O. Therapeutic nanoparticles and their targeted delivery applications. Mole 25(9), 2193 (2020).
    • 24. Naskar S, Kuotsu K, Sharma S. Chitosan-based nanoparticles as drug delivery systems: a review on two decades of research. J. Drug Target. 27(4), 379–393 (2019).
    • 25. Qu H, Fu H, Han Z, Sun Y. Biomaterials for bone tissue engineering scaffolds: a review. RSC Adv. 9(45), 26252–26262 (2019).
    • 26. Gautam G, Kumar S, Kumar K. Processing of biomaterials for bone tissue engineering: state of the art. Mater. Today Proc. 50, 2206–2217 (2022).
    • 27. You Q, Lu M, Li Z, Zhou Y, Tu C. Cell sheet technology as an engineering-based approach to bone regeneration. Int. J. Nanomed. 17, 6491–6511 (2022).
    • 28. Zheng K, Bai J, Yang H et al. Nanomaterial-assisted theranosis of bone diseases. Bioact. Mater. 24, 263–312 (2023).
    • 29. Mcmahon RE, Wang L, Skoracki R, Mathur AB. Development of nanomaterials for bone repair and regeneration. J. Biomed. Mater. Res. B Appl. Biomater. 101(2), 387–397 (2013).
    • 30. Qiao K, Xu L, Tang J et al. The advances in nanomedicine for bone and cartilage repair. J. Nanobiotechnol. 20(1), 141 (2022).
    • 31. Kim YS, Smoak MM, Melchiorri AJ, Mikos AG. An overview of the tissue engineering market in the United States from 2011 to 2018. Tissue Eng. A 25(1–2), 1–8 (2019).
    • 32. Jin S-S, He D-Q, Luo D et al. A biomimetic hierarchical nanointerface orchestrates macrophage polarization and mesenchymal stem cell recruitment to promote endogenous bone regeneration. ACS Nano 13(6), 6581–6595 (2019).
    • 33. Stuckensen K, Schwab A, Knauer M et al. Tissue mimicry in morphology and composition promotes hierarchical matrix remodeling of invading stem cells in osteochondral and meniscus scaffolds. Adv. Mater. 30(28), 1706754 (2018).
    • 34. Laubach M, Suresh S, Herath B et al. Clinical translation of a patient-specific scaffold-guided bone regeneration concept in four cases with large long bone defects. J. Orthop. Translat. 34, 73–84 (2022).
    • 35. Mirkhalaf M, Men Y, Wang R, No Y, Zreiqat H. Personalized 3D printed bone scaffolds: a review. Acta Biomater. 156, 110–124 (2023).
    • 36. Laubach M, Kobbe P, Hutmacher DW. Biodegradable interbody cages for lumbar spine fusion: current concepts and future directions. Biomaterials 288, 121699 (2022).
    • 37. Tavares WM, De França SA, Paiva WS, Teixeira MJ. A systematic review and meta-analysis of fusion rate enhancements and bone graft options for spine surgery. Sci. Rep. 12(1), 7546 (2022).
    • 38. Shen M, Wang L, Gao Y et al. 3D bioprinting of in situ vascularized tissue-engineered bone for repairing large segmental bone defects. Mater. Today Bio 16, 100382 (2022).
    • 39. Brazill JM, Beeve AT, Craft CS, Ivanusic JJ, Scheller EL. Nerves in bone: evolving concepts in pain and anabolism. J. Bone Miner. Res. 34(8), 1393–1406 (2019).
    • 40. Nadine S, Fernandes IJ, Correia CR, Mano JF. Close-to-native bone repair via tissue engineered endochondral ossification approaches. iScience 25(11), 105370 (2022).
    • 41. Bhumiratana S, Bernhard JC, Alfi DM et al. Tissue-engineered autologous grafts for facial bone reconstruction. Sci. Transl. Med. 8(343), 343ra383–343ra383 (2016).
    • 42. Conith AJ, Lam DT, Albertson RC. Muscle-induced loading as an important source of variation in craniofacial skeletal shape. Genesis 57(1), e23263 (2019).
    • 43. Charbe NB, Tambuwala M, Palakurthi SS et al. Biomedical applications of three-dimensional bioprinted craniofacial tissue engineering. Bioeng. Transl. Med. 8(1), e10333 (2023).
    • 44. Xu X, Liao L, Tian W. Strategies of prevascularization in tissue engineering and regeneration of craniofacial tissues. Tissue Eng. B Rev. 28(2), 464–475 (2022).
    • 45. Karanth D, Song K, Martin ML et al. Towards resorbable 3D-printed scaffolds for craniofacial bone regeneration. Orthod. Craniofac. Res. doi: 10.1111/ocr.12645 (2023) (Epub ahead of print).
    • 46. Liu M, Nakasaki M, Shih Y-RV, Varghese S. Effect of age on biomaterial-mediated in situ bone tissue regeneration. Acta Biomater. 78, 329–340 (2018).
    • 47. Crago M, Winlaw DS, Farajikhah S, Dehghani F, Naficy S. Pediatric pulmonary valve replacements: clinical challenges and emerging technologies. Bioeng. Transl. Med. 8(4), e10501 (2023).
    • 48. Gabel L, Macdonald HM, Mckay HA. Sex differences and growth-related adaptations in bone microarchitecture, geometry, density, and strength from childhood to early adulthood: a mixed longitudinal HR-pQCT study. J. Bone Miner. Res. 32(2), 250–263 (2017).
    • 49. Gu Y, Forget A, Shastri VP. Biobridge: an outlook on translational bioinks for 3D bioprinting. Adv. Sci. 9(3), 2103469 (2022).
    • 50. Thrivikraman G, Athirasala A, Gordon R et al. Rapid fabrication of vascularized and innervated cell-laden bone models with biomimetic intrafibrillar collagen mineralization. Nat. Commun. 10(1), 3520 (2019).
    • 51. Martine LC, Holzapfel BM, Mcgovern JA et al. Engineering a humanized bone organ model in mice to study bone metastases. Nat. Protoc. 12(4), 639–663 (2017).
    • 52. Young SA, Heller A-D, Garske DS et al. From breast cancer cell homing to the onset of early bone metastasis: dynamic bone (re) modeling as a driver of metastasis. bioRxiv 2023.2001.2024.525352 https://doi.org/10.1101/2023.01.24.525352 (2023).
    • 53. Ratner BD. Biomaterials science: an interdisciplinary endeavor. In: Biomaterials Science: an Introduction to Materials in Medicine. Elsevier, USA, 1–8 (1996).
    • 54. Chen L, Yu C, Xu W et al. Dual-targeted nanodiscs revealing the cross-talk between osteogenic differentiation of mesenchymal stem cells and macrophages. ACS Nano 17(3), 3153–3167 (2023).
    • 55. Guo J, Kim Y, Xie V et al. Modular, tissue-specific, and biodegradable hydrogel cross-linkers for tissue engineering. Sci. Adv. 5(6), eaaw7396 (2019).
    • 56. Deng C, Lin R, Zhang M et al. Micro/nanometer-structured scaffolds for regeneration of both cartilage and subchondral bone. Adv. Funct. Mater. 29(4), 1806068 (2019).
    • 57. Hu X, Lin Z, He J et al. Recent progress in 3D printing degradable polylactic acid-based bone repair scaffold for the application of cancellous bone defect. MedComm Biomater. Appl. 1(1), e14 (2022).
    • 58. Bohner M, Miron RJ. A proposed mechanism for material-induced heterotopic ossification. Mater. Today 22, 132–141 (2019).
    • 59. Bernardo MP, Da Silva BC, Hamouda AE et al. PLA/hydroxyapatite scaffolds exhibit in vitro immunological inertness and promote robust osteogenic differentiation of human mesenchymal stem cells without osteogenic stimuli. Sci. Rep. 12(1), 2333 (2022).
    • 60. Wilkesmann S, Westhauser F. Tailoring the osteogenic properties of bioactive glasses by incorporation of therapeutic ions for orthopedic applications. In: Bioactive Glasses and Glass-Ceramics: Fundamentals and Applications. Wiley, Hoboken, NJ, USA, 203–226 (2022).
    • 61. Zarrintaj P, Seidi F, Azarfam MY et al. Biopolymer-based composites for tissue engineering applications: a basis for future opportunities. Compos. B Eng. 258, 110701 (2023).
    • 62. Lee SS, Du X, Smit T et al. 3D-printed LEGO®-inspired titanium scaffolds for patient-specific regenerative medicine. bioRxiv 2023.2003.2030.534953 https://doi.org/10.1101/2023.03.30.534953 (2023).
    • 63. Chen W, Zhang H, Zhou Q, Zhou F, Zhang Q, Su J. Smart hydrogels for bone reconstruction via modulating the microenvironment. Research (Wash. DC) 6, 89 (2023).
    • 64. Wei DX, Dao JW, Chen GQ. A micro-ark for cells: highly open porous polyhydroxyalkanoate microspheres as injectable scaffolds for tissue regeneration. Adv. Mater. 30(31), 1802273 (2018).
    • 65. Lin Z, Wu J, Qiao W et al. Precisely controlled delivery of magnesium ions thru sponge-like monodisperse PLGA/nano-MgO-alginate core-shell microsphere device to enable in situ bone regeneration. Biomaterials 174, 1–16 (2018).
    • 66. Tan J, Zhang M, Hai Z et al. Sustained release of two bioactive factors from supramolecular hydrogel promotes periodontal bone regeneration. ACS Nano 13(5), 5616–5622 (2019).
    • 67. Hao L, Wang A, Fu J et al. Biomineralized dipeptide self-assembled hydrogel with ultrahigh mechanical strength and osteoinductivity for bone regeneration. Colloids Surf. A Physicochem. Eng. Asp. 657, 130622 (2023).
    • 68. Lenne P-F, Trivedi V. Sculpting tissues by phase transitions. Nat. Commun. 13(1), 664 (2022).
    • 69. Sri H, Ganapathy D, Venugopalan S, Deshmukh M. Current trends in bone grafts – a review. J. Pharm. Negat. Results 13(7), 474–481 (2022).
    • 70. Chakraborty U, Bhanjana G, Kaur N et al. Design and testing of nanobiomaterials for orthopedic implants. In: Engineered Nanostructures for Therapeutics and Biomedical Applications. Kaushik AKKumar SChaudhary GR (Eds). Elsevier, Amsterdam, 227–271 (2023).
    • 71. Zhao Y, Bai L, Zhang Y et al. Type I collagen decorated nanoporous network on titanium implant surface promotes osseointegration through mediating immunomodulation, angiogenesis, and osteogenesis. Biomaterials 288, 121684 (2022).
    • 72. Luo J, Walker M, Xiao Y, Donnelly H, Dalby MJ, Salmeron-Sanchez M. The influence of nanotopography on cell behaviour through interactions with the extracellular matrix – a review. Bioact. Mater. 15, 145–159 (2022).
    • 73. Gu X, Wang F, Li X. Overview of scaffold reinforcement for tissue repair. In: Tissue Repair: Reinforced Scaffolds. Li X (Ed.). Springer, NY, USA, 1–23 (2017).
    • 74. Elangomannan S, Louis K, Dharmaraj BM, Kandasamy VS, Soundarapandian K, Gopi D. Carbon nanofiber/polycaprolactone/mineralized hydroxyapatite nanofibrous scaffolds for potential orthopedic applications. ACS Appl. Mater. Interfaces 9(7), 6342–6355 (2017).
    • 75. Misra RDK, Nune C, Pesacreta TC, Somani MC, Karjalainen LP. Understanding the impact of grain structure in austenitic stainless steel from a nanograined regime to a coarse-grained regime on osteoblast functions using a novel metal deformation–annealing sequence. Acta Biomater. 9(4), 6245–6258 (2013).
    • 76. Webster TJ, Waid MC, McKenzie JL, Price RL, Ejiofor JU. Nano-biotechnology: carbon nanofibres as improved neural and orthopaedic implants. Nanotechnology 15(1), 9 (2004).
    • 77. Mohandas G, Oskolkov N, McMahon M, Walczak P, Janowski M. Porous tantalum and tantalum oxide nanoparticles for regenerative medicine. Acta Neurobiologiae Experimentalis 74(2), 188–196 (2014).
    • 78. Morelli S, Salerno S, Holopainen J, Ritala M, De Bartolo L. Osteogenic and osteoclastogenic differentiation of co-cultured cells in polylactic acid–nanohydroxyapatite fiber scaffolds. J. Biotechnol. 204, 53–62 (2015).
    • 79. Webster TJ, Ergun C, Doremus RH, Siegel RW, Bizios R. Enhanced functions of osteoblasts on nanophase ceramics. Biomaterials 21(17), 1803–1810 (2000).
    • 80. Sayyar S, Murray E, Thompson BC, Gambhir S, Officer DL, Wallace GG. Covalently linked biocompatible graphene/polycaprolactone composites for tissue engineering. Carbon 52, 296–304 (2013).
    • 81. Ogihara N, Usui Y, Aoki K et al. Biocompatibility and bone tissue compatibility of alumina ceramics reinforced with carbon nanotubes. Nanomedicine 7(7), 981–993 (2012).
    • 82. Kong H, Jang J. Antibacterial properties of novel poly (methyl methacrylate) nanofiber containing silver nanoparticles. Langmuir 24(5), 2051–2056 (2008).
    • 83. Buzarovska A, Gualandi C, Parrilli A, Scandola M. Effect of TiO2 nanoparticle loading on poly(L-lactic acid) porous scaffolds fabricated by TIPS. Composites B Eng. 81, 189–195 (2015).
    • 84. Jayakumar R, Menon D, Manzoor K, Nair SV, Tamura H. Biomedical applications of chitin and chitosan based nanomaterials – a short review. Carbohydr. Polym. 82(2), 227–232 (2010).
    • 85. Shih YRV, Chen CN, Tsai SW, Wang YJ, Lee OK. Growth of mesenchymal stem cells on electrospun type I collagen nanofibers. Stem Cells 24(11), 2391–2397 (2006).
    • 86. Yamanaka K, Mori M, Chiba A. Nanoarchitectured Co–Cr–Mo orthopedic implant alloys: nitrogen-enhanced nanostructural evolution and its effect on phase stability. Acta Biomater. 9(4), 6259–6267 (2013).
    • 87. Rahaman MN, Day DE, Bal BS et al. Bioactive glass in tissue engineering. Acta Biomater. 7(6), 2355–2373 (2011).
    • 88. Kharaziha M, Fathi M. Synthesis and characterization of bioactive forsterite nanopowder. Ceram. Int. 35(6), 2449–2454 (2009).
    • 89. Cai Y, Tang R. Calcium phosphate nanoparticles in biomineralization and biomaterials. J. Mater. Chem. 18(32), 3775–3787 (2008).
    • 90. Jeffrey DS. One-and three-dimensional growth of hydroxyapatite nanowires during sol–gel–hydrothermal synthesis. (2012).
    • 91. Chen H, Clarkson BH, Sun K, Mansfield JF. Self-assembly of synthetic hydroxyapatite nanorods into an enamel prism-like structure. J. Colloid Interface Sci. 288(1), 97–103 (2005).
    • 92. Zhou H, Lee J. Nanoscale hydroxyapatite particles for bone tissue engineering. Acta Biomater. 7(7), 2769–2781 (2011).
    • 93. Webster TJ, Ejiofor JU. Increased osteoblast adhesion on nanophase metals: Ti, Ti6Al4V, and CoCrMo. Biomaterials 25(19), 4731–4739 (2004).
    • 94. Abazari MF, Torabinejad S, Karizi SZ et al. Promoted osteogenic differentiation of human induced pluripotent stem cells using composited polycaprolactone/polyvinyl alcohol/carbopol nanofibrous scaffold. J. Drug Deliv. Sci. Technol. 71, 103318 (2022).
    • 95. Giaconia MA, Dos Passos Ramos S, Araújo TA, De Almeida Cruz M, Renno AC, Braga ARC. Scaffold production and bone tissue healing using electrospinning: trends and gap of knowledge. Regen. Eng. Transl. Med. 8(4), 506–522 (2022).
    • 96. Yao Q, Cosme JG, Xu T et al. Three dimensional electrospun PCL/PLA blend nanofibrous scaffolds with significantly improved stem cells osteogenic differentiation and cranial bone formation. Biomaterials 115, 115–127 (2017).
    • 97. Dhand C, Ong ST, Dwivedi N et al. Bio-inspired in situ cross-linking and mineralization of electrospun collagen scaffolds for bone tissue engineering. Biomaterials 104, 323–338 (2016).
    • 98. Cheng X, Liu Y-T, Si Y, Yu J, Ding B. Direct synthesis of highly stretchable ceramic nanofibrous aerogels via 3D reaction electrospinning. Nat. Commun. 13(1), 2637 (2022).
    • 99. Chen Z, Wang L, Chen C et al. NSC-derived extracellular matrix-modified GelMA hydrogel fibrous scaffolds for spinal cord injury repair. NPG Asia Mater. 14(1), 20 (2022).
    • 100. Shafiei SS, Shavandi M, Ahangari G, Shokrolahi F. Electrospun layered double hydroxide/poly (ϵ-caprolactone) nanocomposite scaffolds for adipogenic differentiation of adipose-derived mesenchymal stem cells. Appl. Clay Sci. 127, 52–63 (2016).
    • 101. Belgheisi G, Nazarpak MH, Hashjin MS. Bone tissue engineering electrospun scaffolds based on layered double hydroxides with the ability to release vitamin D3: fabrication, characterization and in vitro study. Appl. Clay Sci. 185, 105434 (2020).
    • 102. Wu S, Wang J, Jin L, Li Y, Wang Z. Effects of polyacrylonitrile/MoS2 composite nanofibers on the growth behavior of bone marrow mesenchymal stem cells. ACS Appl. Nano Mater. 1(1), 337–343 (2017).
    • 103. Ma K, Liao C, Huang L et al. Electrospun PCL/MoS2 nanofiber membranes combined with NIR-triggered photothermal therapy to accelerate bone regeneration. Small 17(51), 2104747 (2021).
    • 104. Di Pompo G, Liguori A, Carlini M et al. Electrospun fibers coated with nanostructured biomimetic hydroxyapatite: a new platform for regeneration at the bone interfaces. Biomater. Adv. 144, 213231 (2023).
    • 105. Okamoto M, John B. Synthetic biopolymer nanocomposites for tissue engineering scaffolds. Prog. Polym. Sci. 38(10–11), 1487–1503 (2013).
    • 106. Ye G, Bao F, Zhang X et al. Nanomaterial-based scaffolds for bone tissue engineering and regeneration. Nanomedicine 15(20), 1995–2017 (2020).
    • 107. Asghar MS, Li J, Ahmed I et al. Antioxidant, and enhanced flexible nano porous scaffolds for bone tissue engineering applications. Nano Select 2(7), 1356–1367 (2021).
    • 108. Wang M, Yao J, Shen S et al. A scaffold with zinc–whitlockite nanoparticles accelerates bone reconstruction by promoting bone differentiation and angiogenesis. Nano Res. 16(1), 757–770 (2023).
    • 109. Chen J, Ashames A, Buabeid MA, Fahelelbom KM, Ijaz M, Murtaza G. Nanocomposites drug delivery systems for the healing of bone fractures. Int. J. Pharm. 585, 119477 (2020).
    • 110. Gong T, Xie J, Liao J, Zhang T, Lin S, Lin Y. Nanomaterials and bone regeneration. Bone Res. 3, 15029 (2015).
    • 111. Han L, Wang M, Sun H et al. Porous titanium scaffolds with self-assembled micro/nano-hierarchical structure for dual functions of bone regeneration and anti-infection. J. Biomed. Mater. Res. A 105(12), 3482–3492 (2017).
    • 112. Gogoi S, Kumar M, Mandal BB, Karak N. A renewable resource based carbon dot decorated hydroxyapatite nanohybrid and its fabrication with waterborne hyperbranched polyurethane for bone tissue engineering. RSC Adv. 6(31), 26066–26076 (2016).
    • 113. Lu Y, Li L, Li M et al. Zero-dimensional carbon dots enhance bone regeneration, osteosarcoma ablation, and clinical bacterial eradication. Bioconjug. Chem. 29(9), 2982–2993 (2018).
    • 114. Ghorghi M, Rafienia M, Nasirian V, Bitaraf FS, Gharravi AM, Zarrabi A. Electrospun captopril-loaded PCL–carbon quantum dots nanocomposite scaffold: fabrication, characterization, and in vitro studies. Polym. Adv. Technol. 31(12), 3302–3315 (2020).
    • 115. Erdal NB, Hakkarainen M. Construction of bioactive and reinforced bioresorbable nanocomposites by reduced nano-graphene oxide carbon dots. Biomacromolecules 19(3), 1074–1081 (2018).
    • 116. Kim J, Kim HN, Lim K-T et al. Designing nanotopographical density of extracellular matrix for controlled morphology and function of human mesenchymal stem cells. Sci. Rep. 3(1), 3552 (2013).
    • 117. Kim J, Bae W-G, Choung H-W et al. Multiscale patterned transplantable stem cell patches for bone tissue regeneration. Biomaterials 35(33), 9058–9067 (2014).
    • 118. Cheng Y, Pang SW. Effects of nanopillars and surface coating on dynamic traction force. Microsystems Nanoengineer. 9, 6 (2023).
    • 119. Soriente A, Fasolino I, Gomez-Sánchez A et al. Chitosan/hydroxyapatite nanocomposite scaffolds to modulate osteogenic and inflammatory response. J. Biomed. Mater Res. A 110(2), 266–272 (2022).
    • 120. Xu Z, Qi X, Bao M et al. Biomineralization-inspired 3D printed bioactive glass nanocomposite scaffolds orchestrate diabetic bone regeneration by remodeling micromilieu. Bioact. Mater. 25, 239–255 (2023).
    • 121. Eivazzadeh-Keihan R, Maleki A, De La Guardia M et al. Carbon based nanomaterials for tissue engineering of bone: building new bone on small black scaffolds: a review. J. Adv. Res. 18, 185–201 (2019).
    • 122. Gwon Y, Park S, Kim W, Han T, Kim H, Kim J. Radially patterned transplantable biodegradable scaffolds as topographically defined contact guidance platforms for accelerating bone regeneration. J. Biol. Eng. 15(1), 12 (2021).
    • 123. Chen X, Xu Y, Cheng Y, Pang S. Engineered barriers regulate osteoblast cell migration in vertical direction. Sci. Rep. 12(1), 4459 (2022).
    • 124. Walmsley GG, Mcardle A, Tevlin R et al. Nanotechnology in bone tissue engineering. Nanomedicine 11(5), 1253–1263 (2015).
    • 125. Xue J, Wu T, Dai Y, Xia Y. Electrospinning and electrospun nanofibers: methods, materials, and applications. Chem. Rev. 119(8), 5298–5415 (2019).
    • 126. Kumar S, Nehra M, Kedia D, Dilbaghi N, Tankeshwar K, Kim K-H. Nanotechnology-based biomaterials for orthopaedic applications: recent advances and future prospects. Mater. Sci. Eng. C 106, 110154 (2020).
    • 127. Xuan L, Ju Z, Skonieczna M, Zhou PK, Huang R. Nanoparticles‐induced potential toxicity on human health: applications, toxicity mechanisms, and evaluation models. MedComm. 4(4), e327 (2023).