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

Engineered cells along with smart scaffolds: critical factors for improving tissue engineering approaches

    Zahra Abpeikar

    Department of Tissue Engineering & Applied Cell Sciences, School of Advance Medical Science & Technology, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran

    ,
    Ali Akbar Alizadeh

    *Author for correspondence: Tel.: +98 713 230 5471;

    E-mail Address: alizadehaa@sums.ac.ir

    Department of Tissue Engineering & Applied Cell Sciences, School of Advance Medical Science & Technology, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran

    ,
    Yaghoub Ahmadyousefi

    Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 6517838687, Iran

    ,
    Ali Akbar Najafi

    Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, 7919693116, Iran

    &
    Mohsen Safaei

    **Author for correspondence:

    E-mail Address: st-safaei.m@skums.ac.ir

    Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, 8815713471, Iran

    Published Online:https://doi.org/10.2217/rme-2022-0059

    In this review, gene delivery and its applications are discussed in tissue engineering (TE); also, new techniques such as the CRISPR-Cas9 system, synthetics biology and molecular dynamics simulation to improve the efficiency of the scaffolds have been studied. CRISPR-Cas9 is expected to make significant advances in TE in the future. The fundamentals of synthetic biology have developed powerful and flexible methods for programming cells via artificial genetic circuits. The combination of regenerative medicine and artificial biology allows the engineering of cells and organisms for use in TE, biomaterials, bioprocessing and scaffold development. The dynamics of protein adsorption at the scaffold surface at the atomic level can provide valuable guidelines for the future design of TE scaffolds /implants.

    Tweetable abstract

    Engineered cells and smart scaffolds for improving tissue engineering approaches.

    Graphical abstract

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

    References

    • 1. Davies JA, Cachat E. Synthetic biology meets tissue engineering. Biochem. Soc. Trans. 44(3), 696–701 (2016).
    • 2. Hoffman T, Antovski P, Tebon P et al. Synthetic biology and tissue engineering: toward fabrication of complex and smart cellular constructs. Adv. Funct. Mater. 30(26), 1909882 (2020). • Recent advances in the field of synthetic biology have resulted in powerful and flexible strategies for programming cells with artificial genetic circuits, where cell function and behavior can be controlled.
    • 3. O'brien FJ. Biomaterials & scaffolds for tissue engineering. Mater. Today 14(3), 88–95 (2011).
    • 4. Wang T, Naing MW. The potential role of synthetic biology in manufacturing of regenerative medicine. Presented at: IET/SynbiCITE Engineering Biology Conference. IET Conference, London, UK, 13–15 December 2016.
    • 5. Savoji H, Mohammadi MH, Rafatian N et al. Cardiovascular disease models: a game changing paradigm in drug discovery and screening. Biomaterials 198, 3–26 (2019).
    • 6. Madry H, Venkatesan JK, Carballo-Pedrares N, Rey-Rico A, Cucchiarini M. Scaffold-mediated gene delivery for osteochondral repair. Pharmaceutics 12(10), 930 (2020).
    • 7. Jang J-H, Houchin TL, Shea LD. Gene delivery from polymer scaffolds for tissue engineering. Expert Rev. Med. Devices 1(1), 127–138 (2004).
    • 8. Yan X, Chen Y-R, Song Y-F et al. Scaffold-based gene therapeutics for osteochondral tissue engineering. Front. Pharmacol. 10, 1534 (2020).
    • 9. Jafarlou M, Baradaran B, Saedi T et al. An overview of the history, applications, advantages, disadvantages and prospects of gene therapy. J. Biol. Regul. Homeost. Agents 30(2), 315–321 (2016).
    • 10. Balmayor ER, Van Griensven M. Gene therapy for bone engineering. Front. Bioeng. Biotechnol. 3, 9 (2015).
    • 11. Hosseinkhani H, Hosseinkhani M, Gabrielson NP, Pack DW, Khademhosseini A, Kobayashi H. DNA nanoparticles encapsulated in 3D tissue-engineered scaffolds enhance osteogenic differentiation of mesenchymal stem cells. J. Biomed. Mater. Res. A 85(1), 47–60 (2008).
    • 12. Bez M, Sheyn D, Tawackoli W et al. In situ bone tissue engineering via ultrasound-mediated gene delivery to endogenous progenitor cells in mini-pigs. Sci. Transl. Med. 9(390), doi: 10.1126/scitranslmed.aal3128 (2017).
    • 13. Jiang X, Zhao J, Wang S et al. Mandibular repair in rats with premineralized silk scaffolds and BMP-2-modified bMSCs. Biomaterials 30(27), 4522–4532 (2009).
    • 14. Qing W, Guang-Xing C, Lin G, Liu Y. The osteogenic study of tissue engineering bone with BMP2 and BMP7 gene-modified rat adipose-derived stem cell. J. Biomed. Biotechnol. 2012, 410879 (2012).
    • 15. Liu X, Bao C, Xu HHK et al. Osteoprotegerin gene-modified BMSCs with hydroxyapatite scaffold for treating critical-sized mandibular defects in ovariectomized osteoporotic rats. Acta Biomater. 42, 378–388 (2016).
    • 16. Liu Z, Yuan X, Fernandes G et al. The combination of nano-calcium sulfate/platelet rich plasma gel scaffold with BMP2 gene-modified mesenchymal stem cells promotes bone regeneration in rat critical-sized calvarial defects. Stem Cell Res. Ther. 8(1), 122 (2017). • In critical-sized bone defects, scaffolds containing BMP2-modified mesenchymal stem cells successfully promote bone regeneration.
    • 17. Sun XJ, Xia LG, Chou LL et al. Maxillary sinus floor elevation using a tissue engineered bone complex with BMP-2 gene modified bMSCs and a novel porous ceramic scaffold in rabbits. Arch. Oral Biol. 55(3), 195–202 (2010).
    • 18. Wu D-J, Liu S-L, Hao A-H et al. Enhanced repair of segmental bone defects of rats with hVEGF-165 gene-modified endothelial progenitor cells seeded in nanohydroxyapatite/collagen/poly(l-lactic acid) scaffolds. J. Bioact. Compat. Polym. 27(3), 227–243 (2012).
    • 19. Zhu C, Chang Q, Zou D et al. LvBMP-2 gene-modified BMSCs combined with calcium phosphate cement scaffolds for the repair of calvarial defects in rats. J. Mater. Sci. Mater. Med. 22(8), 1965–1973 (2011).
    • 20. Loozen LD, Wegman F, Oner FC, Dhert WJA, Alblas J. Porous bioprinted constructs in BMP-2 non-viral gene therapy for bone tissue engineering. J. Mater. Chem. B 1(48), 6619–6626 (2013).
    • 21. Leng P, Ding CR, Zhang HN, Wang YZ. Reconstruct large osteochondral defects of the knee with hIGF-1 gene enhanced Mosaicplasty. Knee 19(6), 804–811 (2012).
    • 22. Rowland CR, Glass KA, Ettyreddy AR et al. Regulation of decellularized tissue remodeling via scaffold-mediated lentiviral delivery in anatomically-shaped osteochondral constructs. Biomaterials 177, 161–175 (2018).
    • 23. Raftery RM, Mencia Castano I, Chen G et al. Translating the role of osteogenic-angiogenic coupling in bone formation: highly efficient chitosan-pDNA activated scaffolds can accelerate bone regeneration in critical-sized bone defects. Biomaterials 149, 116–127 (2017).
    • 24. Raftery RM, Mencia-Castano I, Sperger S et al. Delivery of the improved BMP-2-Advanced plasmid DNA within a gene-activated scaffold accelerates mesenchymal stem cell osteogenesis and critical size defect repair. J. Control Rel. 283, 20–31 (2018).
    • 25. Li H, Ji Q, Chen X et al. Accelerated bony defect healing based on chitosan thermosensitive hydrogel scaffolds embedded with chitosan nanoparticles for the delivery of BMP2 plasmid DNA. J. Biomed. Mater. Res. A 105(1), 265–273 (2017).
    • 26. Keeney M, Chung MT, Zielins ER et al. Scaffold-mediated BMP-2 minicircle DNA delivery accelerated bone repair in a mouse critical-size calvarial defect model. J. Biomed. Mater. Res. A 104(8), 2099–2107 (2016).
    • 27. Komatsu K, Shibata T, Shimada A et al. Cationized gelatin hydrogels mixed with plasmid DNA induce stronger and more sustained gene expression than atelocollagen at calvarial bone defects in vivo. J. Biomater. Sci. Polym. Ed. 27(5), 419–430 (2016).
    • 28. Gonzalez-Fernandez T, Tierney EG, Cunniffe GM, O'brien FJ, Kelly DJ. Gene delivery of TGF-beta3 and BMP2 in an MSC-laden alginate hydrogel for articular cartilage and endochondral bone tissue engineering. Tissue Eng. Part A 22(9-10), 776–787 (2016).
    • 29. Plonka AB, Khorsand B, Yu N et al. Effect of sustained PDGF nonviral gene delivery on repair of tooth-supporting bone defects. Gene Ther. 24(1), 31–39 (2017).
    • 30. Lee YH, Wu HC, Yeh CW et al. Enzyme-crosslinked gene-activated matrix for the induction of mesenchymal stem cells in osteochondral tissue regeneration. Acta Biomater. 63, 210–226 (2017).
    • 31. Brunger JM, Huynh NP, Guenther CM et al. Scaffold-mediated lentiviral transduction for functional tissue engineering of cartilage. Proc. Natl Acad. Sci. USA 111(9), E798–E806 (2014).
    • 32. Guo CA, Liu XG, Huo JZ, Jiang C, Wen XJ, Chen ZR. Novel gene-modified-tissue engineering of cartilage using stable transforming growth factor-beta1-transfected mesenchymal stem cells grown on chitosan scaffolds. J. Biosci. Bioeng. 103(6), 547–556 (2007).
    • 33. Yang S, Qian Z, Liu D, Wen N, Xu J, Guo X. Integration of C-type natriuretic peptide gene-modified bone marrow mesenchymal stem cells with chitosan/silk fibroin scaffolds as a promising strategy for articular cartilage regeneration. Cell Tissue Bank. 20(2), 209–220 (2019).
    • 34. Li B, Li F, Ma L et al. Poly(lactide-co-glycolide)/fibrin gel construct as a 3D model to evaluate gene therapy of cartilage in vivo. Mol. Pharm. 11(7), 2062–2070 (2014).
    • 35. Wang X, Li Y, Han R et al. Demineralized bone matrix combined bone marrow mesenchymal stem cells, bone morphogenetic protein-2 and transforming growth factor-beta3 gene promoted pig cartilage defect repair. PLOS ONE 9(12), e116061 (2014).
    • 36. Cao L, Yang F, Liu G et al. The promotion of cartilage defect repair using adenovirus mediated SOX9 gene transfer of rabbit bone marrow mesenchymal stem cells. Biomaterials 32(16), 3910–3920 (2011).
    • 37. Li B, Yang J, Ma L, Li F, Tu Z, Gao C. Fabrication of poly(lactide-co-glycolide) scaffold filled with fibrin gel, mesenchymal stem cells, and poly(ethylene oxide)-b-poly(L-lysine)/TGF-beta1 plasmid DNA complexes for cartilage restoration in vivo. J. Biomed. Mater. Res. A 101(11), 3097–3108 (2013). • Shows that the synthesized construct has low cytotoxicity and high biocompatibility, making it an effective candidate for cartilage regeneration in vivo.
    • 38. Lolli A, Narcisi R, Lambertini E et al. Silencing of antichondrogenic microRNA-221 in human mesenchymal stem cells promotes cartilage repair in vivo. Stem Cells 34(7), 1801–1811 (2016).
    • 39. Moutos FT, Glass KA, Compton SA et al. Anatomically shaped tissue-engineered cartilage with tunable and inducible anticytokine delivery for biological joint resurfacing. Proc. Natl Acad. Sci. USA 113(31), E4513–4522 (2016).
    • 40. Griffin DJ, Ortved KF, Nixon AJ, Bonassar LJ. Mechanical properties and structure-function relationships in articular cartilage repaired using IGF-I gene-enhanced chondrocytes. J. Orthop. Res. 34(1), 149–153 (2016).
    • 41. Liang W, Zhu C, Liu F et al. Integrin beta1 gene therapy enhances in vitro creation of tissue-engineered cartilage under periodic mechanical stress. Cell Physiol. Biochem. 37(4), 1301–1314 (2015).
    • 42. Venkatesan JK, Gardner O, Rey-Rico A et al. Improved chondrogenic differentiation of rAAV SOX9-modified human MSCs seeded in fibrin-polyurethane scaffolds in a hydrodynamic environment. Int. J. Mol. Sci. 19(9), 2635 (2018).
    • 43. Venkatesan JK, Moutos FT, Rey-Rico A et al. Chondrogenic differentiation processes in human bone-marrow aspirates seeded in three-dimensional-woven poly(varepsilon-caprolactone) scaffolds enhanced by recombinant adeno-associated virus-mediated SOX9 gene transfer. Hum. Gene Ther. 29(11), 1277–1286 (2018).
    • 44. Lu CH, Yeh TS, Yeh CL et al. Regenerating cartilages by engineered ASCs: prolonged TGF-beta3/BMP-6 expression improved articular cartilage formation and restored zonal structure. Mol. Ther. 22(1), 186–195 (2014).
    • 45. Rey-Rico A, Venkatesan JK, Schmitt G et al. rAAV-mediated overexpression of TGF-beta via vector delivery in polymeric micelles stimulates the biological and reparative activities of human articular chondrocytes in vitro and in a human osteochondral defect model. Int. J. Nanomed. 12, 6985–6996 (2017).
    • 46. He S, Xia T, Wang H, Wei L, Luo X, Li X. Multiple release of polyplexes of plasmids VEGF and bFGF from electrospun fibrous scaffolds towards regeneration of mature blood vessels. Acta Biomater. 8(7), 2659–2669 (2012).
    • 47. Guo R, Xu S, Ma L, Huang A, Gao C. The healing of full-thickness burns treated by using plasmid DNA encoding VEGF-165 activated collagen-chitosan dermal equivalents. Biomaterials 32(4), 1019–1031 (2011).
    • 48. Liu X, Ma L, Liang J, Zhang B, Teng J, Gao C. RNAi functionalized collagen-chitosan/silicone membrane bilayer dermal equivalent for full-thickness skin regeneration with inhibited scarring. Biomaterials 34(8), 2038–2048 (2013).
    • 49. Hu DH, Zhang ZF, Zhang YG et al. A potential skin substitute constructed with hEGF gene modified HaCaT cells for treatment of burn wounds in a rat model. Burns 38(5), 702–712 (2012).
    • 50. Nelson CE, Gupta MK, Adolph EJ, Guelcher SA, Duvall CL. siRNA delivery from an injectable scaffold for wound therapy. Adv. Wound Care (New Rochelle) 2(3), 93–99 (2013).
    • 51. Thiersch M, Rimann M, Panagiotopoulou V et al. The angiogenic response to PLL-g-PEG-mediated HIF-1alpha plasmid DNA delivery in healthy and diabetic rats. Biomaterials 34(16), 4173–4182 (2013).
    • 52. De Laporte L, Yan AL, Shea LD. Local gene delivery from ECM-coated poly(lactide-co-glycolide) multiple channel bridges after spinal cord injury. Biomaterials 30(12), 2361–2368 (2009).
    • 53. De Laporte L, Huang A, Ducommun MM et al. Patterned transgene expression in multiple-channel bridges after spinal cord injury. Acta Biomater. 6(8), 2889–2897 (2010).
    • 54. Li LM, Huang LL, Jiang XC, Chen JC, Ouyang HW, Gao JQ. Transplantation of BDNF gene recombinant mesenchymal stem cells and adhesive peptide-modified hydrogel scaffold for spinal cord repair. Curr. Gene Ther. 18(1), 29–39 (2018).
    • 55. Li SL, Liu Y, Hui L. Construction of engineering adipose-like tissue in vivo utilizing human insulin gene-modified umbilical cord mesenchymal stromal cells with silk fibroin 3D scaffolds. J. Tissue Eng. Regen. Med. 9(12), E267–275 (2015). • The adenovirus-transfected human umbilical cord mesenchymal stem cells are compatible with the silk fibroin scaffold, and adenoviral transfection of the human insulin gene can be applied to construct tissue-engineered adipose tissue.
    • 56. Guan Y, Ou L, Hu G et al. Tissue engineering of urethra using human vascular endothelial growth factor gene-modified bladder urothelial cells. Artif. Organs 32(2), 91–99 (2008).
    • 57. Zhou W, He DQ, Liu JY et al. Angiogenic gene-modified myoblasts promote vascularization during repair of skeletal muscle defects. J. Tissue Eng. Regen. Med. 9(12), 1404–1416 (2015).
    • 58. Yang F, Cho SW, Son SM et al. Genetic engineering of human stem cells for enhanced angiogenesis using biodegradable polymeric nanoparticles. Proc. Natl Acad. Sci. USA 107(8), 3317–3322 (2010).
    • 59. Charpentier E, Doudna JA. Rewriting a genome. Nature 495(7439), 50–51 (2013).
    • 60. Charpentier E, Marraffini LA. Harnessing CRISPR-Cas9 immunity for genetic engineering. Curr. Opin. Microbiol. 19, 114–119 (2014). • The authors describe recent advancements for type II CRISPR-Cas immunity and how this research led to revolutionary genome editing applications.
    • 61. Zhang Y, Heidrich N, Ampattu BJ et al. Processing-independent CRISPR RNAs limit natural transformation in Neisseria meningitidis. Mol. Cell 50(4), 488–503 (2013).
    • 62. Davis B, Farhang N, Bowles R. Use of CRISPR genome-editing technology to promote osteogenic differentiation in human adipose-derived mesenchymal stem cells (hAD-MSCs). Presented at: UCUR 2016 (2022). https://epubs.utah.edu/index.php/URJ/article/view/2718 • The current applications of CRISPR-Cas systems in tissue engineering have highlighted tissue architecture construction, disease model development, cell differentiation, and immune response circumvention. Stem cells are excellent cell sources with enormous promise for tissue engineering applications.
    • 63. Rodrigues LR, Kluskens LD. Synthetic biology & bioinformatics: prospects in the cancer arena. J. Appl. Bioinforma. Comput. Biol. 8, 159–186 (2011).
    • 64. Evans A, Ratcliffe E. Rising influence of synthetic biology in regenerative medicine. Eng. Biol. 1(1), 24–29 (2017).
    • 65. Cachat E, Davies J. Application of synthetic biology to regenerative medicine. J. Bioeng. Biomed. Sci. 2(3) doi: 10.4172/2155-9538.S2-003 (2011).
    • 66. Huang B, Lou Y, Li T et al. Molecular dynamics simulations of adsorption and desorption of bone morphogenetic protein-2 on textured hydroxyapatite surfaces. Acta Biomater. 80, 121–130 (2018).
    • 67. Sarmadi M, Shamloo A, Mohseni M. Utilization of molecular dynamics simulation coupled with experimental assays to optimize biocompatibility of an electrospun PCL/PVA scaffold. PLOS ONE 12(1), e0169451 (2017). • A computational approach that estimates the adhesion of cells to the constructs may be effective for reducing the number of specimens and accelerating the optimization process. This approach is especially useful for composite constructs, which are made up of several polymers and for developing new polymers.
    • 68. Damasceno PKF, De Santana TA, Santos GC et al. Genetic engineering as a strategy to improve the therapeutic efficacy of mesenchymal stem/stromal cells in regenerative medicine. Front. Cell Dev. Biol. 8, 737 (2020).
    • 69. Nie W-B, Zhang D, Wang L-S. Growth factor gene-modified mesenchymal stem cells in tissue regeneration. Drug Des. Devel. Ther. 14, 1241 (2020). • Nonviral vectors (like polymers or lipids) and viral vectors (including retroviruses, lentiviruses, adenoviruses and adeno-associated viruses) have been employed to overexpress genes in cells such as mesenchymal stem cells.
    • 70. Frenz DA, Liu W, Williams JD et al. Induction of chondrogenesis: requirement for synergistic interaction of basic fibroblast growth factor and transforming growth factor-beta. Development 120(2), 415–424 (1994).
    • 71. Chen Z, Wei J, Zhu J et al. Chm-1 gene-modified bone marrow mesenchymal stem cells maintain the chondrogenic phenotype of tissue-engineered cartilage. Curr. Stem Cell Res. Ther. 7(1), 1–11 (2016).
    • 72. Peng H, Wright V, Usas A et al. Synergistic enhancement of bone formation and healing by stem cell-expressed VEGF and bone morphogenetic protein-4. J. Clin. Investig. 110(6), 751–759 (2002).
    • 73. Huang YC, Kaigler D, Rice KG, Krebsbach PH, Mooney DJ. Combined angiogenic and osteogenic factor delivery enhances bone marrow stromal cell‐driven bone regeneration. J. Bone Miner. Res. 20(5), 848–857 (2005).
    • 74. Jiang J, Fan C-Y, Zeng B-F. Experimental construction of BMP2 and VEGF gene modified tissue engineering bone in vitro. Int. J. Mol. Sci. 12(3), 1744–1755 (2011).
    • 75. Dai F, Shi D, He W et al. hCTLA4-gene modified human bone marrow-derived mesenchymal stem cells as allogeneic seed cells in bone tissue engineering. Tissue Eng. 12(9), 2583–2590 (2006).
    • 76. Deng Y, Zhou H, Yan C et al. In vitro osteogenic induction of bone marrow stromal cells with encapsulated gene-modified bone marrow stromal cells and in vivo implantation for orbital bone repair. Tissue Eng. Part A 20(13-14), 2019–2029 (2014).
    • 77. Dong J-L, Li L-X, Mu W-D et al. Bone regeneration with BMP-2 gene-modified mesenchymal stem cells seeded on nano-hydroxyapatite/collagen/poly (l-lactic Acid) scaffolds. J. Bioact. Compat. Polym. 25(6), 547–566 (2010).
    • 78. Han Y, Han Y, Pan Y, Chen Y, Chai J. Transplantation of microencapsulated VEGF genetically modified cells improves angiogenesis of implanted xenogeneic acellular dermis on wound. Transplant. Proc. 42, 1935–1943 (2010).
    • 79. Han Y, Tao R, Han Y et al. Microencapsulated VEGF gene–modified umbilical cord mesenchymal stromal cells promote the vascularization of tissue-engineered dermis: an experimental study. Cytotherapy 16(2), 160–169 (2014).
    • 80. Hong W, Huang M, Wei Y, Wei X. A new and promising application of gene editing: CRISPR-controlled smart materials for tissue engineering, bioelectronics, and diagnostics. Sci. China Life Sci. 62(11), 1547–1549 (2019).
    • 81. English MA, Soenksen LR, Gayet RV et al. Programmable CRISPR-responsive smart materials. Science 365(6455), 780–785 (2019).
    • 82. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337(6096), 816–821 (2012).
    • 83. Armstrong JPK, Stevens MM. Emerging technologies for tissue engineering: from gene editing to personalized medicine. Tissue Eng. Part A 25(9-10), 688–692 (2019).
    • 84. Ali Q, Malik S, Malik A, Hafeez MN, Salman S. Role of modern technologies in tissue engineering. Arch. Neurosci. 7(1), e90394 (2020).
    • 85. Bakhshian-Dehkordi E, Safaei M, Fattahi S, Faghani M, Deris F, Chaleshtori MH. The association of VEGF rs833061 and rs2010963 polymorphisms with susceptibility to colorectal cancer in an Iranian population. Cancer Epidemiol. 75, 102041 (2021).
    • 86. Raftery RM, Walsh DP, Blokpoel Ferreras L et al. Highly versatile cell-penetrating peptide loaded scaffold for efficient and localised gene delivery to multiple cell types: from development to application in tissue engineering. Biomaterials 216, 119277 (2019).
    • 87. Doudna JA, Charpentier E. The new frontier of genome engineering with CRISPR-Cas9. Science 346(6213), 1258096 (2014).
    • 88. Polstein LR, Gersbach CA. A light-inducible CRISPR-Cas9 system for control of endogenous gene activation. Nat. Chem. Biol. 11(3), 198–200 (2015).
    • 89. Ablain J, Durand EM, Yang S, Zhou Y, Zon LI. A CRISPR/Cas9 vector system for tissue-specific gene disruption in zebrafish. Dev. Cell 32(6), 756–764 (2015).
    • 90. Chew SY. Sequential drug/gene delivery in tissue engineering & regenerative medicine. Adv. Drug Deliv. Rev. 149–150, 1 (2019).
    • 91. Malek-Khatabi A, Javar HA, Dashtimoghadam E, Ansari S, Hasani-Sadrabadi MM, Moshaverinia A. In situ bone tissue engineering using gene delivery nanocomplexes. Acta Biomater. 108, 326–336 (2020).
    • 92. Park K. Bioink-guided spatio-temporal gene delivery for tissue engineering. J. Control. Rel. 301, 190 (2019).
    • 93. Gonzalez-Fernandez T, Rathan S, Hobbs C et al. Pore-forming bioinks to enable spatio-temporally defined gene delivery in bioprinted tissues. J. Control. Rel. 301, 13–27 (2019).
    • 94. Gonzalez-Fernandez T, Sathy B, Hobbs C et al. Mesenchymal stem cell fate following non-viral gene transfection strongly depends on the choice of delivery vector. Acta Biomater. 55, 226–238 (2017).
    • 95. Curtin CM, Tierney EG, Mcsorley K, Cryan SA, Duffy GP, O'brien FJ. Combinatorial gene therapy accelerates bone regeneration: non‐viral dual delivery of VEGF and BMP2 in a collagen‐nanohydroxyapatite scaffold. Adv. Healthc. Mater. 4(2), 223–227 (2015).
    • 96. Gholami A, Hashemi SA, Yousefi K et al. 3D nanostructures for tissue engineering, cancer therapy, and gene delivery. J. Nanomater. 2020, 1–24 (2020).
    • 97. Walsh DP, Raftery RM, Murphy R et al. Gene activated scaffolds incorporating star-shaped polypeptide-pDNA nanomedicines accelerate bone tissue regeneration in vivo. Biomater. Sci. 9(14), 4984–4999 (2021).
    • 98. Wu P, Chen H, Jin R et al. Non-viral gene delivery systems for tissue repair and regeneration. J. Transl. Med. 16(1), 29 (2018).
    • 99. Curtin CM, Cunniffe GM, Lyons FG et al. Innovative collagen nano‐hydroxyapatite scaffolds offer a highly efficient non‐viral gene delivery platform for stem cell‐mediated bone formation. Adv. Mater. 24(6), 749–754 (2012).
    • 100. Godbey W, Wu K, Hirasaki G, Mikos A. Improved packing of poly (ethylenimine)/DNA complexes increases transfection efficiency. Gene Ther. 6(8), 1380–1388 (1999).
    • 101. Kulkarni M, Greiser U, O'brien T, Pandit A. Liposomal gene delivery mediated by tissue-engineered scaffolds. Trends Biotechnol. 28(1), 28–36 (2010).
    • 102. Monteiro N, Ribeiro D, Martins A et al. Instructive nanofibrous scaffold comprising runt-related transcription factor 2 gene delivery for bone tissue engineering. ACS Nano 8(8), 8082–8094 (2014).
    • 103. Abbasi E, Aval SF, Akbarzadeh A et al. Dendrimers: synthesis, applications, and properties. Nanoscale Res. Lett. 9(1), 1–10 (2014).
    • 104. Walsh DP, Heise A, O'brien FJ, Cryan SA. An efficient, non-viral dendritic vector for gene delivery in tissue engineering. Gene Ther. 24(11), 681–691 (2017).
    • 105. Safaei M, Mobini GR, Abiri A, Shojaeian A. Synthetic biology in various cellular and molecular fields: applications, limitations, and perspective. Mol. Biol. Rep. 47(8), 6207–6216 (2020).
    • 106. Hodgkinson CP, Gomez JA, Mirotsou M, Dzau VJ. Genetic engineering of mesenchymal stem cells and its application in human disease therapy. Hum. Gene Ther. 21(11), 1513–1526 (2010).
    • 107. Karlsson M, Weber W, Fussenegger M. De novo design and construction of an inducible gene expression system in mammalian cells. In: Methods in Enzymology. 239–253, Elsevier, USA (2011).
    • 108. Liu H, Yu X, Li K et al. Photoexcited CRY2 interacts with CIB1 to regulate transcription and floral initiation in Arabidopsis. Science 322(5907), 1535–1539 (2008).
    • 109. Sadelain M, Brentjens R, Rivière I. The basic principles of chimeric antigen receptor design. Cancer Discov. 3(4), 388–398 (2013).
    • 110. Ma Y, Li J, Yao Y, Wei D, Wang R, Wu Q. A controlled double-duration inducible gene expression system for cartilage tissue engineering. Sci. Rep. 6(1), 1–13 (2016).
    • 111. Sakar MS, Neal D, Boudou T et al. Formation and optogenetic control of engineered 3D skeletal muscle bioactuators. Lab Chip 12(23), 4976–4985 (2012).
    • 112. Chan V, Neal DM, Uzel SG, Kim H, Bashir R, Asada HH. Fabrication and characterization of optogenetic, multi-strip cardiac muscles. Lab Chip 15(10), 2258–2268 (2015).
    • 113. Ehrbar M, Schoenmakers R, Christen EH, Fussenegger M, Weber W. Drug-sensing hydrogels for the inducible release of biopharmaceuticals. Nat. Mater. 7(10), 800–804 (2008).
    • 114. Kämpf MM, Christen EH, Ehrbar M et al. A gene therapy technology‐based biomaterial for the trigger‐inducible release of biopharmaceuticals in mice. Adv. Funct. Mater. 20(15), 2534–2538 (2010).
    • 115. Rajagopal K, Schneider JP. Self-assembling peptides and proteins for nanotechnological applications. Curr. Opin. Struct. Biol. 14(4), 480–486 (2004).
    • 116. Vogel AM, Persson KM, Seamons TR, Deans TL. Synthetic biology for improving cell fate decisions and tissue engineering outcomes. Emerg. Top Life Sci. 3(5), 631–643 (2019).
    • 117. Shamloo A, Pedram MZ, Heidari H, Alasty A. Computing the blood–brain barrier (BBB) diffusion coefficient: a molecular dynamics approach. J. Magn. Magn. Mater. 410, 187–197 (2016).
    • 118. Ebrahimi S, Ghafoori-Tabrizi K, Rafii-Tabar H. Molecular dynamics simulation of the adhesive behavior of collagen on smooth and randomly rough TiO2 and Al2O3 surfaces. Comput. Mater. Sci. 71, 172–178 (2013).
    • 119. Raffaini G, Ganazzoli F. Molecular modelling of protein adsorption on the surface of titanium dioxide polymorphs. Philos. Trans. Royal Soc. A 370(1963), 1444–1462 (2012).
    • 120. Regis S, Youssefian S, Jassal M, Phaneuf MD, Rahbar N, Bhowmick S. Fibronectin adsorption on functionalized electrospun polycaprolactone scaffolds: experimental and molecular dynamics studies. J. Biomed. Mater. Res. A 102(6), 1697–1706 (2014).
    • 121. Gray JJ. The interaction of proteins with solid surfaces. Curr. Opin. Struct. Biol. 14(1), 110–115 (2004).
    • 122. Hlady V, Buijs J. Protein adsorption on solid surfaces. Curr. Opin. Biotechnol. 7(1), 72–77 (1996).
    • 123. Marquetti I. Molecular modeling of bone morphogenetic protein for tissue engineering applications. Presented at: Proceedings of the 2018 IISE Annual Conference. 1759–1764 (2018).
    • 124. Utesch T, Daminelli G, Mroginski MA. Molecular dynamics simulations of the adsorption of bone morphogenetic protein-2 on surfaces with medical relevance. Langmuir 27(21), 13144–13153 (2011).
    • 125. Oliveira A, Gemming S, Seifert G. Molecular dynamics simulations of BMP‐2 adsorption on a hydrophobic surface. Materialwissenschaft und Werkstofftechnik 41(12), 1048–1053 (2010).
    • 126. Mücksch C, Urbassek HM. Adsorption of BMP-2 on a hydrophobic graphite surface: a molecular dynamics study. Chem. Phy. Lett. 510(4-6), 252–256 (2011).
    • 127. Shen J-W, Wu T, Wang Q, Pan H-H. Molecular simulation of protein adsorption and desorption on hydroxyapatite surfaces. Biomaterials 29(5), 513–532 (2008).
    • 128. Lai ZB, Wang M, Yan C, Oloyede A. Molecular dynamics simulation of mechanical behavior of osteopontin-hydroxyapatite interfaces. J. Mech. Behav. Biomed. Mater. 36, 12–20 (2014).
    • 129. Plow EF, Haas TA, Zhang L, Loftus J, Smith JW. Ligand binding to integrins. J. Biol. Chem. 275(29), 21785–21788 (2000).
    • 130. Reverdatto S, Burz DS, Shekhtman A. Peptide aptamers: development and applications. Curr. Top Med. Chem. 15(12), 1082 (2015).
    • 131. Debaisieux S, Rayne F, Yezid H, Beaumelle B. The ins and outs of HIV-1 Tat. Traffic 13(3), 355–363 (2012).
    • 132. Clima L, Rotara A, Cojocaru C, Pinteala M, Simionescu B. Polymer engineering focusing on DRUG/GENE delivery and tissue engineering. Presented at: 2015 E-Health and Bioengineering Conference (EHB). doi: 10.1109/EHB.2015.7391491, 19–21 November 2015.
    • 133. Perez JJ, Perez RA, Perez A. Computational modeling as a tool to investigate PPI: from drug design to tissue engineering. Front. Mol. Biosci. 8, 681617 (2021). • A scaffold based on computational methods with the feature of self-assembly peptide can be designed. These self-assembling peptides should contain a sequence capable of self-assembly and a sequence motif that is accessible and recognized via integrin in order to control their behavior.
    • 134. Zhu J, Zhu J, Negri A et al. Closed headpiece of integrin αIIbβ3 and its complex with an αIIbβ3-specific antagonist that does not induce opening. Blood 116(23), 5050–5059 (2010).
    • 135. Yin H, Slusky JS, Berger BW et al. Computational design of peptides that target transmembrane helices. Science 315(5820), 1817–1822 (2007).
    • 136. Shandler SJ, Korendovych IV, Moore DT et al. Computational design of a β-peptide that targets transmembrane helices. J. Am. Chem. Soc. 133(32), 12378–12381 (2011).
    • 137. Lesch HP, Makkonen K-E, Laitinen A et al. Requirements for baculoviruses for clinical gene therapy applications. J. Invertebr. Pathol 107 S106–S112 (2011).
    • 138. Segura MM, Garnier A, Durocher Y, Ansorge S, Kamen A. New protocol for lentiviral vector mass production. In: Lentivirus Gene Engineering Protocols. Springer, NJ, USA, 39–52 (2010).
    • 139. Walther W, Stein U. Viral vectors for gene transfer. Drugs 60(2), 249–271 (2000).
    • 140. Park S-Y, Kim K-H, Kim S, Lee Y-M, Seol Y-J. BMP-2 gene delivery-based bone regeneration in dentistry. Pharmaceutics 11(8), 393 (2019).
    • 141. Paolini Sguazzi G, Muto V, Tartaglia M, Bertini E, Compagnucci C. Induced pluripotent stem cells (iPSCs) and gene therapy: a new era for the treatment of neurological diseases. Int. J. Mol. Sci. 22(24), 13674 (2021).
    • 142. Johnson MB, March AR, Morsut L. Engineering multicellular systems: using synthetic biology to control tissue self-organization. Curr. Opin. Biomed. Eng. 4, 163–173 (2017).
    • 143. Mitchell SM, Pajovich HT, Broas SM, Hugo MM, Banerjee IA. Molecular dynamics simulations and in vitro studies of hybrid decellularized leaf-peptide-polypyrrole composites for potential tissue engineering applications. J. Biomol. Struct. Dyn. 1–16 (2021).
    • 144. Kofron MD, Laurencin CT. Bone tissue engineering by gene delivery. Adv. Drug Del. Rev. 58(4), 555–576 (2006).
    • 145. Giacca M. Ethical and social problems of gene therapy. In: Gene Therapy Springer, Milano, 283–292 (2010).
    • 146. Paolini Sguazzi G, Muto V, Tartaglia M, Bertini E, Compagnucci C. Induced pluripotent stem cells (iPSCs) and gene therapy: a new era for the treatment of neurological diseases. Int. J. Mol. Sci. 22(24), 13674 (2021).
    • 147. Trump BD, Cegan JC, Wells E, Keisler J, Linkov I. A critical juncture for synthetic biology: lessons from nanotechnology could inform public discourse and further development of synthetic biology. EMBO Rep. 19(7), e46153 (2018).
    • 148. Hospital A, Goñi JR, Orozco M, Gelpí JL. Molecular dynamics simulations: advances and applications. Adv. Appl. Bio. Chem. 8, 37 (2015).