1,029
Views
0
CrossRef citations to date
0
Altmetric
Review Article

A mini-review on gene delivery technique using nanoparticles-mediated photoporation induced by nanosecond pulsed laser

, , , , , , & show all
Article: 2306231 | Received 06 Apr 2023, Accepted 29 Dec 2023, Published online: 21 Jan 2024

References

  • Barber MA. (1911). A technic for the inoculation of bacteria and other substances into living cells. The J Infect Diseases 8:1–9. doi:10.1093/infdis/8.3.348.
  • Baumgart J, Humbert L, Boulais E, et al. (2012). Off-resonance plasmonic enhanced femtosecond laser optoporation and transfection of cancer cells. Biomaterials 33:2345–50. doi:10.1016/j.biomaterials.2011.11.062.
  • Bergeron E, Boutopoulos C, Martel R, et al. (2015). Cell-specific optoporation with near-infrared ultrafast laser and functionalized gold nanoparticles. Nanoscale 7:17836–47. doi:10.1039/c5nr05650k.
  • Breunig HG, Uchugonova A, Batista A, König K. (2014). High-throughput continuous flow femtosecond laser-assisted cell optoporation and transfection. Microsc Res Tech 77:974–9. doi:10.1002/jemt.22423.
  • Chapman CF, Liu Y, Sonek GJ, Tromberg BJ. (1995). The use of exogenous fluorescent probes for temperature measurements in single living cells. Photochem Photobiol 62:416–25. doi:10.1111/j.1751-1097.1995.tb02362.x.
  • Chiappini C, De Rosa E, Martinez JO, et al. (2015). Biodegradable silicon nanoneedles delivering nucleic acids intracellularly induce localized in vivo neovascularization. Nat Mater 14:532–9. doi:10.1038/nmat4249.
  • Clark IB, Hanania EG, Stevens J, et al. (2006). Optoinjection for efficient targeted delivery of a broad range of compounds and macromolecules into diverse cell types. J Biomed Opt 11:014034. doi:10.1117/1.2168148.
  • Du X, Wang J, Chen L, et al. (2021). Delivery of Foreign Materials into Adherent Cells by Gold Nanoparticle-Mediated Photoporation. Membranes 11:550. doi:10.3390/membranes11080550.
  • Du X, Wang J, Zhou Q, et al. (2018). Advanced physical techniques for gene delivery based on membrane perforation. Drug Deliv 25:1516–25. doi:10.1080/10717544.2018.1480674.
  • Duckert B, Vinkx S, Braeken D, Fauvart M. (2021). Single-cell transfection technologies for cell therapies and gene editing. J Control Release 330:963–75. doi:10.1016/j.jconrel.2020.10.068.
  • Esrick EB, Lehmann LE, Biffi A, et al. (2021). Post-transcriptional genetic silencing of BCL11A to treat sickle cell disease. N Engl J Med 384:205–15. doi:10.1056/NEJMoa2029392.
  • Fu L, Wang S, Xin J, et al. (2018). Experimental investigation on multiple breakdown in water induced by focused nanosecond laser. Opt Express 26:28560–75. doi:10.1364/OE.26.028560.
  • Gao X, Kim KS, Liu D. (2007). Nonviral gene delivery: What we know and what is next. Aaps J 9:E92–104. doi:10.1208/aapsj0901009.
  • Gu L, Koymen AR, Mohanty SK. (2014). Crystalline magnetic carbon nanoparticle assisted photothermal delivery into cells using CW near-infrared laser beam. Sci Rep 4:5106. doi:10.1038/srep05106.
  • Gupta P, Kar S, Kumar A, et al. (2021). Pulsed laser assisted high-throughput intracellular delivery in hanging drop based three dimensional cancer spheroids. Analyst 146:4756–66. doi:10.1039/d0an02432e.
  • Harizaj A, Descamps B, Mangodt C, et al. (2021a). Cytosolic delivery of gadolinium via photoporation enables improved in vivo magnetic resonance imaging of cancer cells. Biomater Sci 9:4005–18. doi:10.1039/d1bm00479d.
  • Harizaj A, Van Hauwermeiren F, Stremersch S, et al. (2021b). Nanoparticle-sensitized photoporation enables inflammasome activation studies in targeted single cells. Nanoscale 13:6592–604. doi:10.1039/d0nr05067a.
  • Harizaj A, Wels M, Raes L, et al. (2021c). Photoporation with biodegradable polydopamine nanosensitizers enables safe and efficient delivery of mRNA in human T cells. Adv Funct Materials 31:2102472. doi:10.1002/adfm.202102472.
  • Heinemann D, Kalies S, Schomaker M, et al. (2014). Delivery of proteins to mammalian cells via gold nanoparticle mediated laser transfection. Nanotechnology 25:245101. doi:10.1088/0957-4484/25/24/245101.
  • Heinemann D, Schomaker M, Kalies S, et al. (2013). Gold nanoparticle mediated laser transfection for efficient siRNA mediated gene knock down. PLoS One 8:e58604. doi:10.1371/journal.pone.0058604.
  • Heller LC, Heller R. (2006). In vivo electroporation for gene therapy. Hum Gene Ther 17:890–7. doi:10.1089/hum.2006.17.890.
  • Hosseinpour S, Walsh LJ. (2021). Laser-assisted nucleic acid delivery: a systematic review. J Biophotonics 14:e202000295. doi:10.1002/jbio.202000295.
  • Houthaeve G, Xiong R, Robijns J, et al. (2018). Targeted perturbation of nuclear envelope integrity with vapor nanobubble-mediated photoporation. ACS Nano 12:7791–802. doi:10.1021/acsnano.8b01860.
  • Kaladharan K, Kumar A, Gupta P, et al. (2021). Microfluidic based physical approaches towards single-cell intracellular delivery and analysis. Micromachines 12:631. doi:10.3390/mi12060631.
  • Kalies S, Birr T, Heinemann D, et al. (2014). Enhancement of extracellular molecule uptake in plasmonic laser perforation. J Biophotonics 7:474–82. doi:10.1002/jbio.201200200.
  • Kar S, Loganathan M, Dey K, et al. (2018). Single-cell electroporation: current trends, applications and future prospects. J Micromech Microeng 28:123002. doi:10.1088/1361-6439/aae5ae.
  • Karki A, Giddings E, Carreras A, et al. (2019). Sonoporation as an Approach for siRNA delivery into T cells. Ultrasound Med Biol 45:3222–31. doi:10.1016/j.ultrasmedbio.2019.06.406.
  • Kim TK, Eberwine JH. (2010). Mammalian cell transfection: the present and the future. Anal Bioanal Chem 397:3173–8. doi:10.1007/s00216-010-3821-6.
  • Klein RM, Wolf ED, Wu R, Sanford JC. (1992). High-velocity microprojectiles for delivering nucleic acids into living cells. 1987. Biotechnology 24:384–6.
  • Kohn DB, Booth C, Shaw KL, et al. (2021). Autologous ex vivo lentiviral gene therapy for adenosine deaminase. N Engl J Med 384:2002–13. doi:10.1056/NEJMoa2027675.
  • Krasieva TB, Chapman CF, LaMorte VJ, et al. (1998). Cell permeabilization and molecular transport by laser microirradiation, in: Optical Investigations of Cells in Vitro and in Vivo, pp 38–44.
  • Lachaine R, Boutopoulos C, Lajoie PY, et al. (2016). Rational design of plasmonic nanoparticles for enhanced cavitation and cell perforation. Nano Lett 16:3187–94. doi:10.1021/acs.nanolett.6b00562.
  • Lakshmanan S, Gupta GK, Avci P, et al. (2014). Physical energy for drug delivery; poration, concentration and activation. Adv Drug Deliv Rev 71:98–114. doi:10.1016/j.addr.2013.05.010.
  • Liu D, Wang L, Wang Z, Cuschieri A. (2012). Magnetoporation and magnetolysis ofcancer cells via carbon nanotubes induced by rotating magnetic field. Nano Lett 12:5117–21. doi:10.1021/nl301928z.
  • Liu J, Li C, Brans T, et al. (2020). Surface functionalization with polyethylene glycol and polyethyleneimine improves the performance of graphene-based materials for safe and efficient intracellular delivery by laser-induced photoporation. Int J Mol Sci 21:1540.
  • Liu J, Xiong R, Brans T, et al. (2018). Repeated photoporation with graphene quantum dots enables homogeneous labeling of live cells with extrinsic markers for fluorescence microscopy. Light Sci Appl 7:47. doi:10.1038/s41377-018-0048-3.
  • Madrid M, Saklayen N, Shen W, et al. (2018). Laser-activated self-assembled thermoplasmonic nanocavity substrates for intracellular delivery. ACS Appl Bio Mater 1:1793–9. doi:10.1021/acsabm.8b00447.
  • Man T, Zhu X, Chow YT, et al. (2019). Intracellular photothermal delivery for suspension cells using sharp nanoscale tips in microwells. ACS Nano 13:10835–44. doi:10.1021/acsnano.9b06025.
  • Mehier-Humbert S, Guy RH. (2005). Physical methods for gene transfer: improving the kinetics of gene delivery into cells. Adv Drug Deliv Rev 57:733–53. doi:10.1016/j.addr.2004.12.007.
  • Mellott AJ, Forrest ML, Detamore MS. (2013). Physical non-viral gene delivery methods for tissue engineering. Ann Biomed Eng 41:446–68. doi:10.1007/s10439-012-0678-1.
  • Minai L, Zeidan A, Yeheskely-Hayon D, et al. (2016). Experimental proof for the role of nonlinear photoionization in plasmonic phototherapy. Nano Lett 16:4601–7. doi:10.1021/acs.nanolett.6b01901.
  • Mohan L, Kar S, Nagai M, Santra TS. (2021). Electrochemical fabrication of TiO2 micro-flowers for an efficient intracellular delivery using nanosecond light pulse. Mater Chem Phys 267:124604. doi:10.1016/j.matchemphys.2021.124604.
  • Pasi KJ, Rangarajan S, Mitchell N, et al. (2020). Multiyear Follow-up of AAV5-hFVIII-SQ Gene Therapy for Hemophilia A. N Engl J Med 382:29–40. doi:10.1056/NEJMoa1908490.
  • Patskovsky S, Qi M, Meunier M. (2020). Single point single-cell nanoparticle mediated pulsed laser optoporation. Analyst 145:523–9. doi:10.1039/c9an01869g.
  • Patskovsky S, Bergeron E, Rioux D, Meunier M. (2015). Wide-field hyperspectral 3D imaging of functionalized gold nanoparticles targeting cancer cells by reflected light microscopy. J Biophotonics 8:401–7. doi:10.1002/jbio.201400025.
  • Paunovska K, Loughrey D, Dahlman JE. (2022). Drug delivery systems for RNA therapeutics. Nat Rev Genet 23:265–80. doi:10.1038/s41576-021-00439-4.
  • Pylaev T, Vanzha E, Avdeeva E, et al. (2019). A novel cell transfection platform based on laser optoporation mediated by Au nanostar layers. J Biophotonics 12:e201800166. doi:10.1002/jbio.201800166.
  • Qin Z, Bischof JC. (2012). Thermophysical and biological responses of gold nanoparticle laser heating. Chem Soc Rev 41:1191–217. doi:10.1039/c1cs15184c.
  • Raes L, Pille M, Harizaj A, et al. (2021). Cas9 RNP transfection by vapor nanobubble photoporation for ex vivo cell engineering. Mol Ther Nucleic Acids 25:696–707. doi:10.1016/j.omtn.2021.08.014.
  • Raes L, Van Hecke C, Michiels J, et al. (2019). Gold nanoparticle-mediated photoporation enables delivery of macromolecules over a wide range of molecular weights in human CD4+ T cells. Crystals 9:411. doi:10.3390/cryst9080411.
  • Ramon J, Xiong R, De Smedt SC, et al. (2021). Vapor nanobubble-mediated photoporation constitutes a versatile intracellular delivery technology. Curr Opin Colloid Interface Sci 54:101453. doi:10.1016/j.cocis.2021.101453.
  • Rhodes K, Clark I, Zatcoff M, et al. (2007). Cellular laserfection. Methods Cell Biol 82:309–33. doi:10.1016/S0091-679X(06)82010-8.
  • Saklayen N, Huber M, Madrid M, et al. (2017). Intracellular delivery using nanosecond-laser excitation of large-area plasmonic substrates. ACS Nano 11:3671–80. doi:10.1021/acsnano.6b08162.
  • Santra TS, Wang PC, Tseng FG. (2013). Electroporation based drug delivery and its applications. Adv Micro/Nano Electromechan Syst Fabricat Technol 2013:61–98.
  • Sauvage F, Fraire JC, Remaut K, et al. (2019). Photoablation of human vitreous opacities by light-induced vapor nanobubbles. ACS Nano 13:8401–16. doi:10.1021/acsnano.9b04050.
  • Sauvage F, Nguyen VP, Li Y, et al. (2022). Laser-induced nanobubbles safely ablate vitreous opacities in vivo. Nat Nanotechnol 17:552–9. doi:10.1038/s41565-022-01086-4.
  • Schneckenburger H. (2019). Laser-assisted optoporation of cells and tissues – a mini-review. Biomed Opt Express 10:2883–8. doi:10.1364/BOE.10.002883.
  • Schneckenburger H, Hendinger A, Sailer R, et al. (2002). Laser-assisted optoporation of single cells. J Biomed Opt 7:410–6. doi:10.1117/1.1485758.
  • Sengupta A, Kelly SC, Dwivedi N, et al. (2014). Efficient intracellular delivery of molecules with high cell viability using nanosecond-pulsed laser-activated carbon nanoparticles. ACS Nano 8:2889–99. doi:10.1021/nn500100x.
  • Shaabani E, Sharifiaghdam M, De Keersmaecker H, et al. (2021). Layer by layer assembled chitosan-coated gold nanoparticles for enhanced sirna delivery and silencing. Int J Mol Sci 22:831. doi:10.3390/ijms22020831.
  • Shinde A, Kar S, Nagai M, et al. (2021). Light-induced cellular delivery and analysis. pp 3–30. Singapore: Springer Singapore.
  • Shinde P, Kar S, Loganathan M, et al. (2020). Infrared pulse laser-activated highly efficient intracellular delivery using titanium microdish device. ACS Biomater Sci Eng 6:5645–52. doi:10.1021/acsbiomaterials.0c00785.
  • Shinde P, Kumar A, Dey KK, et al. (2020). Physical approaches for drug delivery: an overview. delivery of drugs: 161–90.
  • Srinivasan R. (1986). Ablation of polymers and biological tissue by ultraviolet lasers. Science 234:559–65. doi:10.1126/science.3764428.
  • Srinivasan R, Leigh WJ. (1982). Ablative photodecomposition: action of far-ultraviolet (193 nm) laser radiation on poly (ethylene terephthalate) films. J Am Chem Soc 104:6784–5. doi:10.1021/ja00388a052.
  • Stevenson DJ, Gunn-Moore FJ, Campbell P, Dholakia K. (2010). Single cell optical transfection. J R Soc Interface 7:863–71. doi:10.1098/rsif.2009.0463.
  • Stewart MP, Langer R, Jensen KF. (2018). Intracellular delivery by membrane disruption: mechanisms, strategies, and concepts. Chem Rev 118:7409–531. doi:10.1021/acs.chemrev.7b00678.
  • Stewart MP, Sharei A, Ding X, et al. (2016). In vitro and ex vivo strategies for intracellular delivery. Nature (London) 538:183–92. doi:10.1038/nature19764.
  • St-Louis LB, Boulais E, Lebrun JJ, Meunier M. (2013). Visible and near infrared resonance plasmonic enhanced nanosecond laser optoporation of cancer cells. Biomed Opt Express 4:490–9. doi:10.1364/BOE.4.000490.
  • Teirlinck E, Fraire JC, Van Acker H, et al. (2019). Laser-induced vapor nanobubbles improve diffusion in biofilms of antimicrobial agents for wound care. Biofilm 1:100004. doi:10.1016/j.bioflm.2019.100004.
  • Teirlinck E, Xiong R, Brans T, et al. (2018). Laser-induced vapour nanobubbles improve drug diffusion and efficiency in bacterial biofilms. Nat Commun 9:4518. doi:10.1038/s41467-018-06884-w.
  • Tirlapur UK, König K. (2002). Targeted transfection by femtosecond laser. nature 418:290–1. doi:10.1038/418290a.
  • Tsukakoshi M, Kurata S, Nomiya Y, et al. (1984). A novel method of DNA transfection by laser microbeam cell surgery. Appl Phys B 35:135–40. doi:10.1007/BF00697702.
  • Van Hoecke L, Raes L, Stremersch S, et al. (2019). Delivery of mixed-lineage kinase domain-like protein by vapor nanobubble photoporation induces necroptotic-like cell death in tumor cells. Int J Mol Sci 20:4254. doi:10.3390/ijms20174254.
  • Varkouhi AK, Scholte M, Storm G, Haisma HJ. (2011). Endosomal escape pathways for delivery of biologicals. J Control Release 151:220–8. doi:10.1016/j.jconrel.2010.11.004.
  • Vogel A, Noack J, Hüttman G, Paltauf G. (2005). Mechanisms of femtosecond laser nanosurgery of cells and tissues. Appl Phys B 81:1015–47. doi:10.1007/s00340-005-2036-6.
  • Wang J, Harizaj A, Wu Y, et al. (2021). Black phosphorus mediated photoporation: a broad absorption nanoplatform for intracellular delivery of macromolecules. Nanoscale 13:17049–56. doi:10.1039/d1nr05461a.
  • Wang M, Zhang Y, Cai C, et al. (2018). Sonoporation-induced cell membrane permeabilization and cytoskeleton disassembly at varied acoustic and microbubble-cell parameters. Sci Rep 8:3885. doi:10.1038/s41598-018-22056-8.
  • Wang S, Fu L, Xin J, et al. (2018). Photoacoustic response induced by nanoparticle-mediated photothermal bubbles beyond the thermal expansion for potential theranostics. J Biomed Opt 23:1–12. doi:10.1117/1.JBO.23.12.125002.
  • Wang S, Fu L, Zhang Y, et al. (2018). Quantitative evaluation and optimization of photothermal bubble generation around overheated nanoparticles excited by pulsed lasers. J Phys Chem C 122:24421–35. doi:10.1021/acs.jpcc.8b07672.
  • Wilson AM, Mazzaferri J, Bergeron E, et al. (2018). In vivo laser-mediated retinal ganglion cell optoporation Using KV1.1 conjugated gold nanoparticles. Nano Lett 18:6981–8. doi:10.1021/acs.nanolett.8b02896.
  • Wu YC, Wu TH, Clemens DL, et al. (2015). Massively parallel delivery of large cargo into mammalian cells with light pulses. Nat Methods 12:439–44. doi:10.1038/nmeth.3357.
  • Wu Z, Yang H, Colosi P. (2010). Effect of genome size on AAV vector packaging. Mol Ther 18:80–6. doi:10.1038/mt.2009.255.
  • Xiong R, Drullion C, Verstraelen P, et al. (2017). Fast spatial-selective delivery into live cells. J Control Release 266:198–204. doi:10.1016/j.jconrel.2017.09.033.
  • Xiong R, Hua D, Van Hoeck J, et al. (2021). Photothermal nanofibres enable safe engineering of therapeutic cells. Nat Nanotechnol 16:1281–91. doi:10.1038/s41565-021-00976-3.
  • Xiong R, Joris F, Liang S, et al. (2016). Cytosolic delivery of nanolabels prevents their asymmetric inheritance and enables extended quantitative in vivo cell imaging. Nano Lett 16:5975–86. doi:10.1021/acs.nanolett.6b01411.
  • Xiong R, Raemdonck K, Peynshaert K, et al. (2014). Comparison of gold nanoparticle mediated photoporation: vapor nanobubbles outperform direct heating for delivering macromolecules in live cells. ACS Nano 8:6288–96. doi:10.1021/nn5017742.
  • Xiong R, Samal SK, Demeester J, et al. (2016). Laser-assisted photoporation: fundamentals, technological advances and applications. Advances in Physics: X 1:596–620. doi:10.1080/23746149.2016.1228476.
  • Xiong R, Vandenbroucke RE, Broos K, et al. (2016). Sizing nanomaterials in bio-fluids by cFRAP enables protein aggregation measurements and diagnosis of bio-barrier permeability. Nat Commun 7:12982. doi:10.1038/ncomms12982.
  • Xu Y, De Keersmaecker H, Braeckmans K, et al. (2020). Targeted nanoparticles towards increased L cell stimulation as a strategy to improve oral peptide delivery in incretin-based diabetes treatment. Biomaterials 255:120209. doi:10.1016/j.biomaterials.2020.120209.
  • Yadav D, Sandeep K, Pandey D, Dutta RK. (2017). Liposomes for drug delivery. J Biotechnol Biomat 7:1000276.
  • Yao C, Qu X, Zhang Z. (2009). Laser-based transfection with conjugated gold nanoparticles. Chin Opt Lett 7:898–900. doi:10.3788/COL20090710.0898.
  • Yao C, Qu X, Zhang Z, et al. (2009). Influence of laser parameters on nanoparticle-induced membrane permeabilization. J Biomed Opt 14:054034. doi:10.1117/1.3253320.
  • Yao C, Rahmanzadeh R, Endl E, et al. (2005). Elevation of plasma membrane permeability by laser irradiation of selectively bound nanoparticles. J Biomed Opt 10:064012. doi:10.1117/1.2137321.
  • Yao C, Rudnitzki F, He Y, et al. (2020). Cancer cell-specific protein delivery by optoporation with laser-irradiated gold nanorods. J Biophotonics 13:e202000017. doi:10.1002/jbio.202000017.
  • Yao C, Rudnitzki F, Hüttmann G, et al. (2017). Important factors for cell-membrane permeabilization by gold nanoparticles activated by nanosecond-laser irradiation. Int J Nanomedicine 12:5659–72. doi:10.2147/IJN.S140620.
  • Yu Z, Li H, Zhong T, et al. (2022). Wavefront shaping: a versatile tool to conquer multiple scattering in multidisciplinary fields. Innovation 3:100292. doi:10.1016/j.xinn.2022.100292.
  • Zhang D, Das DB, Rielly CD. (2014). Potential of microneedle-assisted micro-particle delivery by gene guns: a review. Drug Deliv 21:571–87. doi:10.3109/10717544.2013.864345.