167
Views
7
CrossRef citations to date
0
Altmetric
Original Research

Uptake and Intracellular Trafficking Studies of Multiple Dye-Doped Core-Shell Silica Nanoparticles in Lymphoid and Myeloid Cells

ORCID Icon, , , , , , , , , , & show all
Pages 29-48 | Published online: 08 Mar 2021

References

  • McNeil SE. Nanotechnology for the biologist. J Leukoc Biol. 2005;78(3):585–594. doi:10.1189/jlb.0205074
  • Nagamune T. Biomolecular engineering for nanobio/bionanotechnology. Nano Converg. 2017;4(1):9. doi:10.1186/s40580-017-0103-4
  • Mahapatro A, Singh DK. Biodegradable nanoparticles are excellent vehicle for site directed in-vivo delivery of drugs and vaccines. J Nanobiotechnology. 2011;9(1):55. doi:10.1186/1477-3155-9-55
  • Foroozandeh P, Aziz AA. Insight into cellular uptake and intracellular trafficking of nanoparticles. Nanoscale Res Lett. 2018;13(1):1–12. doi:10.1186/s11671-018-2728-6
  • Saraiva C, Praça C, Ferreira R, Santos T, Ferreira L, Bernardino L. Nanoparticle-mediated brain drug delivery: overcoming blood-brain barrier to treat neurodegenerative diseases. J Control Release. 2016;235:34–47. doi:10.1016/j.jconrel.2016.05.044
  • Luo Y, Yang H, Zhou YF, Hu B. Dual and multi-targeted nanoparticles for site-specific brain drug delivery. J Control Release. 2020;317:195–215. doi:10.1016/j.jconrel.2019.11.037
  • Dos santos T, Varela J, Lynch I, Salvati A, Dawson KA, Schnur JM. Effects of transport inhibitors on the cellular uptake of carboxylated polystyrene nanoparticles in different cell lines. Schnur JM, ed. PLoS One. 2011;6(9):e24438. doi:10.1371/journal.pone.0024438
  • Peñaloza JP, Márquez-Miranda V, Cabaña-Brunod M, et al. Intracellular trafficking and cellular uptake mechanism of PHBV nanoparticles for targeted delivery in epithelial cell lines. J Nanobiotechnology. 2017;15(1):1. doi:10.1186/s12951-016-0241-6
  • Liu X, Wu F, Tian Y, et al. Size dependent cellular uptake of rod-like bionanoparticles with different aspect ratios. Sci Rep. 2016;6(1):1–10. doi:10.1038/srep24567
  • Donahue ND, Acar H, Wilhelm S. Concepts of nanoparticle cellular uptake, intracellular trafficking, and kinetics in nanomedicine. Adv Drug Deliv Rev. 2019;143:68–96. doi:10.1016/j.addr.2019.04.008
  • Ding L, Lian Y, Lin Z, Zhang Z, Wang X. Long-term quantitatively imaging intracellular chloride concentration using a core-/shell-structured nanosensor and time-domain dual-lifetime referencing method. ACS Sens. 2020;5(12):3971–3978. doi:10.1021/acssensors.0c01671
  • Panariti A, Miserocchi G, Rivolta I. The effect of nanoparticle uptake on cellular behavior: disrupting or enabling functions? Nanotechnol Sci Appl. 2012;5(1):87–100. doi:10.2147/NSA.S25515
  • Kou L, Sun J, Zhai Y, He Z. The endocytosis and intracellular fate of nanomedicines: implication for rational design. Asian J Pharm Sci. 2013;8(1):1–10. doi:10.1016/j.ajps.2013.07.001
  • Oh N, Park JH. Endocytosis and exocytosis of nanoparticles in mammalian cells. Int J Nanomedicine. 2014;9(SUPPL.1):51–63. doi:10.2147/IJN.S26592
  • Wang Y, Chen L, Liu P. Biocompatible triplex Ag@SiO 2@mTiO 2 core-shell nanoparticles for simultaneous fluorescence-SERS bimodal imaging and drug delivery. Chem Eur J. 2012;18(19):5935–5943. doi:10.1002/chem.201103571
  • Wang F, Xu G, Gu X, et al. Realizing highly chemoselective detection of H2S in vitro and in vivo with fluorescent probes inside core-shell silica nanoparticles. Biomaterials. 2018;159:82–90. doi:10.1016/j.biomaterials.2018.01.009
  • Chu Z, Huang Y, Tao Q, Li Q. Cellular uptake, evolution, and excretion of silica nanoparticles in human cells. Nanoscale. 2011;3(8):3291–3299. doi:10.1039/c1nr10499c
  • Blechinger J, Bauer AT, Torrano AA, Gorzelanny C, Bräuchle C, Schneider SW. Uptake kinetics and nanotoxicity of silica nanoparticles are cell type dependent. Small. 2013;9(23):3970–3980. doi:10.1002/smll.201301004
  • Sriramulu D, Reed EL, Annamalai M, Venkatesan TV, Valiyaveettil S. Synthesis and characterization of superhydrophobic, self-cleaning NIR-reflective silica nanoparticles. Sci Rep. 2016;6(1):1–10. doi:10.1038/srep35993
  • Hsiao I-L, Gramatke AM, Joksimovic R, Sokolowski M, Gradzielski M, Haase A. Size and cell type dependent uptake of silica nanoparticles. J Nanomed Nanotechnol. 2014;05:06. doi:10.4172/2157-7439.1000248
  • Pellegrino C, Volpe A, Juris R, et al. Multiple dye doped core-shell silica nanoparticles: outstanding stability and signal intensity exploiting FRET phenomenon for biomedical applications. J Nanomater Mol Nanotechnol. 2018;s6. doi:10.4172/2324-8777.s6-003
  • Zhang X, Chen F, Turker MZ, et al. Targeted melanoma radiotherapy using ultrasmall 177Lu-labeled α-melanocyte stimulating hormone-functionalized core-shell silica nanoparticles. Biomaterials. 2020;241(August 2019):119858. doi:10.1016/j.biomaterials.2020.119858
  • Jokerst JV, Lobovkina T, Zare RN, Gambhir SS. Nanoparticle PEGylation for imaging and therapy. Nanomedicine. 2011;6(4):715–728. doi:10.2217/nnm.11.19
  • Zucker RM, Ortenzio J, Degn LL, Lerner JM, Boyes WK. Biophysical comparison of four silver nanoparticles coatings using microscopy, hyperspectral imaging and flow cytometry. Zelikin AN, ed. PLoS One. 2019;14(7):e0219078. doi:10.1371/journal.pone.0219078
  • De La Cruz GG, Rodríguez-Fragoso P, Reyes-Esparza J, Rodríguez-López A, Gómez-Cansino R, Rodriguez-Fragoso L. Interaction of nanoparticles with blood components and associated pathophysiological effects. In: Gomes AC, Sarria MP, editors. Unraveling the Safety Profile of Nanoscale Particles and Materials – from Biomedical to Environmental Applications. InTechOpen; 2018:37–59. doi10.5772/intechopen.69386
  • Johnson S, Nguyen V, Coder D. Assessment of cell viability. Curr Protoc Cytom. 2013;64(1):9.2.1–9.2.26. doi:10.1002/0471142956.cy0902s64
  • Chazotte B. Labeling lysosomes in live cells with lysotracker. Cold Spring Harb Protoc. 2011;6:2. doi:10.1101/pdb.prot5571
  • Canonico B, Cesarini E, Montanari M, et al. Rapamycin re-directs lysosome network, stimulates ER-remodeling, involving membrane CD317 and affecting exocytosis, in campylobacter jejuni-lysate-infected U937 cells. Int J Mol Sci. 2020;21(6):2207. doi:10.3390/ijms21062207
  • Vázquez CL, Colombo MI. Assays to assess autophagy induction and fusion of autophagic vacuoles with a degradative compartment, using monodansylcadaverine (MDC) and DQ-BSA. Methods Enzymol. 2009. doi:10.1016/S0076-6879(08)03606-9
  • Canonico B, Cesarini E, Salucci S, et al. Defective autophagy, mitochondrial clearance and lipophagy in niemann-pick type B lymphocytes. PLoS One. 2016;11(10):e0165780. doi:10.1371/journal.pone.0165780
  • Eruslanov E, Kusmartsev S. Identification of ROS using oxidized DCFDA and flow-cytometry. Methods Mol Biol. 2010;594:57–72. doi:10.1007/978-1-60761-411-1_4
  • Bolte S, Cordelières FP. A guided tour into subcellular colocalization analysis in light microscopy. J Microsc. 2006;224(3):213–232. doi:10.1111/j.1365-2818.2006.01706.x
  • Luchetti F, Canonico B, Arcangeletti M, et al. Fas signalling promotes intercellular communication in T cells. Badley AD, ed. PLoS One. 2012;7(4):e35766. doi:10.1371/journal.pone.0035766
  • van Blaaderen A, Vrij A. Synthesis and characterization of monodisperse colloidal organo-silica spheres. J Colloid Interface Sci. 1993;156(1):1–18. doi:10.1006/jcis.1993.1073
  • Filipe V, Hawe A, Jiskoot W. Critical evaluation of nanoparticle tracking analysis (NTA) by NanoSight for the measurement of nanoparticles and protein aggregates. Pharm Res. 2010;27(5):796–810. doi:10.1007/s11095-010-0073-2
  • Morgan JJ, Wiley J. Aquatic Chemistry Chemical Equilibria and Rates in Natural Waters. A Wiley-Interscience Publication; 1996.
  • Baalousha M, Motelica-Heino M, Le Coustumer P. Conformation and size of humic substances: effects of major cation concentration and type, pH, salinity, and residence time. Colloids Surf A Physicochem Eng Asp. 2006;272(1–2):48–55. doi:10.1016/j.colsurfa.2005.07.010
  • Lead JR, Wilkinson KJ. Aquatic colloids and nanoparticles: current knowledge and future trends. Environ Chem. 2006;3(3):159–171. doi:10.1071/EN06025
  • Nowack B, Bucheli TD. Occurrence, behavior and effects of nanoparticles in the environment. Environ Pollut. 2007;150(1):5–22. doi:10.1016/j.envpol.2007.06.006
  • Boxall ABA, Tiede K, Chaudhry Q. Engineered nanomaterials in soils and water: how do they behave and could they pose a risk to human health? Nanomedicine. 2007;2(6):919–927. doi:10.2217/17435889.2.6.919
  • Christian P, Von Der Kammer F, Baalousha M, Hofmann T. Nanoparticles: structure, properties, preparation and behaviour in environmental media. Ecotoxicology. 2008;17(5):326–343. doi:10.1007/s10646-008-0213-1
  • Spooner RA, Smith DC, Easton AJ, Roberts LM, Lord JM. Retrograde transport pathways utilised by viruses and protein toxins. Virol J. 2006;3(1):26. doi:10.1186/1743-422X-3-26
  • Kumar S, Aswal VK, Callow P. PH-dependent interaction and resultant structures of silica nanoparticles and lysozyme protein. Langmuir. 2014;30(6):1588–1598. doi:10.1021/la403896h
  • Casey JR, Grinstein S, Orlowski J. Sensors and regulators of intracellular pH. Nat Rev Mol Cell Biol. 2010;11(1):50–61. doi:10.1038/nrm2820
  • Suzuki H, Toyooka T, Ibuki Y. Simple and easy method to evaluate uptake potential of nanoparticles in mammalian cells using a flow cytometric light scatter analysis. Environ Sci Technol. 2007;41(8):3018–3024. doi:10.1021/es0625632
  • Busch W, Bastian S, Trahorsch U, et al. Internalisation of engineered nanoparticles into mammalian cells in vitro: influence of cell type and particle properties. J Nanoparticle Res. 2011;13(1):293–310. doi:10.1007/s11051-010-0030-3
  • Iacopetta BJ, Morgan EH. The Kinetics of Transferrin Endocytosis and Iron Uptake from Transferrin in Rabbit Reticulocytes. J Biol Chem.1983;258(15):9108-9115.
  • Saraste J, Palade GE, Farquhar MG. Temperature-sensitive steps in the transport of secretory proteins through the golgi complex in exocrine pancreatic cells. Proc Natl Acad Sci U S A. 1986;83(17):6425–6429. doi:10.1073/pnas.83.17.6425
  • Zeng X, Zhang Y, Nyström AM. Endocytic uptake and intracellular trafficking of bis-MPA-based hyperbranched copolymer micelles in breast cancer cells. Biomacromolecules. 2012;13(11):3814–3822. doi:10.1021/bm301281k
  • Faklaris O, Joshi V, Irinopoulou T, et al. Photoluminescent diamond nanoparticles for cell labeling: study of the uptake mechanism in mammalian cells. ACS Nano. 2009;3(12):3955–3962. doi:10.1021/nn901014j
  • Bozavikov P, Rajshankar D, Lee W, McCulloch CA. Particle size influences fibronectin internalization and degradation by fibroblasts. Exp Cell Res. 2014;328(1):172–185. doi:10.1016/j.yexcr.2014.06.018
  • Fiorentino I, Gualtieri R, Barbato V, et al. Energy independent uptake and release of polystyrene nanoparticles in primary mammalian cell cultures. Exp Cell Res. 2015;330(2):240–247. doi:10.1016/j.yexcr.2014.09.017
  • Francia V, Reker-Smit C, Boel G, Salvati A. Limits and challenges in using transport inhibitors to characterize how nano-sized drug carriers enter cells. Nanomedicine. 2019;14(12):1533–1549. doi:10.2217/nnm-2018-0446
  • Vercauteren D, Vandenbroucke RE, Jones AT, et al. The use of inhibitors to study endocytic pathways of gene carriers: optimization and pitfalls. Mol Ther. 2010;18(3):561–569. doi:10.1038/mt.2009.281
  • Iversen TG, Skotland T, Sandvig K. Endocytosis and intracellular transport of nanoparticles: present knowledge and need for future studies. Nano Today. 2011;6(2):176–185. doi:10.1016/j.nantod.2011.02.003
  • Lehman SE, Morris AS, Mueller PS, Salem AK, Grassian VH, Larsen SC. Silica nanoparticle-generated ROS as a predictor of cellular toxicity: mechanistic insights and safety by design. Environ Sci Nano. 2016;3(1):56–66. doi:10.1039/c5en00179j
  • Liu C-G, Han Y-H, Kankala RK, Wang S-B, Chen A-Z. Subcellular performance of nanoparticles in cancer therapy. Int J Nanomedicine. 2020;15:675–704. doi:10.2147/IJN.S226186
  • Cao Y, Long J, Liu L, et al. A review of endoplasmic reticulum (ER) stress and nanoparticle (NP) exposure. Life Sci. 2017;186:33–42. doi:10.1016/j.lfs.2017.08.003
  • Cooper GM. Lysosomes. Sinauer Associates; 2000. Available from: https://www.ncbi.nlm.nih.gov/books/NBK9953/. Accessed July 13, 2020.
  • Pathak RK, Kolishetti N, Dhar S. Targeted nanoparticles in mitochondrial medicine. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2015;7(3):315–329. doi:10.1002/wnan.1305
  • Brand MD. Mitochondrial generation of superoxide and hydrogen peroxide as the source of mitochondrial redox signaling. Free Radic Biol Med. 2016;100:14–31. doi:10.1016/j.freeradbiomed.2016.04.001
  • Hao N, Yang H, Li L, Li L, Tang F. The shape effect of mesoporous silica nanoparticles on intracellular reactive oxygen species in A375 cells. New J Chem. 2014;38(9):4258–4266. doi:10.1039/c4nj00736k
  • Ortega FG, Roefs MT, de Miguel Perez D, et al. Interfering with endolysosomal trafficking enhances release of bioactive exosomes. Nanomedicine. 2019;20:102014. doi:10.1016/j.nano.2019.102014
  • Pfaller T, Colognato R, Nelissen I, et al. The suitability of different cellular in vitro immunotoxicity and genotoxicity methods for the analysis of nanoparticle-induced events. Nanotoxicology. 2010;4(1):52–72. doi:10.3109/17435390903374001
  • Lai L, Alaverdi N, Maltais L, Morse HC III. Mouse cell surface antigens: nomenclature and immunophenotyping. J Immunol. 1998;160(8):3861–3868.
  • Oostingh GJ, Casals E, Italiani P, et al. Problems and challenges in the development and validation of human cell-based assays to determine nanoparticle-induced immunomodulatory effects. Part Fibre Toxicol. 2011;8(1):8. doi:10.1186/1743-8977-8-8
  • Wu Q, Jin R, Feng T, et al. Iron oxide nanoparticles and induced autophagy in human monocytes. Int J Nanomedicine. 2017;12:3993–4005. doi:10.2147/IJN.S135189
  • Sakhtianchi R, Minchin RF, Lee KB, Alkilany AM, Serpooshan V, Mahmoudi M. Exocytosis of nanoparticles from cells: role in cellular retention and toxicity. Adv Colloid Interface Sci. 2013;201–202:18–29. doi:10.1016/j.cis.2013.10.013
  • Canonico B, Luchetti F, Arcangeletti M, Guescini M, Degli Esposti M, Papa S. Flow cytometric analyses disclose intercellular communications in FasL-stimulated T cells: results and trouble shooting. Cytom Part A. 2012;81 A(1):5–8. doi:10.1002/cyto.a.21151
  • Shelke GV, Lässer C, Gho YS, Lötvall J. Importance of exosome depletion protocols to eliminate functional and RNA-containing extracellular vesicles from fetal bovine serum. J Extracell Vesicles. 2014;3(1):24783. doi:10.3402/jev.v3.24783
  • Al-Rawi M, Diabaté S, Weiss C. Uptake and intracellular localization of submicron and nano-sized SiO 2 particles in HeLa cells. Arch Toxicol. 2011;85(7):813–826. doi:10.1007/s00204-010-0642-5
  • Auten RL, Davis JM. Oxygen toxicity and reactive oxygen species: the devil is in the details. Pediatr Res. 2009;66(2):121–127. doi:10.1203/PDR.0b013e3181a9eafb