- -

Gold Nanoparticle-Assisted Virus Formation by Means of the Delivery of an Oncolytic Adenovirus Genome

RiuNet: Repositorio Institucional de la Universidad Politécnica de Valencia

Compartir/Enviar a

Citas

Estadísticas

  • Estadisticas de Uso

Gold Nanoparticle-Assisted Virus Formation by Means of the Delivery of an Oncolytic Adenovirus Genome

Mostrar el registro sencillo del ítem

Ficheros en el ítem

dc.contributor.author Sendra, Luis es_ES
dc.contributor.author Miguel, Antonio es_ES
dc.contributor.author Navarro-Plaza, M. Carmen es_ES
dc.contributor.author Herrero, María José es_ES
dc.contributor.author de la Higuera, José es_ES
dc.contributor.author Cháfer-Pericás, Consuelo es_ES
dc.contributor.author Aznar, Elena es_ES
dc.contributor.author Marcos Martínez, María Dolores es_ES
dc.contributor.author Martínez-Máñez, Ramón es_ES
dc.contributor.author Rojas, Luis Alfonso es_ES
dc.contributor.author Alemany, Ramón es_ES
dc.contributor.author Aliño, Salvador F. es_ES
dc.date.accessioned 2021-02-25T04:49:29Z
dc.date.available 2021-02-25T04:49:29Z
dc.date.issued 2020-06 es_ES
dc.identifier.uri http://hdl.handle.net/10251/162367
dc.description.abstract [EN] Oncolytic adenoviruses are a therapeutic alternative to treat cancer based on their ability to replicate selectively in tumor cells. However, their use is limited mainly by the neutralizing antibody (Nab) immune response that prevents repeated dosing. An alternative to facilitate the DNA access to the tumor even in the presence of anti-viral Nabs could be gold nanoparticles able to transfer DNA molecules. However, the ability of these nanoparticles to carry large DNA molecules, such as an oncolytic adenovirus genome, has not been studied. In this work, gold nanoparticles were functionalized with different amounts of polyethylenimine to transfer in a safe and efficient manner a large oncolytic virus genome. Their transfer efficacy and final effect of the oncolytic virus in cancer cells are studied. For each synthesized nanoparticle, (a) DNA loading capacity, (b) complex size, (c) DNA protection ability, (d) transfection efficacy and (e) cytotoxic effect were studied. We observed that small gold nanoparticles (70-80 nm in diameter) protected DNA against nucleases and were able to transfect the ICOVIR-15 oncolytic virus genome encoded in pLR1 plasmid. In the present work, efficient transgene RNA expression, luciferase activity and viral cytopathic effect on cancer cells are reported. These results suggest gold nanoparticles to be an efficient and safe vector for oncolytic adenovirus genome transfer. es_ES
dc.description.sponsorship This research was supported by University of Valencia 'Ayuda a la Investigacion', Asociacion Pablo Ugarte and European Regional Development Fund (VLC-CAMPUS). es_ES
dc.language Inglés es_ES
dc.publisher MDPI AG es_ES
dc.relation.ispartof Nanomaterials es_ES
dc.rights Reconocimiento (by) es_ES
dc.subject Gold nanoparticles es_ES
dc.subject Delivery es_ES
dc.subject Gene therapy es_ES
dc.subject Non-viral vectors es_ES
dc.subject Oncolytic virus es_ES
dc.subject Virotherapy es_ES
dc.subject Cancer es_ES
dc.subject.classification QUIMICA INORGANICA es_ES
dc.subject.classification CIENCIA DE LOS MATERIALES E INGENIERIA METALURGICA es_ES
dc.title Gold Nanoparticle-Assisted Virus Formation by Means of the Delivery of an Oncolytic Adenovirus Genome es_ES
dc.type Artículo es_ES
dc.identifier.doi 10.3390/nano10061183 es_ES
dc.rights.accessRights Abierto es_ES
dc.contributor.affiliation Universitat Politècnica de València. Instituto de Reconocimiento Molecular y Desarrollo Tecnológico - Institut de Reconeixement Molecular i Desenvolupament Tecnològic es_ES
dc.contributor.affiliation Universitat Politècnica de València. Departamento de Química - Departament de Química es_ES
dc.description.bibliographicCitation Sendra, L.; Miguel, A.; Navarro-Plaza, MC.; Herrero, MJ.; De La Higuera, J.; Cháfer-Pericás, C.; Aznar, E.... (2020). Gold Nanoparticle-Assisted Virus Formation by Means of the Delivery of an Oncolytic Adenovirus Genome. Nanomaterials. 10(6):1-16. https://doi.org/10.3390/nano10061183 es_ES
dc.description.accrualMethod S es_ES
dc.relation.publisherversion https://doi.org/10.3390/nano10061183 es_ES
dc.description.upvformatpinicio 1 es_ES
dc.description.upvformatpfin 16 es_ES
dc.type.version info:eu-repo/semantics/publishedVersion es_ES
dc.description.volume 10 es_ES
dc.description.issue 6 es_ES
dc.identifier.eissn 2079-4991 es_ES
dc.identifier.pmid 32560474 es_ES
dc.identifier.pmcid PMC7353451 es_ES
dc.relation.pasarela S\418274 es_ES
dc.contributor.funder Asociación Pablo Ugarte es_ES
dc.contributor.funder Universitat de València es_ES
dc.contributor.funder European Regional Development Fund es_ES
dc.description.references Cebrián, V., Martín-Saavedra, F., Yagüe, C., Arruebo, M., Santamaría, J., & Vilaboa, N. (2011). Size-dependent transfection efficiency of PEI-coated gold nanoparticles. Acta Biomaterialia, 7(10), 3645-3655. doi:10.1016/j.actbio.2011.06.018 es_ES
dc.description.references Shukla, R., Bansal, V., Chaudhary, M., Basu, A., Bhonde, R. R., & Sastry, M. (2005). Biocompatibility of Gold Nanoparticles and Their Endocytotic Fate Inside the Cellular Compartment: A Microscopic Overview. Langmuir, 21(23), 10644-10654. doi:10.1021/la0513712 es_ES
dc.description.references Niidome, T., Nakashima, K., Takahashi, H., & Niidome, Y. (2004). Preparation of primary amine-modified gold nanoparticles and their transfection ability into cultivated cellsElectronic Supplementary Information (ESI) available: A TEM image of the complex at a w/w ratio of 11. See http://www.rsc.org/suppdata/cc/b4/b406189f/. Chemical Communications, (17), 1978. doi:10.1039/b406189f es_ES
dc.description.references Sandhu, K. K., McIntosh, C. M., Simard, J. M., Smith, S. W., & Rotello, V. M. (2001). Gold Nanoparticle-Mediated Transfection of Mammalian Cells. Bioconjugate Chemistry, 13(1), 3-6. doi:10.1021/bc015545c es_ES
dc.description.references Thomas, M., & Klibanov, A. M. (2003). Conjugation to gold nanoparticles enhances polyethylenimine’s transfer of plasmid DNA into mammalian cells. Proceedings of the National Academy of Sciences, 100(16), 9138-9143. doi:10.1073/pnas.1233634100 es_ES
dc.description.references Noh, S. M., Kim, W.-K., Kim, S. J., Kim, J. M., Baek, K.-H., & Oh, Y.-K. (2007). Enhanced cellular delivery and transfection efficiency of plasmid DNA using positively charged biocompatible colloidal gold nanoparticles. Biochimica et Biophysica Acta (BBA) - General Subjects, 1770(5), 747-752. doi:10.1016/j.bbagen.2007.01.012 es_ES
dc.description.references Chan, T. G., Morse, S. V., Copping, M. J., Choi, J. J., & Vilar, R. (2018). Targeted Delivery of DNA-Au Nanoparticles across the Blood-Brain Barrier Using Focused Ultrasound. ChemMedChem, 13(13), 1311-1314. doi:10.1002/cmdc.201800262 es_ES
dc.description.references Mbatha, L. S., & Singh, M. (2019). Starburst Poly(amidoamine) Dendrimer Grafted Gold Nanoparticles as a Scaffold for Folic Acid-Targeted Plasmid DNA Delivery In Vitro. Journal of Nanoscience and Nanotechnology, 19(4), 1959-1970. doi:10.1166/jnn.2019.15798 es_ES
dc.description.references Cobley, C. M., Chen, J., Cho, E. C., Wang, L. V., & Xia, Y. (2011). Gold nanostructures: a class of multifunctional materials for biomedical applications. Chem. Soc. Rev., 40(1), 44-56. doi:10.1039/b821763g es_ES
dc.description.references Cho, E. C., Au, L., Zhang, Q., & Xia, Y. (2010). The Effects of Size, Shape, and Surface Functional Group of Gold Nanostructures on Their Adsorption and Internalization by Cells. Small, 6(4), 517-522. doi:10.1002/smll.200901622 es_ES
dc.description.references Pissuwan, D., Niidome, T., & Cortie, M. B. (2011). The forthcoming applications of gold nanoparticles in drug and gene delivery systems. Journal of Controlled Release, 149(1), 65-71. doi:10.1016/j.jconrel.2009.12.006 es_ES
dc.description.references Rosi, N. L., Giljohann, D. A., Thaxton, C. S., Lytton-Jean, A. K. R., Han, M. S., & Mirkin, C. A. (2006). Oligonucleotide-Modified Gold Nanoparticles for Intracellular Gene Regulation. Science, 312(5776), 1027-1030. doi:10.1126/science.1125559 es_ES
dc.description.references Ghosh, P. S., Kim, C.-K., Han, G., Forbes, N. S., & Rotello, V. M. (2008). Efficient Gene Delivery Vectors by Tuning the Surface Charge Density of Amino Acid-Functionalized Gold Nanoparticles. ACS Nano, 2(11), 2213-2218. doi:10.1021/nn800507t es_ES
dc.description.references Massich, M. D., Giljohann, D. A., Seferos, D. S., Ludlow, L. E., Horvath, C. M., & Mirkin, C. A. (2009). Regulating Immune Response Using Polyvalent Nucleic Acid−Gold Nanoparticle Conjugates. Molecular Pharmaceutics, 6(6), 1934-1940. doi:10.1021/mp900172m es_ES
dc.description.references Ryou, S.-M., Kim, S., Jang, H. H., Kim, J.-H., Yeom, J.-H., Eom, M. S., … Lee, K. (2010). Delivery of shRNA using gold nanoparticle–DNA oligonucleotide conjugates as a universal carrier. Biochemical and Biophysical Research Communications, 398(3), 542-546. doi:10.1016/j.bbrc.2010.06.115 es_ES
dc.description.references Stobiecka, M., & Hepel, M. (2011). Double-shell gold nanoparticle-based DNA-carriers with poly-l-lysine binding surface. Biomaterials, 32(12), 3312-3321. doi:10.1016/j.biomaterials.2010.12.064 es_ES
dc.description.references Sharma, A., Tandon, A., Tovey, J. C. K., Gupta, R., Robertson, J. D., Fortune, J. A., … Mohan, R. R. (2011). Polyethylenimine-conjugated gold nanoparticles: Gene transfer potential and low toxicity in the cornea. Nanomedicine: Nanotechnology, Biology and Medicine, 7(4), 505-513. doi:10.1016/j.nano.2011.01.006 es_ES
dc.description.references Yan, X., Blacklock, J., Li, J., & Möhwald, H. (2011). One-Pot Synthesis of Polypeptide–Gold Nanoconjugates for in Vitro Gene Transfection. ACS Nano, 6(1), 111-117. doi:10.1021/nn202939s es_ES
dc.description.references Shan, Y., Luo, T., Peng, C., Sheng, R., Cao, A., Cao, X., … Shi, X. (2012). Gene delivery using dendrimer-entrapped gold nanoparticles as nonviral vectors. Biomaterials, 33(10), 3025-3035. doi:10.1016/j.biomaterials.2011.12.045 es_ES
dc.description.references Trigueros, Domènech, Toulis, & Marfany. (2019). In Vitro Gene Delivery in Retinal Pigment Epithelium Cells by Plasmid DNA-Wrapped Gold Nanoparticles. Genes, 10(4), 289. doi:10.3390/genes10040289 es_ES
dc.description.references Munsell, E. V., Fang, B., & Sullivan, M. O. (2018). Histone-Mimetic Gold Nanoparticles as Versatile Scaffolds for Gene Transfer and Chromatin Analysis. Bioconjugate Chemistry, 29(11), 3691-3704. doi:10.1021/acs.bioconjchem.8b00611 es_ES
dc.description.references Dhanya, G. R., Caroline, D. S., Rekha, M. R., & Sreenivasan, K. (2018). Histidine and arginine conjugated starch-PEI and its corresponding gold nanoparticles for gene delivery. International Journal of Biological Macromolecules, 120, 999-1008. doi:10.1016/j.ijbiomac.2018.08.142 es_ES
dc.description.references Hersey, P., & Gallagher, S. (2013). Intralesional immunotherapy for melanoma. Journal of Surgical Oncology, 109(4), 320-326. doi:10.1002/jso.23494 es_ES
dc.description.references Mastrangelo, M. J., Maguire, H. C., Eisenlohr, L. C., Laughlin, C. E., Monken, C. E., McCue, P. A., … Lattime, E. C. (1999). Intratumoral recombinant GM-CSF-encoding virus as gene therapy in patients with cutaneous melanoma. Cancer Gene Therapy, 6(5), 409-422. doi:10.1038/sj.cgt.7700066 es_ES
dc.description.references Senzer, N. N., Kaufman, H. L., Amatruda, T., Nemunaitis, M., Reid, T., Daniels, G., … Nemunaitis, J. J. (2009). Phase II Clinical Trial of a Granulocyte-Macrophage Colony-Stimulating Factor–Encoding, Second-Generation Oncolytic Herpesvirus in Patients With Unresectable Metastatic Melanoma. Journal of Clinical Oncology, 27(34), 5763-5771. doi:10.1200/jco.2009.24.3675 es_ES
dc.description.references Goins, W. F., Huang, S., Cohen, J. B., & Glorioso, J. C. (2014). Engineering HSV-1 Vectors for Gene Therapy. Herpes Simplex Virus, 63-79. doi:10.1007/978-1-4939-0428-0_5 es_ES
dc.description.references Dummer, R., Rochlitz, C., Velu, T., Acres, B., Limacher, J.-M., Bleuzen, P., … Urosevic, M. (2008). Intralesional Adenovirus-mediated Interleukin-2 Gene Transfer for Advanced Solid Cancers and Melanoma. Molecular Therapy, 16(5), 985-994. doi:10.1038/mt.2008.32 es_ES
dc.description.references GUPTA, P., SU, Z., LEBEDEVA, I., SARKAR, D., SAUANE, M., EMDAD, L., … DENT, P. (2006). mda-7/IL-24: Multifunctional cancer-specific apoptosis-inducing cytokine. Pharmacology & Therapeutics, 111(3), 596-628. doi:10.1016/j.pharmthera.2005.11.005 es_ES
dc.description.references Abbink, P., Lemckert, A. A. C., Ewald, B. A., Lynch, D. M., Denholtz, M., Smits, S., … Barouch, D. H. (2007). Comparative Seroprevalence and Immunogenicity of Six Rare Serotype Recombinant Adenovirus Vaccine Vectors from Subgroups B and D. Journal of Virology, 81(9), 4654-4663. doi:10.1128/jvi.02696-06 es_ES
dc.description.references Mast, T. C., Kierstead, L., Gupta, S. B., Nikas, A. A., Kallas, E. G., Novitsky, V., … Shiver, J. W. (2010). International epidemiology of human pre-existing adenovirus (Ad) type-5, type-6, type-26 and type-36 neutralizing antibodies: Correlates of high Ad5 titers and implications for potential HIV vaccine trials. Vaccine, 28(4), 950-957. doi:10.1016/j.vaccine.2009.10.145 es_ES
dc.description.references Barouch, D. H., Kik, S. V., Weverling, G. J., Dilan, R., King, S. L., Maxfield, L. F., … Goudsmit, J. (2011). International seroepidemiology of adenovirus serotypes 5, 26, 35, and 48 in pediatric and adult populations. Vaccine, 29(32), 5203-5209. doi:10.1016/j.vaccine.2011.05.025 es_ES
dc.description.references Na, Y., Nam, J.-P., Hong, J., Oh, E., Shin, H. C., Kim, H. S., … Yun, C.-O. (2019). Systemic administration of human mesenchymal stromal cells infected with polymer-coated oncolytic adenovirus induces efficient pancreatic tumor homing and infiltration. Journal of Controlled Release, 305, 75-88. doi:10.1016/j.jconrel.2019.04.040 es_ES
dc.description.references Kasala, D., Yoon, A.-R., Hong, J., Kim, S. W., & Yun, C.-O. (2016). Evolving lessons on nanomaterial-coated viral vectors for local and systemic gene therapy. Nanomedicine, 11(13), 1689-1713. doi:10.2217/nnm-2016-0060 es_ES
dc.description.references Kwon, O.-J., Kang, E., Kim, S., & Yun, C.-O. (2011). Viral genome DNA/lipoplexes elicit in situ oncolytic viral replication and potent antitumor efficacy via systemic delivery. Journal of Controlled Release, 155(2), 317-325. doi:10.1016/j.jconrel.2011.06.014 es_ES
dc.description.references YOSHIHARA, C., HAMADA, K., KURODA, M., & KOYAMA, Y. (2011). Oncolytic plasmid: A novel strategy for tumor immuno-gene therapy. Oncology Letters, 3(2), 387-390. doi:10.3892/ol.2011.467 es_ES
dc.description.references Rojas, J. J., Guedan, S., Searle, P. F., Martinez-Quintanilla, J., Gil-Hoyos, R., Alcayaga-Miranda, F., … Alemany, R. (2010). Minimal RB-responsive E1A Promoter Modification to Attain Potency, Selectivity, and Transgene-arming Capacity in Oncolytic Adenoviruses. Molecular Therapy, 18(11), 1960-1971. doi:10.1038/mt.2010.173 es_ES
dc.description.references Rincón, E., Cejalvo, T., Kanojia, D., Alfranca, A., Rodríguez-Milla, M. Á., Hoyos, R. A. G., … García-Castro, J. (2017). Mesenchymal stem cell carriers enhance antitumor efficacy of oncolytic adenoviruses in an immunocompetent mouse model. Oncotarget, 8(28), 45415-45431. doi:10.18632/oncotarget.17557 es_ES
dc.description.references Carette, J. E., Graat, H. C. A., Schagen, F. H. E., Abou El Hassan, M. A. I., Gerritsen, W. R., & van Beusechem, V. W. (2005). Replication-dependent transgene expression from a conditionally replicating adenovirus via alternative splicing to a heterologous splice-acceptor site. The Journal of Gene Medicine, 7(8), 1053-1062. doi:10.1002/jgm.754 es_ES
dc.description.references Stanton, R. J., McSharry, B. P., Armstrong, M., Tomasec, P., & Wilkinson, G. W. G. (2008). Re-engineering adenovirus vector systems to enable high-throughput analyses of gene function. BioTechniques, 45(6), 659-668. doi:10.2144/000112993 es_ES
dc.description.references Bayo-Puxan, N., Cascallo, M., Gros, A., Huch, M., Fillat, C., & Alemany, R. (2006). Role of the putative heparan sulfate glycosaminoglycan-binding site of the adenovirus type 5 fiber shaft on liver detargeting and knob-mediated retargeting. Journal of General Virology, 87(9), 2487-2495. doi:10.1099/vir.0.81889-0 es_ES
dc.description.references Brust, M., Fink, J., Bethell, D., Schiffrin, D. J., & Kiely, C. (1995). Synthesis and reactions of functionalised gold nanoparticles. Journal of the Chemical Society, Chemical Communications, (16), 1655. doi:10.1039/c39950001655 es_ES
dc.description.references Guillem, V. M., Tormo, M., Revert, F., Benet, I., García-Conde, J., Crespo, A., & Aliño, S. F. (2002). Polyethyleneimine-based immunopolyplex for targeted gene transfer in human lymphoma celllines. The Journal of Gene Medicine, 4(2), 170-182. doi:10.1002/jgm.228 es_ES
dc.description.references Stuchbury, T., Shipton, M., Norris, R., Malthouse, J. P. G., Brocklehurst, K., Herbert, J. A. L., & Suschitzky, H. (1975). A reporter group delivery system with both absolute and selective specificity for thiol groups and an improved fluorescent probe containing the 7-nitrobenzo-2-oxa-1,3-diazole moiety. Biochemical Journal, 151(2), 417-432. doi:10.1042/bj1510417 es_ES
dc.description.references Moret, I., Esteban Peris, J., Guillem, V. M., Benet, M., Revert, F., Dası́, F., … Aliño, S. F. (2001). Stability of PEI–DNA and DOTAP–DNA complexes: effect of alkaline pH, heparin and serum. Journal of Controlled Release, 76(1-2), 169-181. doi:10.1016/s0168-3659(01)00415-1 es_ES
dc.description.references Lisitsyna, E. S., Lygo, O. N., Durandin, N. A., Dement’eva, O. V., Rudoi, V. M., & Kuzmin, V. A. (2012). Superquenching of SYBRGreen dye fluorescence in complex with DNA by gold nanoparticles. High Energy Chemistry, 46(6), 363-367. doi:10.1134/s0018143912060057 es_ES
dc.description.references Venkiteswaran, S., Thomas, T., & Thomas, T. J. (2016). Selectivity of polyethyleneimines on DNA nanoparticle preparation and gene transport. ChemistrySelect, 1(6), 1144-1150. doi:10.1002/slct.201600026 es_ES
dc.description.references Taranejoo, S., Liu, J., Verma, P., & Hourigan, K. (2015). A review of the developments of characteristics of PEI derivatives for gene delivery applications. Journal of Applied Polymer Science, 132(25), n/a-n/a. doi:10.1002/app.42096 es_ES
dc.description.references Del Papa, J., & Parks, R. (2017). Adenoviral Vectors Armed with Cell Fusion-Inducing Proteins as Anti-Cancer Agents. Viruses, 9(1), 13. doi:10.3390/v9010013 es_ES
dc.description.references Kazemi Oskuee, R., Dabbaghi, M., Gholami, L., Taheri-Bojd, S., Balali-Mood, M., Mousavi, S. H., & Malaekeh-Nikouei, B. (2018). Investigating the influence of polyplex size on toxicity properties of polyethylenimine mediated gene delivery. Life Sciences, 197, 101-108. doi:10.1016/j.lfs.2018.02.008 es_ES
dc.description.references Thomas, T. J., Tajmir-Riahi, H.-A., & Pillai, C. K. S. (2019). Biodegradable Polymers for Gene Delivery. Molecules, 24(20), 3744. doi:10.3390/molecules24203744 es_ES
dc.description.references Miciak, J. J., Hirshberg, J., & Bunz, F. (2018). Seamless assembly of recombinant adenoviral genomes from high-copy plasmids. PLOS ONE, 13(6), e0199563. doi:10.1371/journal.pone.0199563 es_ES


Este ítem aparece en la(s) siguiente(s) colección(ones)

Mostrar el registro sencillo del ítem