Cargando…

Nanotechnological engineering of extracellular vesicles for the development of actively targeted hybrid nanodevices

BACKGROUND: We propose an efficient method to modify B-cell derived EVs by loading them with a nanotherapeutic stimuli-responsive cargo and equipping them with antibodies for efficient targeting of lymphoma cells. RESULTS: The post-isolation engineering of the EVs is accomplished by a freeze–thaw me...

Descripción completa

Detalles Bibliográficos
Autores principales: Dumontel, Bianca, Susa, Francesca, Limongi, Tania, Vighetto, Veronica, Debellis, Doriana, Canta, Marta, Cauda, Valentina
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9107671/
https://www.ncbi.nlm.nih.gov/pubmed/35568919
http://dx.doi.org/10.1186/s13578-022-00784-9
_version_ 1784708533725429760
author Dumontel, Bianca
Susa, Francesca
Limongi, Tania
Vighetto, Veronica
Debellis, Doriana
Canta, Marta
Cauda, Valentina
author_facet Dumontel, Bianca
Susa, Francesca
Limongi, Tania
Vighetto, Veronica
Debellis, Doriana
Canta, Marta
Cauda, Valentina
author_sort Dumontel, Bianca
collection PubMed
description BACKGROUND: We propose an efficient method to modify B-cell derived EVs by loading them with a nanotherapeutic stimuli-responsive cargo and equipping them with antibodies for efficient targeting of lymphoma cells. RESULTS: The post-isolation engineering of the EVs is accomplished by a freeze–thaw method to load therapeutically-active zinc oxide nanocrystals (ZnO NCs), obtaining the so-called TrojanNanoHorse (TNH) to recall the biomimetism and cytotoxic potential of this novel nanoconstruct. TNHs are further modified at their surface with anti-CD20 monoclonal antibodies (TNH(CD20)) achieving specific targeting against lymphoid cancer cell line. The in vitro characterization is carried out on CD20+ lymphoid Daudi cell line, CD20-negative cancerous myeloid cells (HL60) and the healthy counterpart (B lymphocytes). The TNH shows nanosized structure, high colloidal stability, even over time, and good hemocompatibility. The in vitro characterization shows the high biocompatibility, targeting specificity and cytotoxic capability. Importantly, the selectivity of TNH(CD20) demonstrates significantly higher interaction towards the target lymphoid Daudi cell line compared to the CD20-negative cancerous myeloid cells (HL60) and the healthy counterpart (lymphocytes). An enhanced cytotoxicity directed against Daudi cancer cells is demonstrated after the TNH(CD20) activation with high-energy ultrasound shock-waves (SW). CONCLUSION: This work demonstrates the efficient re-engineering of EVs, derived from healthy cells, with inorganic nanoparticles and monoclonal antibodies. The obtained hybrid nanoconstructs can be on-demand activated by an external stimulation, here acoustic pressure waves, to exploit a cytotoxic effect conveyed by the ZnO NCs cargo against selected cancer cells. GRAPHICAL ABSTRACT: [Image: see text] SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13578-022-00784-9.
format Online
Article
Text
id pubmed-9107671
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-91076712022-05-16 Nanotechnological engineering of extracellular vesicles for the development of actively targeted hybrid nanodevices Dumontel, Bianca Susa, Francesca Limongi, Tania Vighetto, Veronica Debellis, Doriana Canta, Marta Cauda, Valentina Cell Biosci Research BACKGROUND: We propose an efficient method to modify B-cell derived EVs by loading them with a nanotherapeutic stimuli-responsive cargo and equipping them with antibodies for efficient targeting of lymphoma cells. RESULTS: The post-isolation engineering of the EVs is accomplished by a freeze–thaw method to load therapeutically-active zinc oxide nanocrystals (ZnO NCs), obtaining the so-called TrojanNanoHorse (TNH) to recall the biomimetism and cytotoxic potential of this novel nanoconstruct. TNHs are further modified at their surface with anti-CD20 monoclonal antibodies (TNH(CD20)) achieving specific targeting against lymphoid cancer cell line. The in vitro characterization is carried out on CD20+ lymphoid Daudi cell line, CD20-negative cancerous myeloid cells (HL60) and the healthy counterpart (B lymphocytes). The TNH shows nanosized structure, high colloidal stability, even over time, and good hemocompatibility. The in vitro characterization shows the high biocompatibility, targeting specificity and cytotoxic capability. Importantly, the selectivity of TNH(CD20) demonstrates significantly higher interaction towards the target lymphoid Daudi cell line compared to the CD20-negative cancerous myeloid cells (HL60) and the healthy counterpart (lymphocytes). An enhanced cytotoxicity directed against Daudi cancer cells is demonstrated after the TNH(CD20) activation with high-energy ultrasound shock-waves (SW). CONCLUSION: This work demonstrates the efficient re-engineering of EVs, derived from healthy cells, with inorganic nanoparticles and monoclonal antibodies. The obtained hybrid nanoconstructs can be on-demand activated by an external stimulation, here acoustic pressure waves, to exploit a cytotoxic effect conveyed by the ZnO NCs cargo against selected cancer cells. GRAPHICAL ABSTRACT: [Image: see text] SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13578-022-00784-9. BioMed Central 2022-05-14 /pmc/articles/PMC9107671/ /pubmed/35568919 http://dx.doi.org/10.1186/s13578-022-00784-9 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Dumontel, Bianca
Susa, Francesca
Limongi, Tania
Vighetto, Veronica
Debellis, Doriana
Canta, Marta
Cauda, Valentina
Nanotechnological engineering of extracellular vesicles for the development of actively targeted hybrid nanodevices
title Nanotechnological engineering of extracellular vesicles for the development of actively targeted hybrid nanodevices
title_full Nanotechnological engineering of extracellular vesicles for the development of actively targeted hybrid nanodevices
title_fullStr Nanotechnological engineering of extracellular vesicles for the development of actively targeted hybrid nanodevices
title_full_unstemmed Nanotechnological engineering of extracellular vesicles for the development of actively targeted hybrid nanodevices
title_short Nanotechnological engineering of extracellular vesicles for the development of actively targeted hybrid nanodevices
title_sort nanotechnological engineering of extracellular vesicles for the development of actively targeted hybrid nanodevices
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9107671/
https://www.ncbi.nlm.nih.gov/pubmed/35568919
http://dx.doi.org/10.1186/s13578-022-00784-9
work_keys_str_mv AT dumontelbianca nanotechnologicalengineeringofextracellularvesiclesforthedevelopmentofactivelytargetedhybridnanodevices
AT susafrancesca nanotechnologicalengineeringofextracellularvesiclesforthedevelopmentofactivelytargetedhybridnanodevices
AT limongitania nanotechnologicalengineeringofextracellularvesiclesforthedevelopmentofactivelytargetedhybridnanodevices
AT vighettoveronica nanotechnologicalengineeringofextracellularvesiclesforthedevelopmentofactivelytargetedhybridnanodevices
AT debellisdoriana nanotechnologicalengineeringofextracellularvesiclesforthedevelopmentofactivelytargetedhybridnanodevices
AT cantamarta nanotechnologicalengineeringofextracellularvesiclesforthedevelopmentofactivelytargetedhybridnanodevices
AT caudavalentina nanotechnologicalengineeringofextracellularvesiclesforthedevelopmentofactivelytargetedhybridnanodevices