: Lipid nanoparticles (LNPs) are currently having an increasing impact on nanomedicines as delivery agents, among others, of RNA molecules (e.g., short interfering RNA for the treatment of hereditary diseases or messenger RNA for the development of COVID-19 vaccines). Despite this, the delivery of plasmid DNA (pDNA) by LNPs in preclinical studies is still unsatisfactory, mainly due to the lack of systematic structural and functional studies on DNA-loaded LNPs. To tackle this issue, we developed, characterized, and tested a library of 16 multicomponent DNA-loaded LNPs which were prepared by microfluidics and differed in lipid composition, surface functionalization, and manufacturing factors. 8 out of 16 formulations exhibited proper size and zeta potential and passed to the validation step, that is, the simultaneous quantification of transfection efficiency and cell viability in human embryonic kidney cells (HEK-293). The most efficient formulation (LNP15) was then successfully validated both in vitro, in an immortalized adult keratinocyte cell line (HaCaT) and in an epidermoid cervical cancer cell line (CaSki), and in vivo as a nanocarrier to deliver a cancer vaccine against the benchmark target tyrosine-kinase receptor HER2 in C57BL/6 mice. Finally, by a combination of confocal microscopy, transmission electron microscopy and synchrotron small-angle X-ray scattering, we were able to show that the superior efficiency of LNP15 can be linked to its disordered nanostructure consisting of small-size unoriented layers of pDNA sandwiched between closely apposed lipid membranes that undergo massive destabilization upon interaction with cellular lipids. Our results provide new insights into the structure-activity relationship of pDNA-loaded LNPs and pave the way to the clinical translation of this gene delivery technology.

Mechanistic insights into the superior DNA delivery efficiency of multicomponent lipid nanoparticles: an in vitro and in vivo study / Quagliarini, E.; Wang, J.; Renzi, S.; Cui, L.; Digiacomo, L.; Ferri, G.; Pesce, L.; De Lorenzi, V.; Matteoli, G.; Amenitsch, H.; Masuelli, L.; Bei, R.; Pozzi, D.; Amici, A.; Cardarelli, F.; Marchini, C.; Caracciolo, G.. - In: ACS APPLIED MATERIALS & INTERFACES. - ISSN 1944-8252. - 14:51(2022), pp. 56666-56677. [10.1021/acsami.2c20019]

Mechanistic insights into the superior DNA delivery efficiency of multicomponent lipid nanoparticles: an in vitro and in vivo study

Quagliarini E.;Renzi S.;Digiacomo L.;Masuelli L.;Pozzi D.;Caracciolo G.
2022

Abstract

: Lipid nanoparticles (LNPs) are currently having an increasing impact on nanomedicines as delivery agents, among others, of RNA molecules (e.g., short interfering RNA for the treatment of hereditary diseases or messenger RNA for the development of COVID-19 vaccines). Despite this, the delivery of plasmid DNA (pDNA) by LNPs in preclinical studies is still unsatisfactory, mainly due to the lack of systematic structural and functional studies on DNA-loaded LNPs. To tackle this issue, we developed, characterized, and tested a library of 16 multicomponent DNA-loaded LNPs which were prepared by microfluidics and differed in lipid composition, surface functionalization, and manufacturing factors. 8 out of 16 formulations exhibited proper size and zeta potential and passed to the validation step, that is, the simultaneous quantification of transfection efficiency and cell viability in human embryonic kidney cells (HEK-293). The most efficient formulation (LNP15) was then successfully validated both in vitro, in an immortalized adult keratinocyte cell line (HaCaT) and in an epidermoid cervical cancer cell line (CaSki), and in vivo as a nanocarrier to deliver a cancer vaccine against the benchmark target tyrosine-kinase receptor HER2 in C57BL/6 mice. Finally, by a combination of confocal microscopy, transmission electron microscopy and synchrotron small-angle X-ray scattering, we were able to show that the superior efficiency of LNP15 can be linked to its disordered nanostructure consisting of small-size unoriented layers of pDNA sandwiched between closely apposed lipid membranes that undergo massive destabilization upon interaction with cellular lipids. Our results provide new insights into the structure-activity relationship of pDNA-loaded LNPs and pave the way to the clinical translation of this gene delivery technology.
2022
gene delivery; lipid nanoparticles; membrane disintegration; transfection efficiency; vaccination
01 Pubblicazione su rivista::01a Articolo in rivista
Mechanistic insights into the superior DNA delivery efficiency of multicomponent lipid nanoparticles: an in vitro and in vivo study / Quagliarini, E.; Wang, J.; Renzi, S.; Cui, L.; Digiacomo, L.; Ferri, G.; Pesce, L.; De Lorenzi, V.; Matteoli, G.; Amenitsch, H.; Masuelli, L.; Bei, R.; Pozzi, D.; Amici, A.; Cardarelli, F.; Marchini, C.; Caracciolo, G.. - In: ACS APPLIED MATERIALS & INTERFACES. - ISSN 1944-8252. - 14:51(2022), pp. 56666-56677. [10.1021/acsami.2c20019]
File allegati a questo prodotto
File Dimensione Formato  
Quagliarini_Mechanistic_2022.pdf

solo gestori archivio

Tipologia: Versione editoriale (versione pubblicata con il layout dell'editore)
Licenza: Tutti i diritti riservati (All rights reserved)
Dimensione 6.2 MB
Formato Adobe PDF
6.2 MB Adobe PDF   Contatta l'autore

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/11573/1671031
Citazioni
  • ???jsp.display-item.citation.pmc??? 1
  • Scopus 6
  • ???jsp.display-item.citation.isi??? 7
social impact