Your browser doesn't support javascript.
loading
A graphene-sandwiched DNA nano-system: regulation of intercalated doxorubicin for cellular localization.
Nandi, Semonti; Kale, Narendra; Patil, Ashwini; Banerjee, Shashwat; Patil, Yuvraj; Khandare, Jayant.
Afiliação
  • Nandi S; MAEER's Maharashtra Institute of Pharmacy Kothrud Pune 411038 India.
  • Kale N; School of Pharmacy, Dr Vishwanath Karad MIT World Peace University MIT Campus, S. No. 124, Paud Road, Kothrud Pune 411038 India jayant.khandare@mippune.edu.in.
  • Patil A; MAEER's Maharashtra Institute of Pharmacy Kothrud Pune 411038 India.
  • Banerjee S; School of Pharmacy, Dr Vishwanath Karad MIT World Peace University MIT Campus, S. No. 124, Paud Road, Kothrud Pune 411038 India jayant.khandare@mippune.edu.in.
  • Patil Y; MAEER's Maharashtra Institute of Pharmacy Kothrud Pune 411038 India.
  • Khandare J; School of Pharmacy, Dr Vishwanath Karad MIT World Peace University MIT Campus, S. No. 124, Paud Road, Kothrud Pune 411038 India jayant.khandare@mippune.edu.in.
Nanoscale Adv ; 2(12): 5746-5759, 2020 Dec 15.
Article em En | MEDLINE | ID: mdl-36133866
ABSTRACT
Control of the sub-cellular localization of nanoparticles (NPs) with enhanced drug-loading capacity, employing graphene oxide (GO), iron oxide (Fe3O4) NPs and sandwiched deoxyribonucleic acid (DNA) bearing intercalated anticancer drug doxorubicin (DOX) has been investigated in this work. The nanosystems G-DNA-DOX-Fe3O4 and Fe3O4-DNA-DOX differentially influence serum protein binding and deliver DOX to lysosomal compartments of cervical cancer (HeLa) cells with enhanced retention. Stern-Volmer plots describing BSA adsorption on the nanosystems demonstrated the quenching constants, K sv for G-DNA-DOX-Fe3O4 and Fe3O4-DNA-DOX (0.025 mL µg-1 and 0.0103 mL µg-1 respectively). Nuclear DOX intensity, measured at 24 h, was ∼2.0 fold higher for Fe3O4-DNA-DOX in HeLa cells. Parallelly, the cytosol displayed ∼2.2 fold higher DOX intensity for Fe3O4-DNA-DOX compared to G-DNA-DOX-Fe3O4. Fe3O4-DNA-DOX was more efficacious in the cytotoxic effect than G-DNA-DOX-Fe3O4 (viability of treated cells 33% and 49% respectively). The DNAnanosystems demonstrated superior cytotoxicity compared to mole-equivalent free DOX administration. The results implicate DNADOX NPs in influencing the cellular uptake mechanism and were critically subject to cellular localization. Furthermore, cell morphology analysis evidenced maximum deformation attributed to free-DOX with 34% increased cell roundness, 63% decreased cell area and ∼1.9 times increased nuclear-to-cytoplasmic (N/C) ratio after 24 h. In the case of Fe3O4-DNA-DOX, the N/C ratio increased 1.2 times and a maximum ∼37% decrease in NSA was noted suggesting involvement of non-canonical cytotoxic pathways. In conclusion, the study makes a case for designing nanosystems with controlled and regulated sub-cellular localization to potentially exploit secondary cytotoxic pathways, in addition to optimized drug-loading for enhanced anticancer efficacy and reduced adverse effects.

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Ano de publicação: 2020 Tipo de documento: Article