Cargando…

Targeted delivery of silibinin via magnetic niosomal nanoparticles: potential application in treatment of colon cancer cells

Introduction: In recent years, various nanoparticles (NPs) have been discovered and synthesized for the targeted therapy of cancer cells. Targeted delivery increases the local concentration of therapeutics and minimizes side effects. Therefore, NPs-mediated targeted drug delivery systems have become...

Descripción completa

Detalles Bibliográficos
Autores principales: Shafiei, Golchin, Jafari-Gharabaghlou, Davoud, Farhoudi-Sefidan-Jadid, Mahdi, Alizadeh, Effat, Fathi, Marziyeh, Zarghami, Nosratollah
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10335571/
https://www.ncbi.nlm.nih.gov/pubmed/37441534
http://dx.doi.org/10.3389/fphar.2023.1174120
_version_ 1785071027007520768
author Shafiei, Golchin
Jafari-Gharabaghlou, Davoud
Farhoudi-Sefidan-Jadid, Mahdi
Alizadeh, Effat
Fathi, Marziyeh
Zarghami, Nosratollah
author_facet Shafiei, Golchin
Jafari-Gharabaghlou, Davoud
Farhoudi-Sefidan-Jadid, Mahdi
Alizadeh, Effat
Fathi, Marziyeh
Zarghami, Nosratollah
author_sort Shafiei, Golchin
collection PubMed
description Introduction: In recent years, various nanoparticles (NPs) have been discovered and synthesized for the targeted therapy of cancer cells. Targeted delivery increases the local concentration of therapeutics and minimizes side effects. Therefore, NPs-mediated targeted drug delivery systems have become a promising approach for the treatment of various cancers. As a result, in the current study, we aimed to design silibinin-loaded magnetic niosomes nanoparticles (MNNPs) and investigate their cytotoxicity property in colorectal cancer cell treatment. Methods: MNPs ferrofluids were prepared and encapsulated into niosomes (NIOs) by the thin film hydration method. Afterward, the morphology, size, and chemical structure of the synthesized MNNPs were evaluated using the TEM, DLS, and FT-IR techniques, respectively. Results and Discussion: The distribution number of MNNPs was obtained at about 50 nm and 70 nm with a surface charge of −19.0 mV by TEM and DLS analysis, respectively. Silibinin loading efficiency in NIOs was about 90%, and the drug release pattern showed a controlled release with a maximum amount of about 49% and 70%, within 4 h in pH = 7.4 and pH = 5.8, respectively. To investigate the cytotoxicity effect, HT-29 cells were treated with the various concentration of the drugs for 24 and 48 h and evaluated by the MTT as well as flow cytometry assays. Obtained results demonstrated promoted cell cytotoxicity of silibinin-loaded MNNPs (5-fold decrease in cell viability) compared to pure silibinin (3-fold decrease in cell viability) while had no significant cytotoxic effect on HEK-293 (normal cell line) cells, and the cellular uptake level of MNNPs by the HT-29 cell line was enhanced compared to the control group. In conclusion, silibinin-loaded MNNPs complex can be considered as an efficient treatment approach for colorectal cancer cells.
format Online
Article
Text
id pubmed-10335571
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-103355712023-07-12 Targeted delivery of silibinin via magnetic niosomal nanoparticles: potential application in treatment of colon cancer cells Shafiei, Golchin Jafari-Gharabaghlou, Davoud Farhoudi-Sefidan-Jadid, Mahdi Alizadeh, Effat Fathi, Marziyeh Zarghami, Nosratollah Front Pharmacol Pharmacology Introduction: In recent years, various nanoparticles (NPs) have been discovered and synthesized for the targeted therapy of cancer cells. Targeted delivery increases the local concentration of therapeutics and minimizes side effects. Therefore, NPs-mediated targeted drug delivery systems have become a promising approach for the treatment of various cancers. As a result, in the current study, we aimed to design silibinin-loaded magnetic niosomes nanoparticles (MNNPs) and investigate their cytotoxicity property in colorectal cancer cell treatment. Methods: MNPs ferrofluids were prepared and encapsulated into niosomes (NIOs) by the thin film hydration method. Afterward, the morphology, size, and chemical structure of the synthesized MNNPs were evaluated using the TEM, DLS, and FT-IR techniques, respectively. Results and Discussion: The distribution number of MNNPs was obtained at about 50 nm and 70 nm with a surface charge of −19.0 mV by TEM and DLS analysis, respectively. Silibinin loading efficiency in NIOs was about 90%, and the drug release pattern showed a controlled release with a maximum amount of about 49% and 70%, within 4 h in pH = 7.4 and pH = 5.8, respectively. To investigate the cytotoxicity effect, HT-29 cells were treated with the various concentration of the drugs for 24 and 48 h and evaluated by the MTT as well as flow cytometry assays. Obtained results demonstrated promoted cell cytotoxicity of silibinin-loaded MNNPs (5-fold decrease in cell viability) compared to pure silibinin (3-fold decrease in cell viability) while had no significant cytotoxic effect on HEK-293 (normal cell line) cells, and the cellular uptake level of MNNPs by the HT-29 cell line was enhanced compared to the control group. In conclusion, silibinin-loaded MNNPs complex can be considered as an efficient treatment approach for colorectal cancer cells. Frontiers Media S.A. 2023-06-27 /pmc/articles/PMC10335571/ /pubmed/37441534 http://dx.doi.org/10.3389/fphar.2023.1174120 Text en Copyright © 2023 Shafiei, Jafari-Gharabaghlou, Farhoudi-Sefidan-Jadid, Alizadeh, Fathi and Zarghami. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Pharmacology
Shafiei, Golchin
Jafari-Gharabaghlou, Davoud
Farhoudi-Sefidan-Jadid, Mahdi
Alizadeh, Effat
Fathi, Marziyeh
Zarghami, Nosratollah
Targeted delivery of silibinin via magnetic niosomal nanoparticles: potential application in treatment of colon cancer cells
title Targeted delivery of silibinin via magnetic niosomal nanoparticles: potential application in treatment of colon cancer cells
title_full Targeted delivery of silibinin via magnetic niosomal nanoparticles: potential application in treatment of colon cancer cells
title_fullStr Targeted delivery of silibinin via magnetic niosomal nanoparticles: potential application in treatment of colon cancer cells
title_full_unstemmed Targeted delivery of silibinin via magnetic niosomal nanoparticles: potential application in treatment of colon cancer cells
title_short Targeted delivery of silibinin via magnetic niosomal nanoparticles: potential application in treatment of colon cancer cells
title_sort targeted delivery of silibinin via magnetic niosomal nanoparticles: potential application in treatment of colon cancer cells
topic Pharmacology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10335571/
https://www.ncbi.nlm.nih.gov/pubmed/37441534
http://dx.doi.org/10.3389/fphar.2023.1174120
work_keys_str_mv AT shafieigolchin targeteddeliveryofsilibininviamagneticniosomalnanoparticlespotentialapplicationintreatmentofcoloncancercells
AT jafarigharabaghloudavoud targeteddeliveryofsilibininviamagneticniosomalnanoparticlespotentialapplicationintreatmentofcoloncancercells
AT farhoudisefidanjadidmahdi targeteddeliveryofsilibininviamagneticniosomalnanoparticlespotentialapplicationintreatmentofcoloncancercells
AT alizadeheffat targeteddeliveryofsilibininviamagneticniosomalnanoparticlespotentialapplicationintreatmentofcoloncancercells
AT fathimarziyeh targeteddeliveryofsilibininviamagneticniosomalnanoparticlespotentialapplicationintreatmentofcoloncancercells
AT zarghaminosratollah targeteddeliveryofsilibininviamagneticniosomalnanoparticlespotentialapplicationintreatmentofcoloncancercells