Your browser doesn't support javascript.
loading
Interrogating the Role of Endocytosis Pathway and Organelle Trafficking for Doxorubicin-Based Combination Ionic Nanomedicines.
Bashiru, Mujeebat; Rayaan, Muhammad; Ali, Nawab; Jenkins, Samir V; Oyebade, Adeniyi; Rahman, Md Shahedur; Griffin, Robert J; Oyelere, Adegboyega K; Siraj, Noureen.
Afiliação
  • Bashiru M; Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States.
  • Rayaan M; Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States.
  • Ali N; Department of Biology, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States.
  • Jenkins SV; Department of Radiation Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, Arkansas 72205, United States.
  • Oyebade A; Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States.
  • Rahman MS; Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States.
  • Griffin RJ; Department of Radiation Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, Arkansas 72205, United States.
  • Oyelere AK; School of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States.
  • Siraj N; Department of Chemistry, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States.
ACS Appl Bio Mater ; 7(8): 5359-5368, 2024 Aug 19.
Article em En | MEDLINE | ID: mdl-39102354
ABSTRACT
We have studied the endocytic mechanisms that determine subcellular localization for three carrier-free chemotherapeutic-photothermal (chemo-PTT) combination ionic nanomedicines (INMs) composed of doxorubicin (DOX) and an near-infrared (NIR) dye (ICG, IR820, or IR783). This study aims to understand the cellular basis for previously published enhanced toxicity results of these combination nanomedicines toward MCF-7 breast cancer cells. The active transport mechanism of INMs, unlike free DOX, which is known to employ passive transport, was validated by conducting temperature-dependent cellular uptake of the drug in MCF-7 cells using confocal microscopy. The internalization pathway of these INMs was further probed in the presence and absence of different endocytosis inhibitors. Detailed examination of the mode of entry of the carrier-free INMs in MCF-7 cells revealed that they are primarily internalized through clathrin-mediated endocytosis. In addition, time-dependent subcellular localization studies were also investigated. Examination of time-dependent confocal images indicated that the INMs targeted multiple organelles, in contrast to free DOX that primarily targets the nucleus. Collectively, the high cellular endocytic uptake in cancerous cells (EPR effect) and the multimode targeting ability demonstrated the main reason for the low half-maxima inhibitory concentration (IC50) value (the high cytotoxicity) of these carrier-free INMs as compared to their respective parent chemo and PTT drugs.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Doxorrubicina / Endocitose / Nanomedicina Limite: Humans Idioma: En Revista: ACS Appl Bio Mater Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Doxorrubicina / Endocitose / Nanomedicina Limite: Humans Idioma: En Revista: ACS Appl Bio Mater Ano de publicação: 2024 Tipo de documento: Article