Increased cellular uptake of peptide-modified PEGylated gold nanoparticles

2017 ◽  
Vol 494 (1-2) ◽  
pp. 339-345 ◽  
Author(s):  
Bo He ◽  
Dan Yang ◽  
Mengmeng Qin ◽  
Yuan Zhang ◽  
Bing He ◽  
...  
2015 ◽  
Vol 25 (22) ◽  
pp. 3433-3439 ◽  
Author(s):  
Zhiyue Zhang ◽  
Katleen Van Steendam ◽  
Samarendra Maji ◽  
Lieve Balcaen ◽  
Yulia Anoshkina ◽  
...  

2016 ◽  
Vol 36 (suppl_1) ◽  
Author(s):  
Vasanthy Narayanaswami ◽  
Skylar T Chuang ◽  
Young-Seok Shon

We have developed a novel HDL-based multifunctional platform for transport and delivery of highly hydrophobic gold nanoparticles (AuNP) bearing photothermic properties. We exploit the ability of apolipoprotein E3 (apoE3) to act as a high affinity ligand for the low-density lipoprotein receptor (LDLr) to gain entry into glioblastoma cells. The issue of poor aqueous solubility of AuNP was overcome by integrating them with phospholipids and apoE3, yielding reconstituted rHDL bearing 3, 10, or 10 nm AuNP. UV-Vis spectra of rHDL-AuNP indicated the presence of stable particles with surface plasmon band at ~530 nm, a signature feature of AuNP. Transmission electron microscopy (TEM) revealed discoidal geometry for rHDL with 3, 10 and 17 nm particles. Co-immunoprecipitation assay using a soluble form of the LDLr indicated robust binding of rHDL-AuNP to the receptor. Immunofluorescence analysis indicated that all 3 rHDL particles were internalized by glioblastoma cells, as revealed by the presence of punctate, peri-nuclear endocytic/lysosomal vesicles; this suggests cellular uptake of rHDL-AuNP by receptor-mediated endocytosis. Cellular uptake was further confirmed by TEM, in which aggregated AuNP were found in the endosomal-lysosomal compartments. Finally, cytotoxicity studies demonstrated that more than 50% of cells were viable with rHDL-AuNP treatment of up to 0.1 mg/ml for 24 h. The findings that apoE3: (i) acts as a detergent in solubilizing and dramatically improving the aqueous solubility of AuNP, and, (ii) facilitates cellular uptake of rHDL-AuNP by the LDLr pathway, are significant since they offer an effective means of delivering AuNP across the cell membrane. This is particularly relevant in tumor cells since they overexpress LDLr to meet the high demand for cholesterol that is required for rapid proliferation and membrane biogenesis.


2017 ◽  
Vol 91 ◽  
pp. 123-133 ◽  
Author(s):  
Suhana Ahmad ◽  
Anes Ateqah Zamry ◽  
Hern-Tze Tina Tan ◽  
Kah Keng Wong ◽  
JitKang Lim ◽  
...  

2019 ◽  
Vol 1052 ◽  
pp. 1-9 ◽  
Author(s):  
Magdalena Matczuk ◽  
Lena Ruzik ◽  
Svetlana S. Aleksenko ◽  
Bernhard K. Keppler ◽  
Maciej Jarosz ◽  
...  

Langmuir ◽  
2018 ◽  
Vol 34 (46) ◽  
pp. 14033-14045 ◽  
Author(s):  
Kwun Hei Samuel Sy ◽  
Lok Wai Cola Ho ◽  
Wilson Chun Yu Lau ◽  
Ho Ko ◽  
Chung Hang Jonathan Choi

2020 ◽  
Author(s):  
Mehvesh Hameed ◽  
Seema Panicker ◽  
Sallam Hasan Abdallah ◽  
Amir A. Khan ◽  
Changseok Han ◽  
...  

We synthesized protein-coated gold nanoparticles using green and chemical reduction routes for cellular uptake study. In the current work, we coated gold-aryl nanoparticles of the type AuNPs-C<sub>6</sub>H<sub>4</sub>-4-COOH with BSA, collagen, zein and lysozyme proteins. Both routes were carried out without phase-transfer catalysts or extraneous stabilizing agents. High crystallinity of the AuNPs synthesized by the green route can be seen in the transmission electron microscopy images. <a>Osteosarcoma cancer cells are malignant bone tumors with abnormal cellular functions. Studies using MG-63 cells will provide mechanistic suggestions on the details of the amplification in tumors. </a>We studied the cellular uptake of the bioconjugates by MG-63 osteosarcoma cells using laser confocal fluorescence microscopy (LCFM) and flow cytometry. In the LCFM study, BSA-AuNPs was uptaken most efficiently of all protein-coated gold nanoparticles synthesized by the green route. Zein and lysozyme coated nanoparticles, though small sizes, prepared by the green method were not efficiently uptaken by MG-63. The two nanoparticles are negatively charged and zein is also a hydrophobic coat. The difference in hydrophobicity and charge might have affected the internalization. All of those coated nanoparticles that were efficiently uptaken can potentially be used as diagnostic and therapeutic agents for osteosarcoma.


2018 ◽  
Vol 140 (13) ◽  
pp. 4469-4472 ◽  
Author(s):  
Jesús Mosquera ◽  
Malou Henriksen-Lacey ◽  
Isabel García ◽  
Miguel Martínez-Calvo ◽  
Jéssica Rodríguez ◽  
...  

Nanomaterials ◽  
2019 ◽  
Vol 9 (3) ◽  
pp. 396 ◽  
Author(s):  
Suhash Chavva ◽  
Sachin Deshmukh ◽  
Rajashekhar Kanchanapally ◽  
Nikhil Tyagi ◽  
Jason Coym ◽  
...  

Epigallocatechin gallate (EGCG) possesses significant antitumor activity and binds to laminin receptors, overexpressed on cancer cells, with high affinity. Gold nanoparticles (GNPs) serve as excellent drug carriers and protect the conjugated drug from enzymatic metabolization. Citrate-gold nanoparticles (C-GNPs) and EGCG-gold nanoparticles (E-GNPs) were synthesized by reduction methods and characterized with UV-visible spectroscopy, transmission electron microscopy (TEM), and dynamic light scattering (DLS). Cytotoxicity of citrate, EGCG, C-GNPs, and E-GNPs was evaluated by the water-soluble tetrazolium salt (WST-1) assay. Nanoparticle cellular uptake studies were performed by TEM and atomic absorption spectroscopy (AAS). Dialysis method was employed to assess drug release. Cell viability studies showed greater growth inhibition by E-GNPs compared to EGCG or C-GNPs. Cellular uptake studies revealed that, unlike C-GNPs, E-GNPs were taken up more efficiently by cancerous cells than noncancerous cells. We found that E-GNP nanoformulation releases EGCG in a sustained fashion. Furthermore, data showed that E-GNPs induced more apoptosis in cancer cells compared to EGCG and C-GNPs. From the mechanistic standpoint, we observed that E-GNPs inhibited the nuclear translocation and transcriptional activity of nuclear factor-kappaB (NF-κB) with greater potency than EGCG, whereas C-GNPs were only minimally effective. Altogether, our data suggest that E-GNPs can serve as potent tumor-selective chemotoxic agents.


Sign in / Sign up

Export Citation Format

Share Document