Cargando…

Delivery of Quantum Dot-siRNA Nanoplexes in SK-N-SH Cells for BACE1 Gene Silencing and Intracellular Imaging

The fluorescent quantum dots (QDs) delivered small interfering RNAs (siRNAs) targeting β-secretase (BACE1) to achieve high transfection efficiency of siRNAs and reduction of β-amyloid (Aβ) in nerve cells. The CdSe/ZnS QDs with the conjugation of amino-polyethylene glycol (PEG) were synthesized. Nega...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Shengliang, Liu, Zhonghua, Ji, Fengtao, Xiao, Zijian, Wang, Minjuan, Peng, Yingji, Zhang, Yulin, Liu, Ling, Liang, Zibing, Li, Feng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2012
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3381642/
https://www.ncbi.nlm.nih.gov/pubmed/23343930
http://dx.doi.org/10.1038/mtna.2012.11
_version_ 1782236433028743168
author Li, Shengliang
Liu, Zhonghua
Ji, Fengtao
Xiao, Zijian
Wang, Minjuan
Peng, Yingji
Zhang, Yulin
Liu, Ling
Liang, Zibing
Li, Feng
author_facet Li, Shengliang
Liu, Zhonghua
Ji, Fengtao
Xiao, Zijian
Wang, Minjuan
Peng, Yingji
Zhang, Yulin
Liu, Ling
Liang, Zibing
Li, Feng
author_sort Li, Shengliang
collection PubMed
description The fluorescent quantum dots (QDs) delivered small interfering RNAs (siRNAs) targeting β-secretase (BACE1) to achieve high transfection efficiency of siRNAs and reduction of β-amyloid (Aβ) in nerve cells. The CdSe/ZnS QDs with the conjugation of amino-polyethylene glycol (PEG) were synthesized. Negatively charged siRNAs were electrostatically adsorbed to the surface of QDs to develop QD-PEG/siRNA nanoplexes. The QD-PEG/siRNAs nanoplexes significantly promote the transfection efficiency of siRNA, and the siRNAs from non-packaged nanoplexes were widely distributed in cell bodies and processes and efficiently silenced BACE1 gene, leading to the reduction of Aβ. The biodegradable PEG polymer coating could protect QDs from being exposed to the intracellular environment and restrained the release of toxic Cd2(+). Therefore, the QD-PEG/siRNA nanoplexes reported here might serve as ideal carriers for siRNAs. We developed a novel method of siRNA delivery into nerve cells. We first reported that the QD-PEG/siRNA nanoplexes were generated by the electrostatic interaction and inhibited the Alzheimer's disease (AD)-associated BACE1 gene. We also first revealed the dynamics of QD-PEG/siRNAs within nerve cells via confocal microscopy and the ultrastructural evidences under transmission electron microscopy (TEM). This technology might hold promise for the treatment of neurodegenerative diseases such as AD.
format Online
Article
Text
id pubmed-3381642
institution National Center for Biotechnology Information
language English
publishDate 2012
publisher Nature Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-33816422012-07-03 Delivery of Quantum Dot-siRNA Nanoplexes in SK-N-SH Cells for BACE1 Gene Silencing and Intracellular Imaging Li, Shengliang Liu, Zhonghua Ji, Fengtao Xiao, Zijian Wang, Minjuan Peng, Yingji Zhang, Yulin Liu, Ling Liang, Zibing Li, Feng Mol Ther Nucleic Acids Original Article The fluorescent quantum dots (QDs) delivered small interfering RNAs (siRNAs) targeting β-secretase (BACE1) to achieve high transfection efficiency of siRNAs and reduction of β-amyloid (Aβ) in nerve cells. The CdSe/ZnS QDs with the conjugation of amino-polyethylene glycol (PEG) were synthesized. Negatively charged siRNAs were electrostatically adsorbed to the surface of QDs to develop QD-PEG/siRNA nanoplexes. The QD-PEG/siRNAs nanoplexes significantly promote the transfection efficiency of siRNA, and the siRNAs from non-packaged nanoplexes were widely distributed in cell bodies and processes and efficiently silenced BACE1 gene, leading to the reduction of Aβ. The biodegradable PEG polymer coating could protect QDs from being exposed to the intracellular environment and restrained the release of toxic Cd2(+). Therefore, the QD-PEG/siRNA nanoplexes reported here might serve as ideal carriers for siRNAs. We developed a novel method of siRNA delivery into nerve cells. We first reported that the QD-PEG/siRNA nanoplexes were generated by the electrostatic interaction and inhibited the Alzheimer's disease (AD)-associated BACE1 gene. We also first revealed the dynamics of QD-PEG/siRNAs within nerve cells via confocal microscopy and the ultrastructural evidences under transmission electron microscopy (TEM). This technology might hold promise for the treatment of neurodegenerative diseases such as AD. Nature Publishing Group 2012-04 2012-04-24 /pmc/articles/PMC3381642/ /pubmed/23343930 http://dx.doi.org/10.1038/mtna.2012.11 Text en Copyright © 2012 American Society of Gene & Cell Therapy http://creativecommons.org/licenses/by-nc-nd/3.0/ Molecular Therapy-Nucleic Acids is an open-access journal published by Nature Publishing Group. This work is licensed under the Creative Commons Attribution-NonCommercial-No Derivative Works 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/3.0/
spellingShingle Original Article
Li, Shengliang
Liu, Zhonghua
Ji, Fengtao
Xiao, Zijian
Wang, Minjuan
Peng, Yingji
Zhang, Yulin
Liu, Ling
Liang, Zibing
Li, Feng
Delivery of Quantum Dot-siRNA Nanoplexes in SK-N-SH Cells for BACE1 Gene Silencing and Intracellular Imaging
title Delivery of Quantum Dot-siRNA Nanoplexes in SK-N-SH Cells for BACE1 Gene Silencing and Intracellular Imaging
title_full Delivery of Quantum Dot-siRNA Nanoplexes in SK-N-SH Cells for BACE1 Gene Silencing and Intracellular Imaging
title_fullStr Delivery of Quantum Dot-siRNA Nanoplexes in SK-N-SH Cells for BACE1 Gene Silencing and Intracellular Imaging
title_full_unstemmed Delivery of Quantum Dot-siRNA Nanoplexes in SK-N-SH Cells for BACE1 Gene Silencing and Intracellular Imaging
title_short Delivery of Quantum Dot-siRNA Nanoplexes in SK-N-SH Cells for BACE1 Gene Silencing and Intracellular Imaging
title_sort delivery of quantum dot-sirna nanoplexes in sk-n-sh cells for bace1 gene silencing and intracellular imaging
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3381642/
https://www.ncbi.nlm.nih.gov/pubmed/23343930
http://dx.doi.org/10.1038/mtna.2012.11
work_keys_str_mv AT lishengliang deliveryofquantumdotsirnananoplexesinsknshcellsforbace1genesilencingandintracellularimaging
AT liuzhonghua deliveryofquantumdotsirnananoplexesinsknshcellsforbace1genesilencingandintracellularimaging
AT jifengtao deliveryofquantumdotsirnananoplexesinsknshcellsforbace1genesilencingandintracellularimaging
AT xiaozijian deliveryofquantumdotsirnananoplexesinsknshcellsforbace1genesilencingandintracellularimaging
AT wangminjuan deliveryofquantumdotsirnananoplexesinsknshcellsforbace1genesilencingandintracellularimaging
AT pengyingji deliveryofquantumdotsirnananoplexesinsknshcellsforbace1genesilencingandintracellularimaging
AT zhangyulin deliveryofquantumdotsirnananoplexesinsknshcellsforbace1genesilencingandintracellularimaging
AT liuling deliveryofquantumdotsirnananoplexesinsknshcellsforbace1genesilencingandintracellularimaging
AT liangzibing deliveryofquantumdotsirnananoplexesinsknshcellsforbace1genesilencingandintracellularimaging
AT lifeng deliveryofquantumdotsirnananoplexesinsknshcellsforbace1genesilencingandintracellularimaging