scholarly journals Protein delivery into cells using inorganic nanoparticle–protein supramolecular assemblies

2018 ◽  
Vol 47 (10) ◽  
pp. 3421-3432 ◽  
Author(s):  
Federica Scaletti ◽  
Joseph Hardie ◽  
Yi-Wei Lee ◽  
David C. Luther ◽  
Moumita Ray ◽  
...  

Direct intracellular delivery of proteins using inorganic nanoparticle–protein supramolecular assemblies.

2019 ◽  
Vol 5 (6) ◽  
pp. eaaw8922 ◽  
Author(s):  
Chongyi Liu ◽  
Tao Wan ◽  
Hui Wang ◽  
Song Zhang ◽  
Yuan Ping ◽  
...  

Cytosolic protein delivery is of central importance for the development of protein-based biotechnologies and therapeutics; however, efficient intracellular delivery of native proteins remains a challenge. Here, we reported a boronic acid–rich dendrimer with unprecedented efficiency for cytosolic delivery of native proteins. The dendrimer could bind with both negatively and positively charged proteins and efficiently delivered 13 cargo proteins into the cytosol of living cells. All the delivered proteins kept their bioactivities after cytosolic delivery. The dendrimer ensures efficient intracellular delivery of Cas9 protein into various cell lines and showed high efficiency in CRISPR-Cas9 genome editing. The rationally designed boronic acid–rich dendrimer permits the development of an efficient platform with high generality for the delivery of native proteins.


2015 ◽  
Vol 3 (47) ◽  
pp. 9115-9121 ◽  
Author(s):  
Bingru Zeng ◽  
Hongdong Shi ◽  
Yangzhong Liu

A highly biocompatible nanoplatform for the intracellular delivery of different proteins, exhibiting pH-responsive release and efficient endosomal escape.


2016 ◽  
Vol 52 (93) ◽  
pp. 13608-13611 ◽  
Author(s):  
Jie Li ◽  
Linlin Zhang ◽  
Yang Liu ◽  
Jing Wen ◽  
Di Wu ◽  
...  

We reported an efficient strategy for the intracellular delivery of proteins based on assembling proteins with a self-crosslinkable polymer. The disulfide-crosslinking structure enhances the stability of the protein–polymer assembly, and also allows effective dissociation of the assembly in response to glutathione.


2018 ◽  
Vol 9 (27) ◽  
pp. 5967-5975 ◽  
Author(s):  
Lele Sun ◽  
Yanjing Gao ◽  
Yaoguang Wang ◽  
Qin Wei ◽  
Jiye Shi ◽  
...  

A DNA-programmed membrane fusion strategy for directing intracellular delivery of proteins into live cells.


2014 ◽  
Vol 1622 ◽  
pp. 123-127
Author(s):  
Sana AHMED ◽  
Kazuaki MATSUMURA

ABSTRACTA number of drug carrier systems such as liposomes, polymeric-nanoparticles, microparticles, polymeric micelles have been investigated for intracellular delivery. Among these liposomes are the potential drug vehicles for efficient cytosolic delivery. They have an adhesive property for cell membrane to encapsulate the drug or protein effectively and showing the enhanced absorption rate. One of the problems could be the difficulty of incorporation of the drug or protein into cell. Therefore many studies of the drug carriers have been developed to enhance the intracellular delivery of materials. Here we propose the novel method to improve the intracellular uptaking by using freeze concentration. Solutes are excluded from ice crystallization and concentrated in the remaining solution during freezing by freezing concentration. We have reported that polymeric cryoprotectant which is carboxylated poly-L-lysine was adsorbed on to the cell membrane during freezing and caused effective freeze concentration. In this study we investigated that delivery of protein effectively taking place by liposome as a carrier agent. It was successfully delivered protein to L929 cells via freeze concentration using polymeric cryoprotectant as a novel drug delivery.


2021 ◽  
Author(s):  
Rebecca L. Hershman ◽  
Yamin Li ◽  
Feihe Ma ◽  
Qioabing Xu ◽  
James A. Van Deventer

AbstractMany intracellular signaling events remain poorly characterized due to a general lack of tools to interfere with “undruggable” targets. Antibodies have the potential to elucidate intracellular mechanisms via targeted disruption of cell signaling cascades because of their ability to bind to a target with high specificity and affinity. However, due to their size and chemical composition, antibodies cannot innately cross the cell membrane, and thus access to the cytosol with these macromolecules has been limited. Here, we describe strategies for accessing the intracellular space with recombinant antibodies mediated by cationic lipid nanoparticles to selectively disrupt intracellular signaling events. To enable such investigations, we first produced a series of antibody constructs, known as scFv-Fcs, containing additional, genetically encoded negative charges located at the C-termini of the constructs. Preparing proteins with negatively charged motifs has previously been shown to enhance intracellular protein delivery with cationic lipids, but usually for the purpose of genome editing or targeted cell death. We started by generating derivatives of scFv-Fc17, an antibody construct previously reported to bind specifically to signal transducer and activator of transcription 3 (STAT3) phosphorylated at Tyr705 (pYSTAT3). We screened a small number of lipids from our combinatorial lipid library with flow cytometry and found that PBA-Q76-O16B facilitated the most efficient delivery of scFv-Fcs under the conditions tested. In HepG2 cells, we observed up to 60.5% delivery efficacy, while in a STAT3-luciferase reporter cell line up to 71.5% delivery efficacy was observed. These results demonstrated the feasibility of accessing the intracellular space with scFv-Fcs. However, we also note that no more than modest changes were observed upon changing the numbers of negative charges in these constructs during delivery. Characterization of the cytotoxicity, size, and encapsulation efficiency of scFv-Fcs with PBA-Q76-O16B revealed that the constructs were generally well-behaved, with addition of differing quantities of negative charge resulting in at most modest effects. Importantly, functional assays monitoring transcriptional activity in luciferase reporter cell lines and HepG2 cells demonstrated significant reduction of gene expression downstream of pYSTAT3 following delivery of scFv-Fc17 constructs. Together, our results establish the use of recombinantly produced antibodies to selectively interfere with cell signaling events driven by a single posttranslational modification. Efficient intracellular delivery of engineered antibodies opens up possibilities for modulation of previously “undruggable” targets, including for potential therapeutic applications.


2019 ◽  
Author(s):  
Rohit Bhadoria ◽  
Kefeng Ping ◽  
Christer Lohk ◽  
Ivar Järving ◽  
Pavel Starkov

<div> <div> <div> <p>Conjugation techniques are central to improving intracellular delivery of bioactive small molecules. However, tracking and assessing the overall biological outcome of these constructs remains poorly understood. We addressed this issue by having developed a focused library of heterobivalent constructs based on Rho kinase inhibitors to probe various scenarios. By comparing induction of a phenotype of interest vs. cell viability vs. cellular uptake, we demonstrate that such conjugates indeed lead to divergent cellular outcomes. </p> </div> </div> </div>


Sign in / Sign up

Export Citation Format

Share Document