1
|
Zhu D, Brückner D, Sosniok M, Skiba M, Feliu N, Gallego M, Liu Y, Schulz F, Falkenberg G, Parak WJ, Sanchez-Cano C. Size-dependent penetration depth of colloidal nanoparticles into cell spheroids. Adv Drug Deliv Rev 2025; 222:115593. [PMID: 40339992 DOI: 10.1016/j.addr.2025.115593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/18/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025]
Abstract
The penetration of nanoparticle (NP)-based drugs into tissue is essential for their use as nanomedicines. Systematic studies about how different NP properties, such as size, influence NP penetration are helpful for the development of NP-based drugs. An overview of how NPs of different sizes may penetrate three-dimensional cell spheroids is given. In particular different techniques for experimental analysis are compared, including mass spectrometry, flow cytometry, optical fluorescence microscopy, X-ray fluorescence microscopy, and transmission electron microscopy. An experimental data set is supplemented exclusively made for this review, in which the results of different techniques are visualized. Limitations of the analysis techniques for different types of NPs, including carbon-based materials, are discussed.
Collapse
Affiliation(s)
- Dingcheng Zhu
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany; Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121 Zhejiang, China
| | - Dennis Brückner
- Deutsches Elektronen-Synchrotron DESY, Photon Science, 22607 Hamburg, Germany
| | - Martin Sosniok
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany; Zentrum für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Marvin Skiba
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany
| | - Neus Feliu
- Zentrum für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Marta Gallego
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE) Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastián, Spain
| | - Yang Liu
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany
| | - Florian Schulz
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany
| | - Gerald Falkenberg
- Deutsches Elektronen-Synchrotron DESY, Photon Science, 22607 Hamburg, Germany.
| | - Wolfgang J Parak
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany.
| | - Carlos Sanchez-Cano
- Donostia International Physics Center, 20018 Donostia-San Sebastian, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain; Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU, 20018 Donostia-San Sebastian, Spain.
| |
Collapse
|
2
|
Noske S, Krueger M, Ewe A, Aigner A. Analysis of Polymer/siRNA Nanoparticle Efficacy and Biocompatibility in 3D Air-Liquid Interface Culture Compared to 2D Cell Culture. Pharmaceutics 2025; 17:339. [PMID: 40143003 PMCID: PMC11946471 DOI: 10.3390/pharmaceutics17030339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Polymeric nanoparticles have been explored as efficient tools for siRNA delivery to induce RNAi-mediated gene knockdown. Chemical modifications of polyethylenimines (PEI) enhance nanoparticle efficacy and biocompatibility. Their in vivo use, however, benefits from prior analyses in relevant in vitro 3D conditions. Methods: We utilize a 3D ALI cell culture model for testing the biological activities and toxicities of a set of different PEI-based nanoparticles with different chemical modifications. This also includes a novel, fluoroalkyl-modified PEI. Reporter gene knockdown is directly compared to 2D cell culture. In parallel, biocompatibility is assessed by measuring cell viability and lactate dehydrogenase (LDH) release. Results: Knockdown efficacies in the 3D ALI model are dependent on the chemical modification and complex preparation conditions. Results only correlate in part with gene knockdown in 2D cell culture, identifying nanoparticle penetration and cellular internalization under 3D conditions as important parameters. The 3D ALI cell culture is also suitable for the quantitative determination of nanoparticle effects on cell viability and acute toxicity, with biocompatibility benefitting from PEI modifications. Conclusions: The 3D ALI cell model allows for a more realistic assessment of biological nanoparticle effects. A novel fluoroalkyl-modified PEI is described. Optimal preparations of PEI-based nanoparticles for siRNA delivery and gene knockdown are identified.
Collapse
Affiliation(s)
- Sandra Noske
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, Leipzig University, 04107 Leipzig, Germany; (S.N.); (A.E.)
| | - Martin Krueger
- Institute of Anatomy, Leipzig University, Liebigstraße 13, 04103 Leipzig, Germany;
| | - Alexander Ewe
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, Leipzig University, 04107 Leipzig, Germany; (S.N.); (A.E.)
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, Leipzig University, 04107 Leipzig, Germany; (S.N.); (A.E.)
| |
Collapse
|
3
|
Kreofsky NW, Roy P, Reineke TM. pH-Responsive Micelles Containing Quinine Functionalities Enhance Intracellular Gene Delivery and Expression. Bioconjug Chem 2024; 35:1762-1778. [PMID: 39467734 DOI: 10.1021/acs.bioconjchem.4c00326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Quinine is a promising building block for creating polymer carriers for intracellular nucleic acid delivery. This is due to its ability to bind to genetic material through intercalation and electrostatic interactions and the balance of hydrophobicity and hydrophilicity dependent on the pH/charge state. Yet, studies utilizing cinchona alkaloid natural products in gene delivery are limited. Herein, we present the incorporation of a quinine functionalized monomer (Q) into block polymer architectures to form self-assembled micelles for highly efficient gene delivery. Q was incorporated into the core and/or the shell of the micelles to introduce the unique advantages of quinine to the system. We found that incorporation of Q into the core of the micelle resulted in acid-induced disassembly of the micelle and a boost in transfection efficiency by promoting endosomal escape. This effect was especially evident in the cancerous cell line, A549, which has a more acidic intracellular environment. Incorporation of Q into the shell of the micelles resulted in intercalative binding to the genetic payload as well as larger micelle-DNA complexes (micelleplexes) from the hydrophobicity of Q in the shell. These factors enable the micelleplexes to be more resistant to serum and have more persistent protein expression post-transfection. Overall, this study is the first to demonstrate the benefits of including quinine functionalities into self-assembled micelles for highly efficient gene delivery and presents a platform for inclusion of other natural products with similar properties into micellar systems.
Collapse
Affiliation(s)
- Nicholas W Kreofsky
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Punarbasu Roy
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
4
|
Sekar RP, Lawson JL, Wright ARE, McGrath C, Schadeck C, Kumar P, Tay JW, Dragavon J, Kumar R. Poly(l-glutamic acid) augments the transfection performance of lipophilic polycations by overcoming tradeoffs among cytotoxicity, pDNA delivery efficiency, and serum stability. RSC APPLIED POLYMERS 2024; 2:701-718. [PMID: 39035825 PMCID: PMC11255917 DOI: 10.1039/d4lp00085d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/27/2024] [Indexed: 07/23/2024]
Abstract
Polycations are scalable and affordable nanocarriers for delivering therapeutic nucleic acids. Yet, cationicity-dependent tradeoffs between nucleic acid delivery efficiency, cytotoxicity, and serum stability hinder clinical translation. Typically, the most efficient polycationic vehicles also tend to be the most toxic. For lipophilic polycations-which recruit hydrophobic interactions in addition to electrostatic interactions to bind and deliver nucleic acids-extensive chemical or architectural modifications sometimes fail to resolve intractable toxicity-efficiency tradeoffs. Here, we employ a facile post-synthetic polyplex surface modification strategy wherein poly(l-glutamic acid) (PGA) rescues toxicity, inhibits hemolysis, and prevents serum inhibition of lipophilic polycation-mediated plasmid (pDNA) delivery. Importantly, the sequence in which polycations, pDNA, and PGA are combined dictates pDNA conformations and spatial distribution. Circular dichroism spectroscopy reveals that PGA must be added last to polyplexes assembled from lipophilic polycations and pDNA; else, PGA will disrupt polycation-mediated pDNA condensation. Although PGA did not mitigate toxicity caused by hydrophilic PEI-based polycations, PGA tripled the population of transfected viable cells for lipophilic polycations. Non-specific adsorption of serum proteins abrogated pDNA delivery mediated by lipophilic polycations; however, PGA-coated polyplexes proved more serum-tolerant than uncoated polyplexes. Despite lower cellular uptake than uncoated polyplexes, PGA-coated polyplexes were imported into nuclei at higher rates. PGA also silenced the hemolytic activity of lipophilic polycations. Our work provides fundamental insights into how polyanionic coatings such as PGA transform intermolecular interactions between lipophilic polycations, nucleic acids, and serum proteins, and facilitate gentle yet efficient transgene delivery.
Collapse
Affiliation(s)
- Ram Prasad Sekar
- Chemical and Biological Engineering, Colorado School of Mines Golden CO 80401 USA
| | | | - Aryelle R E Wright
- Quantitative Biosciences and Engineering, Colorado School of Mines Golden CO 80401 USA
| | - Caleb McGrath
- Quantitative Biosciences and Engineering, Colorado School of Mines Golden CO 80401 USA
| | - Cesar Schadeck
- Materials Science, Colorado School of Mines Golden CO 80401 USA
| | - Praveen Kumar
- Shared Instrumentation Facility, Colorado School of Mines Golden CO USA
| | - Jian Wei Tay
- BioFrontiers Institute, University of Colorado Boulder CO 80303 USA
| | - Joseph Dragavon
- BioFrontiers Institute, University of Colorado Boulder CO 80303 USA
| | - Ramya Kumar
- Chemical and Biological Engineering, Colorado School of Mines Golden CO 80401 USA
| |
Collapse
|
5
|
Wang Y, Wang J, Li J, Mu Y, Ying J, Liu Z, Wu M, Geng Y, Zhou X, Zhou T, Shen Y, Sun L, Liu X, Zhou Q. Sulfoxide-containing polymers conjugated prodrug micelles with enhanced anticancer activity and reduced intestinal toxicity. J Control Release 2024; 371:313-323. [PMID: 38823585 DOI: 10.1016/j.jconrel.2024.05.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Poly(ethylene glycol) (PEG) is widely utilized as a hydrophilic coating to extend the circulation time and improve the tumor accumulation of polymeric micelles. Nonetheless, PEGylated micelles often activate complement proteins, leading to accelerated blood clearance and negatively impacting drug efficacy and safety. Here, we have crafted amphiphilic block copolymers that merge hydrophilic sulfoxide-containing polymers (psulfoxides) with the hydrophobic drug 7-ethyl-10-hydroxylcamptothecin (SN38) into drug-conjugate micelles. Our findings show that the specific variant, PMSEA-PSN38 micelles, remarkably reduce protein fouling, prolong blood circulation, and improve intratumoral accumulation, culminating in significantly increased anti-cancer efficacy compared with PEG-PSN38 counterpart. Additionally, PMSEA-PSN38 micelles effectively inhibit complement activation, mitigate leukocyte uptake, and attenuate hyperactivation of inflammatory cells, diminishing their ability to stimulate tumor metastasis and cause inflammation. As a result, PMSEA-PSN38 micelles show exceptional promise in the realm of anti-metastasis and significantly abate SN38-induced intestinal toxicity. This study underscores the promising role of psulfoxides as viable PEG substitutes in the design of polymeric micelles for efficacious anti-cancer drug delivery.
Collapse
Affiliation(s)
- Yechun Wang
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Jiafeng Wang
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - JunJun Li
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Yongli Mu
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Jiajia Ying
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Zimeng Liu
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Mengjie Wu
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yu Geng
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Xuefei Zhou
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Tianhua Zhou
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Leimin Sun
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China; Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
| | - Xiangrui Liu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China.
| | - Quan Zhou
- Department of Cell Biology, and Department of Gastroenterology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
6
|
Hu M, Li X, You Z, Cai R, Chen C. Physiological Barriers and Strategies of Lipid-Based Nanoparticles for Nucleic Acid Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2303266. [PMID: 37792475 DOI: 10.1002/adma.202303266] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/21/2023] [Indexed: 10/06/2023]
Abstract
Lipid-based nanoparticles (LBNPs) are currently the most promising vehicles for nucleic acid drug (NAD) delivery. Although their clinical applications have achieved success, the NAD delivery efficiency and safety are still unsatisfactory, which are, to a large extent, due to the existence of multi-level physiological barriers in vivo. It is important to elucidate the interactions between these barriers and LBNPs, which will guide more rational design of efficient NAD vehicles with low adverse effects and facilitate broader applications of nucleic acid therapeutics. This review describes the obstacles and challenges of biological barriers to NAD delivery at systemic, organ, sub-organ, cellular, and subcellular levels. The strategies to overcome these barriers are comprehensively reviewed, mainly including physically/chemically engineering LBNPs and directly modifying physiological barriers by auxiliary treatments. Then the potentials and challenges for successful translation of these preclinical studies into the clinic are discussed. In the end, a forward look at the strategies on manipulating protein corona (PC) is addressed, which may pull off the trick of overcoming those physiological barriers and significantly improve the efficacy and safety of LBNP-based NADs delivery.
Collapse
Affiliation(s)
- Mingdi Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
- Sino-Danish Center for Education and Research, Beijing, 100049, China
| | - Xiaoyan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Zhen You
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Rong Cai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
- Sino-Danish Center for Education and Research, Beijing, 100049, China
- The GBA National Institute for Nanotechnology Innovation, Guangzhou, 510700, China
| |
Collapse
|
7
|
Ko T, Fumoto S, Kurosaki T, Nakashima M, Miyamoto H, Sasaki H, Nishida K. Interaction of γ-Polyglutamic Acid/Polyethyleneimine/Plasmid DNA Ternary Complexes with Serum Components Plays a Crucial Role in Transfection in Mice. Pharmaceutics 2024; 16:522. [PMID: 38675183 PMCID: PMC11053868 DOI: 10.3390/pharmaceutics16040522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/20/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
Typical examples of non-viral vectors are binary complexes of plasmid DNA with cationic polymers such as polyethyleneimine (PEI). However, problems such as cytotoxicity and hemagglutination, owing to their positively charged surfaces, hinder their in vivo use. Coating binary complexes with anionic polymers, such as γ-polyglutamic acid (γ-PGA), can prevent cytotoxicity and hemagglutination. However, the role of interactions between these complexes and serum components in in vivo gene transfer remains unclear. In this study, we analyzed the contribution of serum components to in vivo gene transfer using PEI/plasmid DNA binary complexes and γ-PGA/PEI/plasmid DNA ternary complexes. In binary complexes, heat-labile components in the serum greatly contribute to the hepatic and splenic gene expression of the luciferase gene. In contrast, serum albumin and salts affected the hepatic and splenic gene expression in the ternary complexes. Changes in physicochemical characteristics, such as increased particle size and decreased absolute values of ζ-potential, might be involved in the enhanced gene expression. These findings would contribute to a better understanding of in vivo non-viral gene transfer using polymers, such as PEI and γ-PGA.
Collapse
Affiliation(s)
- Tomotaka Ko
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Shintaro Fumoto
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Tomoaki Kurosaki
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Moe Nakashima
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Hirotaka Miyamoto
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Hitoshi Sasaki
- Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Koyo Nishida
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| |
Collapse
|
8
|
Müller JA, Schäffler N, Kellerer T, Schwake G, Ligon TS, Rädler JO. Kinetics of RNA-LNP delivery and protein expression. Eur J Pharm Biopharm 2024; 197:114222. [PMID: 38387850 DOI: 10.1016/j.ejpb.2024.114222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/23/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024]
Abstract
Lipid nanoparticles (LNPs) employing ionizable lipids are the most advanced technology for delivery of RNA, most notably mRNA, to cells. LNPs represent well-defined core-shell particles with efficient nucleic acid encapsulation, low immunogenicity and enhanced efficacy. While much is known about the structure and activity of LNPs, less attention is given to the timing of LNP uptake, cytosolic transfer and protein expression. However, LNP kinetics is a key factor determining delivery efficiency. Hence quantitative insight into the multi-cascaded pathway of LNPs is of interest to elucidate the mechanism of delivery. Here, we review experiments as well as theoretical modeling of the timing of LNP uptake, mRNA-release and protein expression. We describe LNP delivery as a sequence of stochastic transfer processes and review a mathematical model of subsequent protein translation from mRNA. We compile probabilities and numbers obtained from time resolved microscopy. Specifically, live-cell imaging on single cell arrays (LISCA) allows for high-throughput acquisition of thousands of individual GFP reporter expression time courses. The traces yield the distribution of mRNA life-times, expression rates and expression onset. Correlation analysis reveals an inverse dependence of gene expression efficiency and transfection onset-times. Finally, we discuss why timing of mRNA release is critical in the context of codelivery of multiple nucleic acid species as in the case of mRNA co-expression or CRISPR/Cas gene editing.
Collapse
Affiliation(s)
- Judith A Müller
- Faculty of Physics and Center for NanoScience, Ludwig Maximilians-University, Munich, Germany
| | - Nathalie Schäffler
- Faculty of Physics and Center for NanoScience, Ludwig Maximilians-University, Munich, Germany
| | - Thomas Kellerer
- Multiphoton Imaging Lab, Munich University of Applied Sciences, Munich, Germany
| | - Gerlinde Schwake
- Faculty of Physics and Center for NanoScience, Ludwig Maximilians-University, Munich, Germany
| | | | - Joachim O Rädler
- Faculty of Physics and Center for NanoScience, Ludwig Maximilians-University, Munich, Germany.
| |
Collapse
|
9
|
Espuche B, Moya SE, Calderón M. Nanogels: Smart tools to enlarge the therapeutic window of gene therapy. Int J Pharm 2024; 653:123864. [PMID: 38309484 DOI: 10.1016/j.ijpharm.2024.123864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
Gene therapy can potentially treat a great number of diseases, from cancer to rare genetic disorders. Very recently, the development and emergency approval of nucleic acid-based COVID-19 vaccines confirmed its strength and versatility. However, gene therapy encounters limitations due to the lack of suitable carriers to vectorize therapeutic genetic material inside target cells. Nanogels are highly hydrated nano-size crosslinked polymeric networks that have been used in many biomedical applications, from drug delivery to tissue engineering and diagnostics. Due to their easy production, tunability, and swelling properties they have called the attention as promising vectors for gene delivery. In this review, nanogels are discussed as vectors for nucleic acid delivery aiming to enlarge gene therapy's therapeutic window. Recent works highlighting the optimization of inherent transfection efficiency and biocompatibility are reviewed here. The importance of the monomer choice, along with the internal structure, surface decoration, and responsive features are outlined for the different transfection modalities. The possible sources of toxicological endpoints in nanogels are analyzed, and the strategies to limit them are compared. Finally, perspectives are discussed to identify the remining challenges for the nanogels before their translation to the market as transfection agents.
Collapse
Affiliation(s)
- Bruno Espuche
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain; POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain
| | - Sergio E Moya
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain.
| | - Marcelo Calderón
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain; IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain.
| |
Collapse
|
10
|
Su Y, Chen H, Liu Q, Ding X, Lian R, Hu Y, Xu FJ. Thermoresponsive Gels with Embedded Starch Microspheres for Optimized Antibacterial and Hemostatic Properties. ACS APPLIED MATERIALS & INTERFACES 2024; 16:12321-12331. [PMID: 38431875 DOI: 10.1021/acsami.3c19581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Apart from single hemostasis, antibacterial and other functionalities are also desirable for hemostatic materials to meet clinical needs. Cationic materials have attracted great interest for antibacterial/hemostatic applications, and it is still desirable to explore rational structure design to address the challenges in balanced hemostatic/antibacterial/biocompatible properties. In this work, a series of cationic microspheres (QMS) were prepared by the facile surface modification of microporous starch microspheres with a cationic tannic acid derivate, the coating contents of which were adopted for the first optimization of surface structure and property. Thermoresponsive gels with embedded QMS (F-QMS) were further prepared by mixing a neutral thermosensitive polymer and QMS for second structure/function optimization through different QMS and loading contents. In vitro and in vivo results confirmed that the coating content plays a crucial role in the hemostatic/antibacterial/biocompatible properties of QMS, but varied coating contents of QMS only lead to a classical imperfect performance of cationic materials. Inspiringly, the F-QMS-4 gel with an optimal loading content of QMS4 (with the highest coating content) achieved a superior balanced in vitro hemostatic/antibacterial/biocompatible properties, the mechanism of which was revealed as the second regulation of cell-material/protein-material interactions. Moreover, the optimal F-QMS-4 gel exhibited a high hemostatic performance in a femoral artery injury model accompanied by the easy on-demand removal for wound healing endowed by the thermoresponsive transformation. The present work offers a promising approach for the rational design and facile preparation of cationic materials with balanced hemostatic/antibacterial/biocompatible properties.
Collapse
Affiliation(s)
- Yang Su
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Hanlu Chen
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Qian Liu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Xiaokang Ding
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Rui Lian
- Emergency Department, China-Japan Friendship Hospital, Beijing 100029, P. R. China
| | - Yang Hu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Fu-Jian Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| |
Collapse
|
11
|
Wang X, Yuan K, Su Y, Li X, Meng L, Zhao N, Hu Y, Duan F, Xu FJ. Tuning Blood-Material Interactions to Generate Versatile Hemostatic Powders and Gels. Adv Healthc Mater 2024; 13:e2301945. [PMID: 37897223 DOI: 10.1002/adhm.202301945] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/05/2023] [Indexed: 10/29/2023]
Abstract
Polymer-based hemostatic materials/devices have been increasingly exploited for versatile clinical scenarios, while there is an urgent need to reveal the rational design/facile approach for procoagulant surfaces through regulating blood-material interactions. In this work, degradable powders (PLPS) and thermoresponsive gels (F127-PLPS) are readily developed as promising hemostatic materials for versatile clinical applications, through tuning blood-material interactions with optimized grafting of cationic polylysine: the former is facilely prepared by conjugating polylysine onto porous starch particle, while F127-PLPS is prepared by the simple mixture of PLPS and commercial thermosensitive polymer. In vitro and in vivo results demonstrate that PLPS2 with the optimal-/medium content of polylysine grafts achieve the superior hemostatic performance. The underlying procoagulant mechanism of PLPS2 surface is revealed as the selective fibrinogen adsorption among the competitive plasma-protein-adsorption process, which is the foundation of other blood-material interactions. Moreover, in vitro results confirm the achieved procoagulant surface of F127-PLPS through optimal PLPS2 loading. Together with the tunable thermoresponsiveness, F127-PLPS exhibits outstanding hemostatic utilization in both femoral-artery-injury and renal-artery-embolization models. The work thereby pioneers an appealing approach for generating versatile polymer-based hemostatic materials/devices.
Collapse
Affiliation(s)
- Xueru Wang
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Kai Yuan
- Department of Interventional Radiology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yang Su
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaoyue Li
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Limin Meng
- Department of Medical Imaging, Air Force Medical Center, PLA, Beijing, 100142, China
| | - Nana Zhao
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yang Hu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Feng Duan
- Department of Interventional Radiology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Fu-Jian Xu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
12
|
Hülsmann J, Lindemann H, Wegener J, Kühne M, Godmann M, Koschella A, Coldewey SM, Heinze T, Heinzel T. Dually Modified Cellulose as a Non-Viral Vector for the Delivery and Uptake of HDAC3 siRNA. Pharmaceutics 2023; 15:2659. [PMID: 38140000 PMCID: PMC10747125 DOI: 10.3390/pharmaceutics15122659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
RNA interference can be applied to different target genes for treating a variety of diseases, but an appropriate delivery system is necessary to ensure the transport of intact siRNAs to the site of action. In this study, cellulose was dually modified to create a non-viral vector for HDAC3 short interfering RNA (siRNA) transfer into cells. A guanidinium group introduced positive charges into the cellulose to allow complexation of negatively charged genetic material. Furthermore, a biotin group fixed by a polyethylene glycol (PEG) spacer was attached to the polymer to allow, if required, the binding of targeting ligands. The resulting polyplexes with HDAC3 siRNA had a size below 200 nm and a positive zeta potential of up to 15 mV. For N/P ratio 2 and higher, the polymer could efficiently complex siRNA. Nanoparticles, based on this dually modified derivative, revealed a low cytotoxicity. Only minor effects on the endothelial barrier integrity and a transfection efficiency in HEK293 cells higher than Lipofectamine 2000TM were found. The uptake and release of the polyplexes were confirmed by immunofluorescence imaging. This study indicates that the modified biopolymer is an auspicious biocompatible non-viral vector with biotin as a promising moiety.
Collapse
Affiliation(s)
- Juliana Hülsmann
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, Hans-Knöll-Straße 2, 07745 Jena, Germany; (J.H.); (M.K.); (M.G.)
| | - Henry Lindemann
- Institute for Organic Chemistry and Macromolecular Chemistry, Center of Excellence for Polysaccharide Research, Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany; (H.L.); (A.K.); (T.H.)
| | - Jamila Wegener
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (J.W.); (S.M.C.)
- Septomics Research Center, Jena University Hospital, Albert-Einstein-Straße 10, 07745 Jena, Germany
| | - Marie Kühne
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, Hans-Knöll-Straße 2, 07745 Jena, Germany; (J.H.); (M.K.); (M.G.)
| | - Maren Godmann
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, Hans-Knöll-Straße 2, 07745 Jena, Germany; (J.H.); (M.K.); (M.G.)
| | - Andreas Koschella
- Institute for Organic Chemistry and Macromolecular Chemistry, Center of Excellence for Polysaccharide Research, Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany; (H.L.); (A.K.); (T.H.)
| | - Sina M. Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (J.W.); (S.M.C.)
- Septomics Research Center, Jena University Hospital, Albert-Einstein-Straße 10, 07745 Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Thomas Heinze
- Institute for Organic Chemistry and Macromolecular Chemistry, Center of Excellence for Polysaccharide Research, Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany; (H.L.); (A.K.); (T.H.)
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Thorsten Heinzel
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, Hans-Knöll-Straße 2, 07745 Jena, Germany; (J.H.); (M.K.); (M.G.)
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| |
Collapse
|
13
|
Thakur S, Saini RV, Thakur N, Sharma R, Das J, Slama P, Tuli HS, Haque S, Niyazi HA, Moulay M, Harakeh S, Saini AK. Chitosan-PEI passivated carbon dots for plasmid DNA and miRNA-153 delivery in cancer cells. Heliyon 2023; 9:e21824. [PMID: 38034707 PMCID: PMC10682126 DOI: 10.1016/j.heliyon.2023.e21824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
These days carbon dots have been developed for multiple biomedical applications. In the current study, the transfection potential of synthesized carbon dots from single biopolymers such as chitosan, PEI-2kDa, and PEI-25kDa (CS-CDs, PEI2-CDs, and PEI25-CDs) and by combining two biopolymers (CP2-CDs and CP25-CDs) through a bottom-up approach have been investigated. The characterization studies revealed successful synthesis of fluorescent, positively charged carbon dots <20 nm in size. Synthesized carbon dots formed a stable complex with plasmid DNA (EGFP-N1) and miRNA-153 that protected DNA/miRNA from serum-induced degradation. In-vitro cytotoxicity analysis revealed minimal cytotoxicity in cancer cell lines (A549 and MDA-MB-231). In-vitro transfection of EGFP-N1 plasmid DNA with PEI2-CDs, PEI25-CDs and CP25-CDs demonstrated that these CDs could strongly transfect A549 and MDA-MB-231 cells. The highest EGFP-N1 plasmid transfection efficiency was observed with PEI2-CDs at a weight ratio of 32:1. PEI25-CDs polyplex showed maximum transfection at a weight ratio of 8:1 in A549 at a weight ratio of 16:1 in MDA-MB-231 cells. CP25-CDs exhibited the highest transfection at a weight ratio of 16:1 in both cell lines. The in-vitro transfection of target miRNA, i.e., miR-153 in A549 and MDA-MB-231 cells with PEI2-CDs, PEI25-CDs, and CP25-CDs suggested successful transfer of miR-153 into cells which induced significant cell death in both cell lines. Importantly, CS-CDs and CP2-CDs could be tolerated by cells up to 200 μg/mL concentration, while PEI2-CDs, PEI25-CDs, and CP25-CDs showed non-cytotoxic behavior at low concentrations (25 μg/mL). Together, these results suggest that a combination of carbon dots synthesized from chitosan and PEI (CP25-CDs) could be a novel vector for transfection nucleic acids that can be utilized in cancer therapy.
Collapse
Affiliation(s)
- Saloni Thakur
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, 173229, India
| | - Reena V. Saini
- Department of Biotechnology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, 133207, India
| | - Neelam Thakur
- School of Advance Chemical Sciences, Shoolini University, Solan, 173229, India
| | - Rohit Sharma
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, 173229, India
| | - Joydeep Das
- Department of Chemistry, Physical Sciences, Mizoram University, Aizawl, 796004, India
| | - Petr Slama
- Laboratory of Animal Immunology and Biotechnology, Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, 61300, Brno, Czech Republic
| | - Hardeep Singh Tuli
- Department of Biotechnology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, 133207, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Hatoon A. Niyazi
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Moulay
- Embryonic Stem Cells Research Unit, King Fahd Medical Research Center. King Abdul Aziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Steve Harakeh
- King Fahd Medical Research Center, and Yousef Abdullatif Jameel Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adesh K. Saini
- Department of Biotechnology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, 133207, India
| |
Collapse
|
14
|
Fornaguera C, Torres-Coll A, Olmo L, Garcia-Fernandez C, Guerra-Rebollo M, Borrós S. Engineering oncogene-targeted anisamide-functionalized pBAE nanoparticles as efficient lung cancer antisense therapies. RSC Adv 2023; 13:29986-30001. [PMID: 37842686 PMCID: PMC10573942 DOI: 10.1039/d3ra05830a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/04/2023] [Indexed: 10/17/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the leading causes of worldwide death, mainly due to the lack of efficient and safe therapies. Currently, NSCLC standard of care for consist on the use of traditional chemotherapeutics, non-selectively distributed through the whole body, thus causing severe side effects while not achieving high efficacy outcomes. Consequently, the need of novel therapies, targeted to modify specific subcellular routes aberrantly expressed only in tumor cells is still urgent. In this context, the delivery of siRNAs that can know-down overexpressed oncogenes, such as mTOR, could become the promised targeted therapy. However, siRNA effective delivery remains a challenge due to its compromised stability in biological fluids and its inability to cross biological and plasmatic membranes. Therefore, polymeric nanoparticles that efficiently encapsulate siRNAs and are selectively targeted to tumor cells could play a pivotal role. Accordingly, we demonstrate in this work that oligopeptide end-modified poly(beta aminoester) (OM-pBAE) polymers can efficiently complex siRNA in small nanometric particles using very low polymer amounts, protecting siRNA from nucleases attack. These nanoparticles are stable in the presence of serum, advantageous fact in terms of in vivo use. We also demonstrated that they efficiently transfect cells in vitro, in the presence of serum and are able to knock down target gene expression. Moreover, we demonstrated their antitumor efficacy by encapsulating mTOR siRNA, as a model antisense therapy, which showed specific lung tumor cell growth inhibition in vitro and in vivo. Finally, through the addition of anisamide functionalization to the surface of the nanoparticles, we proved that they become selective to lung tumor cells, while not affecting healthy cells. Therefore, our results are a first step in the discovery of a tumor cell-targeted efficient silencing nanotherapy for NSCLC patients survival improvement.
Collapse
Affiliation(s)
- Cristina Fornaguera
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL) Spain
| | - Antoni Torres-Coll
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL) Spain
| | - Laura Olmo
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL) Spain
| | - Coral Garcia-Fernandez
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL) Spain
| | - Marta Guerra-Rebollo
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL) Spain
| | - Salvador Borrós
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL) Spain
| |
Collapse
|
15
|
Jin Y, Adams F, Nguyen A, Sturm S, Carnerio S, Müller-Caspary K, Merkel OM. Synthesis and application of spermine-based amphiphilic poly(β-amino ester)s for siRNA delivery. NANOSCALE ADVANCES 2023; 5:5256-5262. [PMID: 37767040 PMCID: PMC10521211 DOI: 10.1039/d3na00272a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/16/2023] [Indexed: 09/29/2023]
Abstract
Small interfering RNA (siRNA) can trigger RNA interference (RNAi) to therapeutically silence disease-related genes in human cells. The approval of siRNA therapeutics by the FDA in recent years generated a new hope in novel and efficient siRNA therapeutics. However, their therapeutic application is still limited by the lack of safe and efficient transfection vehicles. In this study, we successfully synthesized a novel amphiphilic poly(β-amino ester) based on the polyamine spermine, hydrophobic decylamine and 1,4-butanediol diacrylate, which was characterized by 1H NMR spectroscopy and size exclusion chromatography (SEC, Mn = 6000 Da). The polymer encapsulated siRNA quantitatively from N/P 5 on as assessed by fluorescence intercalation while maintaining optimal polyplex sizes and zeta potentials. Biocompatibility and cellular delivery efficacy were also higher than those of the commonly used cationic, hyperbranched polymer polyethylenimine (PEI, 25 kDa). Optimized formulations mediated around 90% gene silencing in enhanced green fluorescence protein expressing H1299 cells (H1299-eGFP) as determined by flow cytometry. These results suggest that spermine-based, amphiphilic poly(β-amino ester)s are very promising candidates for efficient siRNA delivery.
Collapse
Affiliation(s)
- Yao Jin
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics Butenandtstr. 5-13 81377 Munich Germany
| | - Friederike Adams
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics Butenandtstr. 5-13 81377 Munich Germany
| | - Anny Nguyen
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics Butenandtstr. 5-13 81377 Munich Germany
| | - Sebastian Sturm
- Department of Chemistry and Centre for NanoScience, Ludwig-Maximilians-University Munich Butenandtstr. 11 81377 Munich Germany
| | - Simone Carnerio
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics Butenandtstr. 5-13 81377 Munich Germany
| | - Knut Müller-Caspary
- Department of Chemistry and Centre for NanoScience, Ludwig-Maximilians-University Munich Butenandtstr. 11 81377 Munich Germany
| | - Olivia M Merkel
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Pharmaceutical Technology and Biopharmaceutics Butenandtstr. 5-13 81377 Munich Germany
| |
Collapse
|
16
|
Halbi G, Fayer I, Aranovich D, Gat S, Pavan MJ, Nachmias D, Sanchez DS, Brik A, Granek R, Bernheim-Groswasser A. Smart design of universally decorated nanoparticles for drug delivery applications driven by active transport. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2023; 46:74. [PMID: 37653248 DOI: 10.1140/epje/s10189-023-00331-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/25/2023] [Indexed: 09/02/2023]
Abstract
Targeting the cell nucleus remains a challenge for drug delivery. Here, we present a universal platform for the smart design of nanoparticle (NP) decoration that is based on: (i) a spacer polymer, commonly biotin-polyethylene-glycol-thiol, whose grafting density and molecular weight can be tuned for optimized performance, and (ii) protein binding peptides, such as cell penetrating peptides (CPPs), cancer-targeting peptides, or nuclear localization signal (NLS) peptides, that are linked to the PEG free-end by universal chemistry. We manifested our platform with two different bromo-acetamide (Br-Ac) modified NLSs. We used cell extract-based and live cell assays to demonstrate the recruitment of dynein motor proteins, which drive the NP active transport toward the nucleus, and the enhancement of cellular and nuclear entry, manifesting the properties of NLS as a CPP. Our control of the NP decoration scheme, and the modularity of our platform, carry great advantages for nano-carrier design for drug delivery applications.
Collapse
Affiliation(s)
- Gal Halbi
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Itay Fayer
- The Stella and Avram Goren-Goldstein Department of Biotechnology Engineering Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Dina Aranovich
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Shachar Gat
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Mariela J Pavan
- The Ilse Katz Institute for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Dikla Nachmias
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Daniel Sevilla Sanchez
- The Ilse Katz Institute for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Ashraf Brik
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, 3200008, Haifa, Israel
| | - Rony Granek
- The Stella and Avram Goren-Goldstein Department of Biotechnology Engineering Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
- The Ilse Katz Institute for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Anne Bernheim-Groswasser
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel.
- The Ilse Katz Institute for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel.
| |
Collapse
|
17
|
Hartl N, Gabold B, Adams F, Uhl P, Oerter S, Gätzner S, Metzger M, König AC, Hauck SM, Appelt-Menzel A, Mier W, Fricker G, Merkel OM. Overcoming the blood-brain barrier? - prediction of blood-brain permeability of hydrophobically modified polyethylenimine polyplexes for siRNA delivery into the brain with in vitro and in vivo models. J Control Release 2023; 360:613-629. [PMID: 37437848 DOI: 10.1016/j.jconrel.2023.07.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/23/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
The blood-brain barrier (BBB) is a highly selective biological barrier that represents a major bottleneck in the treatment of all types of central nervous system (CNS) disorders. Small interfering RNA (siRNA) offers in principle a promising therapeutic approach, e.g., for brain tumors, by downregulating brain tumor-related genes and inhibiting tumor growth via RNA interference. In an effort to develop efficient siRNA nanocarriers for crossing the BBB, we utilized polyethyleneimine (PEI) polymers hydrophobically modified with either stearic-acid (SA) or dodecylacrylamide (DAA) subunits and evaluated their suitability for delivering siRNA across the BBB in in vitro and in vivo BBB models depending on their structure. Physicochemical characteristics of siRNA-polymer complexes (polyplexes (PXs)), e.g., particle size and surface charge, were measured by dynamic light scattering and laser Doppler anemometry, whereas siRNA condensation ability of polymers and polyplex stability was evaluated by spectrophotometric methods. The composition of the biomolecule corona that absorbs on polyplexes upon encountering physiological fluids was investigated by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and by a liquid chromatography-tandem mass spectrometry (LC-MS-MS) method. Cellular internalization abilities of PXs into brain endothelial cells (hCMEC/D3) was confirmed, and a BBB permeation assay using a human induced pluripotent stem cell (hiPSC)-derived BBB model revealed similar abilities to cross the BBB for all formulations under physiological conditions. However, biodistribution studies of radiolabeled PXs in mice were inconsistent with in vitro results as the detected amount of radiolabeled siRNA in the brain delivered with PEI PXs was higher compared to PEI-SA PXs. Taken together, PEI PXs were shown to be a suitable nanocarrier to deliver small amounts of siRNA across the BBB into the brain but more sophisticated human BBB models that better represent physiological conditions and biodistribution are required to provide highly predictive in vitro data for human CNS drug development in the future.
Collapse
Affiliation(s)
- Natascha Hartl
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Bettina Gabold
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Friederike Adams
- University of Stuttgart, Institute of Polymer Chemistry, Macromolecular Materials and Fiber Chemistry, Pfaffenwaldring 55, 70569 Stuttgart, Germany
| | - Philipp Uhl
- University Hospital Heidelberg, Department of Nuclear Medicine, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Sabrina Oerter
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany; University Hospital Würzburg, Chair of Tissue Engineering and Regenerative Medicine (TERM), 97070 Würzburg, Germany
| | - Sabine Gätzner
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany
| | - Marco Metzger
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany; University Hospital Würzburg, Chair of Tissue Engineering and Regenerative Medicine (TERM), 97070 Würzburg, Germany
| | - Ann-Christine König
- Helmholtz Centrum Munich - German Research Center for Environmental Health, Research Unit Protein Science, Heidemannsstr. 1, 80939, Munich, Germany
| | - Stefanie M Hauck
- Helmholtz Centrum Munich - German Research Center for Environmental Health, Research Unit Protein Science, Heidemannsstr. 1, 80939, Munich, Germany
| | - Antje Appelt-Menzel
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany; University Hospital Würzburg, Chair of Tissue Engineering and Regenerative Medicine (TERM), 97070 Würzburg, Germany
| | - Walter Mier
- University Hospital Heidelberg, Department of Nuclear Medicine, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Gert Fricker
- University of Heidelberg, Institute for Pharmacy & Molekular Biotechnology, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| | - Olivia M Merkel
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany.
| |
Collapse
|
18
|
Budiarta M, Roy S, Katenkamp T, Feliu N, Beck T. Overcoming Non-Specific Interactions for Efficient Encapsulation of Doxorubicin in Ferritin Nanocages for Targeted Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205606. [PMID: 36748864 DOI: 10.1002/smll.202205606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/22/2022] [Indexed: 05/25/2023]
Abstract
Due to its beneficial pharmacological properties, ferritin (Ftn) is considered as an interesting drug delivery vehicle to alleviate the cardiotoxicity of doxorubicin (DOX) in chemotherapy. However, the encapsulation of DOX in Ftn suffers from heavy precipitation and low protein recovery yield which limits its full potential. Here, a new DOX encapsulation strategy by cysteine-maleimide conjugation is proposed. In order to demonstrate that this strategy is more efficient compared to the other approaches, DOX is encapsulated in Ftn variants carrying different surface charges. Furthermore, in contrast to the common belief, this data show that DOX molecules are also found to bind non-specifically to the surface of Ftn. This can be circumvented by the use of Tris(2-carboxyethyl)phosphine (TCEP) during encapsulation or by washing with acidic buffer. The biocompatibility studies of the resulting DOX Ftn variants in MCF-7 and MHS cancer cells shows a complex relationship between the cytotoxicity, the DOX loading and the different surface charges of Ftn. Further investigation on the cell uptake mechanism provides reasonable explanations for the cytotoxicity results and reveals that surface charging of Ftn hinders its transferrin receptor 1 (TfR-1) mediated cellular uptake in MCF-7 cells.
Collapse
Affiliation(s)
- Made Budiarta
- Institute of Inorganic Chemistry, RWTH Aachen University, Landoltweg 1, 52074, Aachen, Germany
| | - Sathi Roy
- Fraunhofer Center for Applied Nanotechnology (CAN), Fraunhofer IAP, Grindelallee 117, 20146, Hamburg, Germany
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22607, Hamburg, Germany
| | - Tobias Katenkamp
- Institute of Physical Chemistry, Department of Chemistry, Universität Hamburg, Grindelallee 117, 20146, Hamburg, Germany
| | - Neus Feliu
- Fraunhofer Center for Applied Nanotechnology (CAN), Fraunhofer IAP, Grindelallee 117, 20146, Hamburg, Germany
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22607, Hamburg, Germany
| | - Tobias Beck
- Hamburg Centre for Ultrafast Imaging, Universität Hamburg, Luruper Chaussee 149, 22761, Hamburg, Germany
| |
Collapse
|
19
|
Fang T, Cao X, Ibnat M, Chen G. Stimuli-responsive nanoformulations for CRISPR-Cas9 genome editing. J Nanobiotechnology 2022; 20:354. [PMID: 35918694 PMCID: PMC9344766 DOI: 10.1186/s12951-022-01570-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 07/22/2022] [Indexed: 12/07/2022] Open
Abstract
The CRISPR-Cas9 technology has changed the landscape of genome editing and has demonstrated extraordinary potential for treating otherwise incurable diseases. Engineering strategies to enable efficient intracellular delivery of CRISPR-Cas9 components has been a central theme for broadening the impact of the CRISPR-Cas9 technology. Various non-viral delivery systems for CRISPR-Cas9 have been investigated given their favorable safety profiles over viral systems. Many recent efforts have been focused on the development of stimuli-responsive non-viral CRISPR-Cas9 delivery systems, with the goal of achieving efficient and precise genome editing. Stimuli-responsive nanoplatforms are capable of sensing and responding to particular triggers, such as innate biological cues and external stimuli, for controlled CRISPR-Cas9 genome editing. In this Review, we overview the recent advances in stimuli-responsive nanoformulations for CRISPR-Cas9 delivery, highlight the rationale of stimuli and formulation designs, and summarize their biomedical applications.
Collapse
Affiliation(s)
- Tianxu Fang
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada.,Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Xiaona Cao
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada.,Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada.,School of Nursing, Tianjin Medical University, Tianjin, China
| | - Mysha Ibnat
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada.,Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada. .,Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada.
| |
Collapse
|
20
|
Meng L, Yang F, Pang Y, Cao Z, Wu F, Yan D, Liu J. Nanocapping-enabled charge reversal generates cell-enterable endosomal-escapable bacteriophages for intracellular pathogen inhibition. SCIENCE ADVANCES 2022; 8:eabq2005. [PMID: 35857522 PMCID: PMC11581130 DOI: 10.1126/sciadv.abq2005] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/28/2022] [Indexed: 06/15/2023]
Abstract
Bacteriophages (phages) are widely explored as antimicrobials for treating infectious diseases due to their specificity and potency to infect and inhibit host bacteria. However, the application of phages to inhibit intracellular pathogens has been greatly restricted by inadequacy in cell entry and endosomal escape. Here, we describe the use of cationic polymers to selectively cap negatively charged phage head rather than positively charged tail by electrostatic interaction, resulting in charge-reversed phages with uninfluenced vitality. Given the positive surface charge and proton sponge effect of the nanocapping, capped phages are able to enter intestinal epithelial cells and subsequently escape from endosomes to lyse harbored pathogens. In a murine model of intestinal infection, oral ingestion of capped phages significantly reduces the translocation of pathogens to major organs, showing a remarkable inhibition efficacy. Our work proposes that simple synthetic nanocapping can manipulate phage bioactivity, offering a facile platform for preparing next-generation antimicrobials.
Collapse
Affiliation(s)
- Lu Meng
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fengmin Yang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yan Pang
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Zhenping Cao
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Feng Wu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Deyue Yan
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
21
|
Costa B, Boueri B, Oliveira C, Silveira I, Ribeiro AJ. Lipoplexes and polyplexes as nucleic acids delivery nanosystems: The current state and future considerations. Expert Opin Drug Deliv 2022; 19:577-594. [DOI: 10.1080/17425247.2022.2075846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Bruno Costa
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Beatriz Boueri
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Claudia Oliveira
- Group Genetics of Cognitive Dysfunction, IBMC - Instituto de Biologia Molecular e Celular, I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Isabel Silveira
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Group Genetics of Cognitive Dysfunction, IBMC - Instituto de Biologia Molecular e Celular, I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Antonio J. Ribeiro
- University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Group Genetics of Cognitive Dysfunction, IBMC - Instituto de Biologia Molecular e Celular, I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| |
Collapse
|
22
|
Jerzykiewicz J, Czogalla A. Polyethyleneimine-Based Lipopolyplexes as Carriers in Anticancer Gene Therapies. MATERIALS 2021; 15:ma15010179. [PMID: 35009324 PMCID: PMC8746209 DOI: 10.3390/ma15010179] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/06/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022]
Abstract
Recent years have witnessed rapidly growing interest in application of gene therapies for cancer treatment. However, this strategy requires nucleic acid carriers that are both effective and safe. In this context, non-viral vectors have advantages over their viral counterparts. In particular, lipopolyplexes—nanocomplexes consisting of nucleic acids condensed with polyvalent molecules and enclosed in lipid vesicles—currently offer great promise. In this article, we briefly review the major aspects of developing such non-viral vectors based on polyethyleneimine and outline their properties in light of anticancer therapeutic strategies. Finally, examples of current in vivo studies involving such lipopolyplexes and possibilities for their future development are presented.
Collapse
|
23
|
Yan H, Cacioppo M, Megahed S, Arcudi F, Đorđević L, Zhu D, Schulz F, Prato M, Parak WJ, Feliu N. Influence of the chirality of carbon nanodots on their interaction with proteins and cells. Nat Commun 2021; 12:7208. [PMID: 34893594 PMCID: PMC8664908 DOI: 10.1038/s41467-021-27406-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 11/11/2021] [Indexed: 12/26/2022] Open
Abstract
Carbon nanodots with opposite chirality possess the same major physicochemical properties such as optical features, hydrodynamic diameter, and colloidal stability. Here, a detailed analysis about the comparison of the concentration of both carbon nanodots is carried out, putting a threshold to when differences in biological behavior may be related to chirality and may exclude effects based merely on differences in exposure concentrations due to uncertainties in concentration determination. The present study approaches this comparative analysis evaluating two basic biological phenomena, the protein adsorption and cell internalization. We find how a meticulous concentration error estimation enables the evaluation of the differences in biological effects related to chirality.
Collapse
Affiliation(s)
- Huijie Yan
- Fachbereich Physik, Center for Hybrid Nanostructures (CHyN), Universitat Hamburg, 22607, Hamburg, Germany
| | - Michele Cacioppo
- Fachbereich Physik, Center for Hybrid Nanostructures (CHyN), Universitat Hamburg, 22607, Hamburg, Germany
- Department of Chemical and Pharmaceutical Sciences, INSTM UdR Trieste, University of Trieste, Via Licio Giorgieri 1, 34127, Trieste, Italy
| | - Saad Megahed
- Fachbereich Physik, Center for Hybrid Nanostructures (CHyN), Universitat Hamburg, 22607, Hamburg, Germany
- Physics Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Francesca Arcudi
- Department of Chemical and Pharmaceutical Sciences, INSTM UdR Trieste, University of Trieste, Via Licio Giorgieri 1, 34127, Trieste, Italy
| | - Luka Đorđević
- Department of Chemical and Pharmaceutical Sciences, INSTM UdR Trieste, University of Trieste, Via Licio Giorgieri 1, 34127, Trieste, Italy
| | - Dingcheng Zhu
- Fachbereich Physik, Center for Hybrid Nanostructures (CHyN), Universitat Hamburg, 22607, Hamburg, Germany
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou, PR China
| | - Florian Schulz
- Fachbereich Physik, Center for Hybrid Nanostructures (CHyN), Universitat Hamburg, 22607, Hamburg, Germany
| | - Maurizio Prato
- Department of Chemical and Pharmaceutical Sciences, INSTM UdR Trieste, University of Trieste, Via Licio Giorgieri 1, 34127, Trieste, Italy.
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 182, 20014, Donostia San Sebastian, Spain.
- Basque Foundation for Science, Ikerbasque, 48013, Bilbao, Spain.
| | - Wolfgang J Parak
- Fachbereich Physik, Center for Hybrid Nanostructures (CHyN), Universitat Hamburg, 22607, Hamburg, Germany.
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 182, 20014, Donostia San Sebastian, Spain.
| | - Neus Feliu
- Fachbereich Physik, Center for Hybrid Nanostructures (CHyN), Universitat Hamburg, 22607, Hamburg, Germany.
- Fraunhofer Center for Applied Nanotechnology (CAN), 20146, Hamburg, Germany.
| |
Collapse
|
24
|
Wang X, Deng B, Yu M, Zeng T, Chen Y, Hu J, Wu Q, Li A. Constructing a passive targeting and long retention therapeutic nanoplatform based on water-soluble, non-toxic and highly-stable core-shell poly(amino acid) nanocomplexes. Biomater Sci 2021; 9:7065-7075. [PMID: 34590101 DOI: 10.1039/d1bm01246k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Drug delivery nanoplatforms have been applied in bioimaging, medical diagnosis, drug delivery and medical therapy. However, insolubility, toxicity, instability, nonspecific targeting and short retention of many hydrophobic drugs limit their extensive applications. Herein, we have constructed a passive targeting and long retention therapeutic nanoplatform of core-shell gefitinib/poly (ethylene glycol)-polytyrosine nanocomplexes (Gef-PY NCs). The Gef-PY NCs have good water-solubility, non-toxicity (correspond to 1/10 dosage of effective gefitinib (hydrochloride) (Gef·HCl) (normal drug administration and slow-release) and high stability (120 days, 80% drug retention at 4 or 25 °C). The core-shell Gef-PY NCs present unexpected kidney targeting and drug slow-release capacity (ca. 72 h). The good water-solubility, non-toxicity and high stability of Gef-PY NCs effectively solve the bottleneck question that Gef-based therapy could be used only in intraperitoneal injection due to its insolubility and severe toxicity. Such excellent properties (e.g., water-solubility, non-toxicity, high stability, kidney targeting and long retention) of Gef-PY NCs create their prominent anti-fibrosis capabilities, such as decreasing approximately 40% tubulointerstitial fibrosis area and 68% expression of collagen I within 7 days. This therapeutic efficacy is well-matched with that of 10 times the dosage of toxic Gef·HCl. It is very hopeful that Gef-PY NCs could realize clinical applications and such a strategy offers an effective route to design high-efficiency treatments for kidney- and tumor-related diseases.
Collapse
Affiliation(s)
- Xin Wang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Bingqing Deng
- Nanobiological Medicine Center, Key Lab of Fuel Cell Technology of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Meng Yu
- Nanobiological Medicine Center, Key Lab of Fuel Cell Technology of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Tao Zeng
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yuyu Chen
- Nanobiological Medicine Center, Key Lab of Fuel Cell Technology of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Jianqiang Hu
- Nanobiological Medicine Center, Key Lab of Fuel Cell Technology of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Qianqing Wu
- Nanobiological Medicine Center, Key Lab of Fuel Cell Technology of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Aiqing Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
25
|
Smolková B, MacCulloch T, Rockwood TF, Liu M, Henry SJW, Frtús A, Uzhytchak M, Lunova M, Hof M, Jurkiewicz P, Dejneka A, Stephanopoulos N, Lunov O. Protein Corona Inhibits Endosomal Escape of Functionalized DNA Nanostructures in Living Cells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:46375-46390. [PMID: 34569777 PMCID: PMC9590277 DOI: 10.1021/acsami.1c14401] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
DNA nanostructures (DNs) can be designed in a controlled and programmable manner, and these structures are increasingly used in a variety of biomedical applications, such as the delivery of therapeutic agents. When exposed to biological liquids, most nanomaterials become covered by a protein corona, which in turn modulates their cellular uptake and the biological response they elicit. However, the interplay between living cells and designed DNs are still not well established. Namely, there are very limited studies that assess protein corona impact on DN biological activity. Here, we analyzed the uptake of functionalized DNs in three distinct hepatic cell lines. Our analysis indicates that cellular uptake is linearly dependent on the cell size. Further, we show that the protein corona determines the endolysosomal vesicle escape efficiency of DNs coated with an endosome escape peptide. Our study offers an important basis for future optimization of DNs as delivery systems for various biomedical applications.
Collapse
Affiliation(s)
- Barbora Smolková
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18221, Czech Republic
| | - Tara MacCulloch
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85287, United States
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Tyler F Rockwood
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85287, United States
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Minghui Liu
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85287, United States
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Skylar J W Henry
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85287, United States
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Adam Frtús
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18221, Czech Republic
| | - Mariia Uzhytchak
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18221, Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18221, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), Prague 14021, Czech Republic
| | - Martin Hof
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague 18223, Czech Republic
| | - Piotr Jurkiewicz
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague 18223, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18221, Czech Republic
| | - Nicholas Stephanopoulos
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85287, United States
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague 18221, Czech Republic
| |
Collapse
|
26
|
Kim J, Vaughan HJ, Zamboni CG, Sunshine JC, Green JJ. High-throughput evaluation of polymeric nanoparticles for tissue-targeted gene expression using barcoded plasmid DNA. J Control Release 2021; 337:105-116. [PMID: 34097924 DOI: 10.1016/j.jconrel.2021.05.047] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 05/03/2021] [Accepted: 05/31/2021] [Indexed: 12/17/2022]
Abstract
Successful systemic gene delivery requires specific tissue targeting as well as efficient intracellular transfection. Increasingly, research laboratories are fabricating libraries of novel nanoparticles, engineering both new biomaterial structures and composition ratios of multicomponent systems. Yet, methods for screening gene delivery vehicles directly in vivo are often low-throughout, limiting the number of candidate nanoparticles that can be investigated. Here, we report a comprehensive, high-throughput method to evaluate a library of polymeric nanoparticles in vivo for tissue-specific gene delivery. The method involves pairing each nanoparticle formulation with a plasmid DNA (pDNA) that harbors a unique nucleotide sequence serving as the identifying "barcode". Using real time quantitative PCR (qPCR) for detection of the barcoded pDNA and quantitative reverse transcription PCR (RT-qPCR) for transcribed barcoded mRNA, we can quantify accumulation and transfection in tissues of interest. The barcode pDNA and primers were designed with sufficient sensitivity and specificity to evaluate multiple nanoparticle formulations per mouse, improving screening efficiency. Using this platform, we evaluated the biodistribution and transfection of 8 intravenously administered poly(beta-amino ester; PBAE) nanoparticle formulations, each with a PBAE polymer of differential structure. Significant levels of nanoparticle accumulation and gene transfection were observed mainly in organs involved in clearance, including spleen, liver, and kidneys. Interestingly, higher levels of transfection of select organs did not necessarily correlate with higher levels of tissue accumulation, highlighting the importance of directly measuring in vivo transfection efficiency as the key barcoded parameter in gene delivery vector optimization. To validate this method, nanoparticle formulations were used individually for luciferase pDNA delivery in vivo. The distribution of luciferase expression in tissues matched the transfection analysis by the barcode qPCR method, confirming that this platform can be used to accurately evaluate systemic gene delivery.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Hannah J Vaughan
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Camila G Zamboni
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Joel C Sunshine
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Jordan J Green
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA; Departments of Neurosurgery, Oncology, and Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
27
|
Zhu D, Yan H, Zhou Z, Tang J, Liu X, Hartmann R, Parak WJ, Shen Y, Feliu N. Influence of the Modulation of the Protein Corona on Gene Expression Using Polyethylenimine (PEI) Polyplexes as Delivery Vehicle. Adv Healthc Mater 2021; 10:e2100125. [PMID: 34086423 PMCID: PMC11469282 DOI: 10.1002/adhm.202100125] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/20/2021] [Indexed: 12/16/2022]
Abstract
The protein corona can significantly modulate the physicochemical properties and gene delivery of polyethylenimine (PEI)/DNA complexes (polyplexes). The effects of the protein corona on the transfection have been well studied in terms of averaged gene expression in a whole cell population. Such evaluation methods give excellent and reliable statistics, but they in general provide the final transfection efficiency without reflecting the dynamic process of gene expression. In this regard the influence of bovine serum albumin (BSA) on the gene expression of PEI polyplexes also on a single cell level via live imaging is analyzed. The results reveal that although the BSA corona causes difference in the overall gene expression and mRNA transcription, the gene expression behavior on the level of individual cell is similar, including the mitosis-dependent expression, distributions of onset time, expression pattern in two daughter cells, and expression kinetics in successfully transfected cells. Comparison of single cell and ensemble data on whole cell cultures indicate that the protein corona does not alter the transfection process after nuclear entry, including cell division, polyplex dissociation, and protein expression. Its influence on other steps of in vitro gene delivery before nuclear entry shall render the difference in the overall transfection.
Collapse
Affiliation(s)
- Dingcheng Zhu
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityZheda road 38Hangzhou310007China
- Fachbereich Physik und Chemie and CHyNUniversität HamburgNotkestraße 85Hamburg22607Germany
- College of Material, Chemistry and Chemical EngineeringHangzhou Normal UniversityHaishu road 58Hangzhou310000China
| | - Huijie Yan
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityZheda road 38Hangzhou310007China
- Fachbereich Physik und Chemie and CHyNUniversität HamburgNotkestraße 85Hamburg22607Germany
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityZheda road 38Hangzhou310007China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityZheda road 38Hangzhou310007China
| | - Xiangrui Liu
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityZheda road 38Hangzhou310007China
| | - Raimo Hartmann
- Fachbereich PhysikPhilipps Universität MarburgRenthof 6Marburg35032Germany
| | - Wolfgang J. Parak
- Fachbereich Physik und Chemie and CHyNUniversität HamburgNotkestraße 85Hamburg22607Germany
- CIC biomaGUNEMiramon Pasealekua 182San Sebastian20014Spain
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityZheda road 38Hangzhou310007China
| | - Neus Feliu
- Fachbereich Physik und Chemie and CHyNUniversität HamburgNotkestraße 85Hamburg22607Germany
- Fraunhofer Center for Applied Nanotechnology (CAN)Grindelallee 117Hamburg20146Germany
| |
Collapse
|
28
|
Ahmad A, Ansari MM, Verma RK, Khan R. Aminocellulose-Grafted Polymeric Nanoparticles for Selective Targeting of CHEK2-Deficient Colorectal Cancer. ACS APPLIED BIO MATERIALS 2021; 4:5324-5335. [PMID: 35007013 DOI: 10.1021/acsabm.1c00437] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
We report the formulation of aminocellulose-grafted polymeric nanoparticles containing LCS-1 for synthetic lethal targeting of checkpoint kinase 2 (CHEK2)-deficient HCT116 colon cancer (CRC) cells to surpass the limitations associated with the solubility of LCS-1 (a superoxide dismutase inhibitor). Aminocellulose (AC), a highly biocompatible and biodegradable hydrophilic polymer, was grafted over polycaprolactone (PCL), and a nanoprecipitation method was employed for formulating nanoparticles containing LCS-1. In this study, we exploited the synthetic lethal interaction between SOD1 and CHEK2 for the specific inhibition of CHEK2-deficient HCT116 CRC cells using LCS-1-loaded PCL-AC NPs. Furthermore, the effects of formation of protein corona on PCL-AC nanoparticles were also assessed in terms of size, cellular uptake, and cell viability. LCS-1-loaded NPs were evaluated for their size, zeta potential, and polydispersity index using a zetasizer, and their morphological characteristics were assessed by transmission electron microscopy, scanning electron microscopy, and atomic force microscopy analyses. Cellular internalization using confocal microscopy exhibited that nanoparticles were uptaken by HCT116 cells. Also, nanoparticles were cytocompatible as they did not induce cytotoxicity in hTERT and HEK-293 cells. The LCS-1-loaded PCL-AC NPs were quite hemocompatible and were 240 times more selective in killing CHEK2-deficient cells as compared to CHEK2-proficient CRC cells. Moreover, PCL-AC NPs exhibited that the protein corona-coated nanoparticles were incubated in the human and fetal bovine sera as visualized by SDS-PAGE. A slight increment in hydrodynamic diameter was observed for corona-coated PCL-AC nanoparticles, and size increment was further confirmed by TEM. Corona-coated PCL-AC NPs also exhibited cellular uptake as demonstrated by flow cytometric analysis and did not cause cytotoxic effects on hTERT cells. The nanoformulation was developed to enhance therapeutic potential of the drug LCS-1 for enhanced lethality of colorectal cancer cells with CHEK2 deficiency.
Collapse
Affiliation(s)
- Anas Ahmad
- Institute of Nano Science and Technology, Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Punjab 140306, India.,Department of Pharmacology, Chandigarh College of Pharmacy, Sector 112, Sahibzada Ajit Singh Nagar, Punjab 140307, India
| | - Md Meraj Ansari
- Institute of Nano Science and Technology, Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Punjab 140306, India
| | - Rahul Kumar Verma
- Institute of Nano Science and Technology, Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Punjab 140306, India
| | - Rehan Khan
- Institute of Nano Science and Technology, Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Punjab 140306, India
| |
Collapse
|
29
|
Zhang Z, Qiu N, Wu S, Liu X, Zhou Z, Tang J, Liu Y, Zhou R, Shen Y. Dose-Independent Transfection of Hydrophobized Polyplexes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102219. [PMID: 33991017 DOI: 10.1002/adma.202102219] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/09/2021] [Indexed: 05/14/2023]
Abstract
Cationic polymers dynamically complex DNA into complexes (polyplexes). So, upon dilution, polyplexes easily dissociate and lose transfection ability, limiting their in vivo systemic gene delivery. Herein, it is found that polyplex's stability and endocytosis pathway determine its transfection dose-dependence. The polyplexes of hydrophilic polycations have dose-dependent integrity and lysosome-trafficking endocytosis; at low doses, most of these polyplexes dissociate, and the remaining few are internalized and trapped in lysosomes, abolishing their transfection ability. In contrast, the polyplexes of the polycations with optimal hydrophobicity remain integrated even at low concentrations and enter cells via macropinocytosis directly into the cytosol evading lysosomes, so each polyplex can accomplish its infection process, leading to dose-independent DNA transfection like viral vectors. Furthermore, the tuned hydrophobicity balancing the affinity of anionic poly(γ-glutamic acid) (γ-PGA) to the polyplex surface enables γ-PGA to stick on the polyplex surface as a shielding layer but peel off on the cell membrane to release the naked polyplexes for dose-independent transfection. These findings may provide guidelines for developing polyplexes that mimick a viral vector's dose-independent transfection for effective in vivo gene delivery.
Collapse
Affiliation(s)
- Zhen Zhang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Shuling Wu
- Department of Respiratory, The First People's Hospital of Xiaoshan, Hangzhou, 311200, China
| | - Xin Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
- Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yanpeng Liu
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
- Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, China
| | - Ruhong Zhou
- Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
- Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215, China
| |
Collapse
|
30
|
Yang Q, Dong Y, Wang X, Lin Z, Yan M, Wang W, Dong A, Zhang J, Huang P, Wang C. pH-Sensitive Polycations for siRNA Delivery: Effect of Asymmetric Structures of Tertiary Amine Groups. Macromol Biosci 2021; 21:e2100025. [PMID: 33769670 DOI: 10.1002/mabi.202100025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/07/2021] [Indexed: 12/13/2022]
Abstract
pH-sensitive polyelectrolytes provide enormous opportunity for siRNA delivery. Especially, their tertiary amine structures can not only bind genes but also act as pH-sensitive hydrophobic structure to control genes release. However, the influence of molecular structures on siRNA delivery still remains elusive, especially for the asymmetric alkyl substituents of the tertiary amine groups. Herein, a library of N-methyl-N-alkyl aminoethyl methacrylate monomers (MsAM) with asymmetric alkyl substituents on the tertiary amine group is synthesized and used to prepare a series of tri-block polycationic copolymers poly(aminoethyl methacrylate)-block-poly (N-methyl-N-alkyl aminoethyl methacrylate)-block-poly(ethylene glycol methacrylate) (PAMA-PMsMA-PEG). And the properties of these polycations and their self-assembled micelles are characterized, including molecular structure, proton buffering capacity, pH-sensitivity, size, and zeta potential. With the length increase of one alkyl substituent, the proton buffering capacity of both monomers and polycations is demonstrated to be narrowed down. The siRNA delivery efficiency and cytotoxicity of these micelles are also evaluated on HepG2 cells. In particular, poly(aminoethyl methacrylate)-block-poly(N-methyl-N-ethyl aminoethyl methacrylate)-block-poly(ethylene glycol methacrylate) (PAMA-PMEMA-PEG) elicited the best luciferase knockdown efficiency and low cytotoxicity. Besides, PAMA-PMEMA-PEG/siRRM2 also induced significant anti-tumor activity in vitro. These results indicated PAMA-PMEMA-PEG has potential for further use in the design of gene vehicles with the improved efficiency of siRNA delivery.
Collapse
Affiliation(s)
- Qinping Yang
- Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Yanliang Dong
- Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Xuanyu Wang
- Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Zhihao Lin
- Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Mingyu Yan
- Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Anjie Dong
- Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Jianhua Zhang
- Department of Polymer Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Changrong Wang
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai, 264003, China
| |
Collapse
|
31
|
Valente J, Pereira P, Sousa A, Queiroz J, Sousa F. Effect of Plasmid DNA Size on Chitosan or Polyethyleneimine Polyplexes Formulation. Polymers (Basel) 2021; 13:793. [PMID: 33807586 PMCID: PMC7962013 DOI: 10.3390/polym13050793] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 01/22/2023] Open
Abstract
Gene therapy could be simply defined as a strategy for the introduction of a functional copy of desired genes in patients, to correct some specific mutation and potentially treat the respective disorder. However, this straightforward definition hides very complex processes related to the design and preparation of the therapeutic genes, as well as the development of suitable gene delivery systems. Within non-viral vectors, polymeric nanocarriers have offered an ideal platform to be applied as gene delivery systems. Concerning this, the main goal of the study was to do a systematic evaluation on the formulation of pDNA delivery systems based on the complexation of different sized plasmids with chitosan (CH) or polyethyleneimine (PEI) polymers to search for the best option regarding encapsulation efficiency, surface charge, size, and delivery ability. The cytotoxicity and the transfection efficiency of these systems were accessed and, for the best p53 encoding pDNA nanosystems, the ability to promote protein expression was also evaluated. Overall, it was showed that CH polyplexes are more efficient on transfection when compared with the PEI polyplexes, resulting in higher P53 protein expression. Cells transfected with CH/p53-pDNA polyplexes presented an increase of around 54.2% on P53 expression, while the transfection with the PEI/p53-pDNA polyplexes resulted in a 32% increase.
Collapse
Affiliation(s)
- J.F.A. Valente
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (P.P.); (A.S.); (J.A.Q.)
- CDRsp—Centre Rapid and Sustainable Product Development, Polytechnic Institute of Leiria, 2411-901 Leiria, Portugal
| | - P. Pereira
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (P.P.); (A.S.); (J.A.Q.)
- CEMMPRE, Department of Chemical Engineering, University of Coimbra, Rua Sílvio Lima-Pólo II, 3030-790 Coimbra, Portugal
| | - A. Sousa
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (P.P.); (A.S.); (J.A.Q.)
| | - J.A. Queiroz
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (P.P.); (A.S.); (J.A.Q.)
| | - F. Sousa
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (P.P.); (A.S.); (J.A.Q.)
| |
Collapse
|
32
|
Liu T, Lin M, Wu F, Lin A, Luo D, Zhang Z. Development of a nontoxic and efficient gene delivery vector based on histidine grafted chitosan. INT J POLYM MATER PO 2021. [DOI: 10.1080/00914037.2021.1885407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Tianhui Liu
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University, Fuzhou, China
- Pharmaceutical and Medical Technology College, Putian University, Putian, China
| | - Mei Lin
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fuzhou, China
| | - Fan Wu
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Aizhu Lin
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Daoshu Luo
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Ziyang Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
33
|
Kumar R, Le N, Tan Z, Brown ME, Jiang S, Reineke TM. Efficient Polymer-Mediated Delivery of Gene-Editing Ribonucleoprotein Payloads through Combinatorial Design, Parallelized Experimentation, and Machine Learning. ACS NANO 2020; 14:17626-17639. [PMID: 33225680 DOI: 10.1021/acsnano.0c08549] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Chemically defined vectors such as cationic polymers are versatile alternatives to engineered viruses for the delivery of genome-editing payloads. However, their clinical translation hinges on rapidly exploring vast chemical design spaces and deriving structure-function relationships governing delivery performance. Here, we discovered a polymer for efficient intracellular ribonucleoprotein (RNP) delivery through combinatorial polymer design and parallelized experimental workflows. A chemically diverse library of 43 statistical copolymers was synthesized via combinatorial RAFT polymerization, realizing systematic variations in physicochemical properties. We selected cationic monomers that varied in their pKa values (8.1-9.2), steric bulk, and lipophilicity of their alkyl substituents. Co-monomers of varying hydrophilicity were also incorporated, enabling elucidation of the roles of protonation equilibria and hydrophobic-hydrophilic balance in vehicular properties and performance. We screened our multiparametric vector library through image cytometry and rapidly uncovered a hit polymer (P38), which outperforms state-of-the-art commercial transfection reagents, achieving nearly 60% editing efficiency via nonhomologous end-joining. Structure-function correlations underlying editing efficiency, cellular toxicity, and RNP uptake were probed through machine learning approaches to uncover the physicochemical basis of P38's performance. Although cellular toxicity and RNP uptake were solely determined by polyplex size distribution and protonation degree, respectively, these two polyplex design parameters were found to be inconsequential for enhancing editing efficiency. Instead, polymer hydrophobicity and the Hill coefficient, a parameter describing cooperativity-enhanced polymer deprotonation, were identified as the critical determinants of editing efficiency. Combinatorial synthesis and high-throughput characterization methodologies coupled with data science approaches enabled the rapid discovery of a polymeric vehicle that would have otherwise remained inaccessible to chemical intuition. The statistically derived design rules elucidated herein will guide the synthesis and optimization of future polymer libraries tailored for therapeutic applications of RNP-based genome editing.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ngoc Le
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Zhe Tan
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mary E Brown
- University Imaging Centers, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Shan Jiang
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
34
|
Tan L, Deng X, Lai X, Zeng T, Li A, Hu J, Xiong Z. Mesoscale nanoparticles encapsulated with emodin for targeting antifibrosis in animal models. OPEN CHEM 2020. [DOI: 10.1515/chem-2020-0163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AbstractThe aim of this study is to explore the kidney-targeting capability of mesoscale nanoparticles (MNPs)-emodin (Em-MNPs) and its potential antifibrosis in the animal model. First, MNPs and Em-MNPs were synthesized via nanoprecipitation method, and their diameters were both ∼400 nm with the uniform size. The entrapment efficiency of MNPs was 45.1% when adding emodin at the concentration of 12 mg/mL. Moreover, cytotoxicity assay showed that Em-MNPs presented excellent biocompatibility in rat proximal tubular cells. Cellular uptake assay demonstrated that Em-MNPs had high-efficiency uptake, especially in the cytoplasm. Ex vivo organ fluorescence imaging revealed that Em-MNPs possessed specific kidney-targeting ability with relative long retention time in the kidney (∼24 h). In the renal unilateral ureteral obstruction model, Em-MNPs treatment could significantly alleviate kidney tubule injury and reduce extracellular matrix deposition compared with free MNPs. Herein, Em-MNPs with specific kidney-targeting and preferable antifibrosis effects in animal model may pave an avenue for treating renal diseases.
Collapse
Affiliation(s)
- Lishan Tan
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| | - Xiulong Deng
- Department of Chemical and Chemical Engineering, Key Laboratory of Organo-Pharmaceutical Chemistry, Gannan Normal University, Ganzhou, Jiangxi Province, 341000, China
| | - Xuandi Lai
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Tao Zeng
- Department of Nephrology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Aiqing Li
- Department of Nephrology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jianqiang Hu
- Department of Nephrology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zuying Xiong
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| |
Collapse
|
35
|
Qin B, Yuan X, Jiang M, Yin H, Luo Z, Zhang J, Zhu C, Li X, Shi Y, Luo L, Du Y, You J. Targeting DNA to the endoplasmic reticulum efficiently enhances gene delivery and therapy. NANOSCALE 2020; 12:18249-18262. [PMID: 32857088 DOI: 10.1039/d0nr03156a] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Gene therapy mediated by non-viral carriers is gaining an increasing popularity due to its high biosafety and the convenience of production on a large scale, yet inefficient gene delivery is a limiting obstacle. Few gene vectors can avoid the endosome-lysosome route, and as a result, their DNA payloads are easily decomposed during transfection. Herein, a peptide (pardaxin, PAR)-modified cationic liposome (PAR-Lipo) targeting the endoplasmic reticulum (ER) was developed for improving the gene delivery efficiency. Interestingly, compared to non-PAR-modified cationic liposomes (Non-Lipos) and Lipofectamine 2000 (Lipo 2000, a commercial genetic vector), PAR-Lipos showed remarkably higher gene delivery efficiency in vitro and better antitumor efficacy in vivo. It was demonstrated that PAR-Lipos could be accumulated into the ER via a non-lysosome intracellular route after cellular internalization, which induced the retention of the DNA payload in the ER close to the nucleus, while Non-Lipos, like most conventional cationic carriers, mainly presented lysosomal retention after their endocytosis. The unique intracellular transport behavior of PAR-Lipos can enhance the protection of the DNA payload, prolong their residence time in the cell, and promote their entry into the nucleus relying on the intimate relationship between the ER and nuclear membrane, which is the explanation for the enhanced gene-therapy effect mediated by PAR-Lipos. Our research may provide alternative means of efficiently delivering genes in cells.
Collapse
Affiliation(s)
- Bing Qin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Roy S, Zhu D, Parak WJ, Feliu N. Lysosomal Proton Buffering of Poly(ethylenimine) Measured In Situ by Fluorescent pH-Sensor Microcapsules. ACS NANO 2020; 14:8012-8023. [PMID: 32568521 DOI: 10.1021/acsnano.9b10219] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Poly(ethylenimine) (PEI) is frequently used as transfection agent for delivery of nucleic acids to the cytosol. After endocytosis of complexes of PEI and nucleic acids, a fraction of them can escape endosomes/lysosomes and reach the cytosol. One proposed mechanism is the so-called proton sponge effect, which involves buffering of the lysosomal pH by PEI. There are however also reports that report the absence of such buffering. In this work, the buffering capacity of PEI of the lysosomal pH was investigated in situ by combining PEI and pH-sensing ratiometric fluorophores in a single carrier particle. As carrier particles, hereby capsules were used, which were composed of polyelectrolyte walls based on layer-by-layer assembly, with the pH sensors located inside the capsule cavities. In this way, the local pH around individual particles could be monitored during the whole process of endocytosis. Results demonstrate the pH-buffering capability of PEI, which prevents the strong acidification of lysosomes containing PEI. This effect was related to the presence of PEI and was not related to the overall charge of the carrier particles. In case PEI was added in molecular form, no buffering of pH could be observed by endocytosed encapsulated pH-sensing ratiometric fluorophores. Co-localization experiments demonstrated that this was due to the fact that internalized free PEI and the encapsulated pH-sensing ratiometric fluorophores were not located in the same lysosomes. Missing co-localization might explain why also in other studies no pH buffering was found; in the case of co-delivery of PEI, the pH sensors could be clearly observed.
Collapse
Affiliation(s)
- Sathi Roy
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22607 Hamburg, Germany
| | - Dingcheng Zhu
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22607 Hamburg, Germany
| | - Wolfgang J Parak
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22607 Hamburg, Germany
- CIC Biomagune, Miramon Pasealekua 182, 20014 San Sebastian, Spain
| | - Neus Feliu
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22607 Hamburg, Germany
| |
Collapse
|
37
|
Liu N, Tang M, Ding J. The interaction between nanoparticles-protein corona complex and cells and its toxic effect on cells. CHEMOSPHERE 2020; 245:125624. [PMID: 31864050 DOI: 10.1016/j.chemosphere.2019.125624] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 05/23/2023]
Abstract
Once nanoparticles (NPs) contact with the biological fluids, the proteins immediately adsorb onto their surface, forming a layer called protein corona (PC), which bestows the biological identity on NPs. Importantly, the NPs-PC complex is the true identity of NPs in physiological environment. Based on the affinity and the binding and dissociation rate, PC is classified into soft protein corona, hard protein corona, and interfacial protein corona. Especially, the hard PC, a protein layer relatively stable and closer to their surface, plays particularly important role in the biological effects of the complex. However, the abundant corona proteins rarely correspond to the most abundant proteins found in biological fluids. The composition profile, formation and conformational change of PC can be affected by many factors. Here, the influence factors, not only the nature of NPs, but also surface chemistry and biological medium, are discussed. Likewise, the formed PC influences the interaction between NPs and cells, and the associated subsequent cellular uptake and cytotoxicity. The uncontrolled PC formation may induce undesirable and sometimes opposite results: increasing or inhibiting cellular uptake, hindering active targeting or contributing to passive targeting, mitigating or aggravating cytotoxicity, and stimulating or mitigating the immune response. In the present review, we discuss these aspects and hope to provide a valuable reference for controlling protein adsorption, predicting their behavior in vivo experiments and designing lower toxicity and enhanced targeting nanomedical materials for nanomedicine.
Collapse
Affiliation(s)
- Na Liu
- Key Laboratory of Environmental Medicine & Engineering, Ministry of Education, School of Public Health, Southeast University, 87 Ding Jia Qiao, Nanjing, 210009, PR China.
| | - Meng Tang
- Key Laboratory of Environmental Medicine & Engineering, Ministry of Education, School of Public Health, Southeast University, 87 Ding Jia Qiao, Nanjing, 210009, PR China.
| | - Jiandong Ding
- Department of Cardiology, Zhongda Hospital, Southeast University, 87 Ding Jia Qiao, Nanjing, 210009, PR China.
| |
Collapse
|
38
|
Laird NZ, Malkawi WI, Chakka JL, Acri TM, Elangovan S, Salem AK. A proof of concept gene-activated titanium surface for oral implantology applications. J Tissue Eng Regen Med 2020; 14:622-632. [PMID: 32078257 DOI: 10.1002/term.3026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 12/10/2019] [Accepted: 01/27/2020] [Indexed: 01/15/2023]
Abstract
Dental implants are very successful medical devices, yet implant failures do occur due to biological and mechanical complications. Peri-implantitis is one such biological complication that is primarily caused by bacteria and their products at the implant soft tissue interface. Bacterial infiltration can be prevented by the formation of a reliable soft tissue seal encircling dental implants. Platelet-derived growth factor-BB (PDGF-BB) has significant chemotactic and proliferative effects on various mesenchymal cell types, including fibroblasts, and therefore can be an effective molecule to enhance the peri-implant soft tissue seal. To overcome the limitations of the recombinant protein form of PDGF-BB, such as cost and the need for supraphysiological doses, we have developed and characterized a titanium surface that is rendered bioactive by coating it with polyethylenimine-plasmid DNA (pDNA) nanoplexes in the presence of sucrose. Human embryonic kidney 293T (HEK293T) cells and human primary gingival fibroblasts (GFs) were successfully transfected in culture with enhanced green fluorescent protein (EGFP)-encoding pDNA or platelet-derived growth factor subunit B (PDGFB)-encoding pDNA loaded into nanoplexes and coated onto titanium disks in a dose-dependent manner. GFs were shown to secrete PDGF-BB for at least 7 days after transfection and displayed both minimal viability loss and increased integrin-α2 expression 4 days posttransfection.
Collapse
Affiliation(s)
- Noah Z Laird
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA
| | - Walla I Malkawi
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA
| | - Jaidev L Chakka
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA
| | - Timothy M Acri
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA
| | - Satheesh Elangovan
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA.,Department of Periodontics, College of Dentistry and Dental Clinics, The University of Iowa, Iowa City, IA
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA.,Department of Chemical and Biochemical Engineering, College of Engineering, The University of Iowa, Iowa City, IA
| |
Collapse
|
39
|
Roy S, Elbaz NM, Parak WJ, Feliu N. Biodegradable Alginate Polyelectrolyte Capsules As Plausible Biocompatible Delivery Carriers. ACS APPLIED BIO MATERIALS 2019; 2:3245-3256. [DOI: 10.1021/acsabm.9b00203] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Sathi Roy
- Faculty of Physics, Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Hamburg, Germany
| | - Nancy M. Elbaz
- Department of Chemistry, University of Liverpool, Crown Street, Liverpool L69 7ZD, United Kingdom
| | - Wolfgang J. Parak
- Faculty of Physics, Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Hamburg, Germany
| | - Neus Feliu
- Faculty of Physics, Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Hamburg, Germany
| |
Collapse
|
40
|
Santo D, Mendonça PV, Lima MS, Cordeiro RA, Cabanas L, Serra A, Coelho JFJ, Faneca H. Poly(ethylene glycol)- block-poly(2-aminoethyl methacrylate hydrochloride)-Based Polyplexes as Serum-Tolerant Nanosystems for Enhanced Gene Delivery. Mol Pharm 2019; 16:2129-2141. [PMID: 30986077 DOI: 10.1021/acs.molpharmaceut.9b00101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Incorporation of poly(ethylene glycol) (PEG) into polyplexes has been used as a promising approach to enhance their stability and reduce unwanted interactions with biomolecules. However, this strategy generally has a negative influence on cellular uptake and, consequently, on transfection of target cells. In this work, we explore the effect of PEGylation on biological and physicochemical properties of poly(2-aminoethyl methacrylate) (PAMA)-based polyplexes. For this purpose, different tailor-made PEG- b-PAMA block copolymers, and the respective homopolymers, were synthesized using the controlled/"living" radical polymerization method based on activators regenerated by electron transfer atom transfer radical polymerization. The obtained data show that PEG- b-PAMA-based polyplexes exhibited a much better transfection activity/cytotoxicity relationship than the corresponding non-PEGylated nanocarriers. The best formulation, prepared with the largest block copolymer (PEG45- b-PAMA168) at a 25:1 N/P ratio, presented a 350-fold higher transfection activity in the presence of serum than that obtained with polyplexes generated with the gold standard bPEI. This higher transfection activity was associated to an improved capability to overcome the intracellular barriers, namely the release from the endolysosomal pathway and the vector unpacking and consequent DNA release from the nanosystem inside cells. Moreover, these nanocarriers exhibit suitable physicochemical properties for gene delivery, namely reduced sizes, high DNA protection, and colloidal stability. Overall, these findings demonstrate the high potential of the PEG45- b-PAMA168 block copolymer as a gene delivery system.
Collapse
Affiliation(s)
- Daniela Santo
- Center for Neuroscience and Cell Biology , University of Coimbra , 3004-504 Coimbra , Portugal
| | - Patrícia V Mendonça
- CEMMPRE, Department of Chemical Engineering , University of Coimbra , 3030-790 Coimbra , Portugal
| | - Mafalda S Lima
- CEMMPRE, Department of Chemical Engineering , University of Coimbra , 3030-790 Coimbra , Portugal
| | - Rosemeyre A Cordeiro
- Center for Neuroscience and Cell Biology , University of Coimbra , 3004-504 Coimbra , Portugal
| | - Luis Cabanas
- Center for Neuroscience and Cell Biology , University of Coimbra , 3004-504 Coimbra , Portugal
| | - Arménio Serra
- CEMMPRE, Department of Chemical Engineering , University of Coimbra , 3030-790 Coimbra , Portugal
| | - Jorge F J Coelho
- CEMMPRE, Department of Chemical Engineering , University of Coimbra , 3030-790 Coimbra , Portugal
| | - Henrique Faneca
- Center for Neuroscience and Cell Biology , University of Coimbra , 3004-504 Coimbra , Portugal
| |
Collapse
|
41
|
Jiang XC, Xiang JJ, Wu HH, Zhang TY, Zhang DP, Xu QH, Huang XL, Kong XL, Sun JH, Hu YL, Li K, Tabata Y, Shen YQ, Gao JQ. Neural Stem Cells Transfected with Reactive Oxygen Species-Responsive Polyplexes for Effective Treatment of Ischemic Stroke. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1807591. [PMID: 30633395 DOI: 10.1002/adma.201807591] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 12/19/2018] [Indexed: 06/09/2023]
Abstract
Neural stem cells (NSCs), capable of ischemia-homing, regeneration, and differentiation, exert strong therapeutic potentials in treating ischemic stroke, but the curative effect is limited in the harsh microenvironment of ischemic regions rich in reactive oxygen species (ROS). Gene transfection to make NSCs overexpress brain-derived neurotrophic factor (BDNF) can enhance their therapeutic efficacy; however, viral vectors must be used because current nonviral vectors are unable to efficiently transfect NSCs. The first polymeric vector, ROS-responsive charge-reversal poly[(2-acryloyl)ethyl(p-boronic acid benzyl)diethylammonium bromide] (B-PDEA), is shown here, that mediates efficient gene transfection of NSCs and greatly enhances their therapeutics in ischemic stroke treatment. The cationic B-PDEA/DNA polyplexes can effectively transfect NSCs; in the cytosol, the B-PDEA is oxidized by intracellular ROS into negatively charged polyacrylic acid, quickly releasing the BDNF plasmids for efficient transcription and secreting a high level of BDNF. After i.v. injection in ischemic stroke mice, the transfected NSCs (BDNF-NSCs) can home to ischemic regions as efficiently as the pristine NSCs but more efficiently produce BDNF, leading to significantly augmented BDNF levels, which in turn enhances the mouse survival rate to 60%, from 0% (nontreated mice) or ≈20% (NSC-treated mice), and enables more rapid and superior functional reconstruction.
Collapse
Affiliation(s)
- Xin-Chi Jiang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Jia-Jia Xiang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
| | - Hong-Hui Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Tian-Yuan Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Dan-Ping Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Qian-Hao Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Xiao-Li Huang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Xiang-Lei Kong
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, P. R. China
| | - Ji-Hong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, P. R. China
| | - Yu-Lan Hu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Kai Li
- Institute of Materials Science and Engineering, A*STAR, Singapore, 138634, Singapore
| | - Yasuhiko Tabata
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - You-Qing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| |
Collapse
|
42
|
Zhang W, Kang X, Yuan B, Wang H, Zhang T, Shi M, Zheng Z, Zhang Y, Peng C, Fan X, Yang H, Shen Y, Huang Y. Nano-Structural Effects on Gene Transfection: Large, Botryoid-Shaped Nanoparticles Enhance DNA Delivery via Macropinocytosis and Effective Dissociation. Theranostics 2019; 9:1580-1598. [PMID: 31037125 PMCID: PMC6485200 DOI: 10.7150/thno.30302] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/03/2019] [Indexed: 12/22/2022] Open
Abstract
Effective delivery is the primary barrier against the clinical translation of gene therapy. Yet there remains too much unknown in the gene delivery mechanisms, even for the most investigated polymeric carrier (i.e., PEI). As a consequence, the conflicting results have been often seen in the literature due to the large variability in the experimental conditions and operations. Therefore, some key parameters should be identified and thus strictly controlled in the formulation process. Methods: The effect of the formulation processing parameters (e.g., concentration or mixture volume) and the resulting nanostructure properties on gene transfection have been rarely investigated. Two types of the PEI/DNA nanoparticles (NPs) were prepared in the same manner with the same dose but at different concentrations. The microstructure of the NPs and the transfection mechanisms were investigated through various microscopic methods. The therapeutic efficacy of the NPs was demonstrated in the cervical subcutaneous xenograft and peritoneal metastasis mouse models. Results: The high-concentration process (i.e., small reaction-volume) for mixture resulted in the large-sized PEI/DNA NPs that had a higher efficiency of gene transfection, compared to the small counterpart that was prepared at a low concentration. The microstructural experiments showed that the prepared small NPs were firmly condensed, whereas the large NPs were bulky and botryoid-shaped. The large NPs entered the tumor cells via the macropinocytosis pathway, and then efficiently dissociated in the cytoplasm and released DNA, thus promoting the intranuclear delivery. The enhanced in vivo therapeutic efficacy of the large NPs was demonstrated, indicating the promise for local-regional administration. Conclusion: This work provides better understanding of the effect of formulation process on nano-structural properties and gene transfection, laying a theoretical basis for rational design of the experimental process.
Collapse
Affiliation(s)
- Wenyuan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuejia Kang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Bo Yuan
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Huiyuan Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Tao Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mingjie Shi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zening Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yuanheng Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Chengyuan Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaoming Fan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Huaiyu Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- East China Normal University School of Life Sciences, Shanghai 200241, China
| | - Youqing Shen
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
43
|
Sun K, Ding T, Xing Y, Mo D, Zhang J, Rosenholm JM. Hybrid mesoporous nanorods with deeply grooved lateral faces toward cytosolic drug delivery. Biomater Sci 2019; 7:5301-5311. [DOI: 10.1039/c9bm01251f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hybrid mesoporous nanorods with six twisted sharp edges can induce effective penetration of intracellular barriers and cytosolic delivery of membrane-impermeable drugs through curvature effects.
Collapse
Affiliation(s)
- Kaiyao Sun
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education
- College of Bioengineering
- Chongqing University
- Chongqing 400044
| | - Tao Ding
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education
- College of Bioengineering
- Chongqing University
- Chongqing 400044
| | - Yuxin Xing
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education
- College of Bioengineering
- Chongqing University
- Chongqing 400044
| | - Dong Mo
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education
- College of Bioengineering
- Chongqing University
- Chongqing 400044
| | - Jixi Zhang
- Key Laboratory of Biorheological Science and Technology
- Ministry of Education
- College of Bioengineering
- Chongqing University
- Chongqing 400044
| | - Jessica M. Rosenholm
- Pharmaceutical Sciences Laboratory
- Faculty of Science and Engineering
- Åbo Akademi University
- Turku 20520
- Finland
| |
Collapse
|
44
|
Huang W, Wang X, Wang C, Du L, Zhang J, Deng L, Cao H, Dong A. Structural exploration of hydrophobic core in polycationic micelles for improving siRNA delivery efficiency and cell viability. J Mater Chem B 2019; 7:965-973. [DOI: 10.1039/c8tb02706d] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Improving siRNA delivery efficiency often encounters a dilemma with poor or decreased biocompatibility for polycationic micelles.
Collapse
Affiliation(s)
- Wenjun Huang
- Department of Polymer Science and Technology
- School of Chemical Engineering and Technology
- Key Laboratory of Systems Bioengineering (Ministry of Education)
- Tianjin University
- Tianjin 300072
| | - Xiaoxia Wang
- Laboratory of Nucleic Acid Technology
- Institute of Molecular Medicine
- Peking University
- Beijing 100871
- China
| | - Changrong Wang
- Department of Polymer Science and Technology
- School of Chemical Engineering and Technology
- Key Laboratory of Systems Bioengineering (Ministry of Education)
- Tianjin University
- Tianjin 300072
| | - Lili Du
- Laboratory of Nucleic Acid Technology
- Institute of Molecular Medicine
- Peking University
- Beijing 100871
- China
| | - Jianhua Zhang
- Department of Polymer Science and Technology
- School of Chemical Engineering and Technology
- Key Laboratory of Systems Bioengineering (Ministry of Education)
- Tianjin University
- Tianjin 300072
| | - Liandong Deng
- Department of Polymer Science and Technology
- School of Chemical Engineering and Technology
- Key Laboratory of Systems Bioengineering (Ministry of Education)
- Tianjin University
- Tianjin 300072
| | - Huiqing Cao
- Laboratory of Nucleic Acid Technology
- Institute of Molecular Medicine
- Peking University
- Beijing 100871
- China
| | - Anjie Dong
- Department of Polymer Science and Technology
- School of Chemical Engineering and Technology
- Key Laboratory of Systems Bioengineering (Ministry of Education)
- Tianjin University
- Tianjin 300072
| |
Collapse
|
45
|
Deng X, Zeng T, Li J, Huang C, Yu M, Wang X, Tan L, Zhang M, Li A, Hu J. Kidney-targeted triptolide-encapsulated mesoscale nanoparticles for high-efficiency treatment of kidney injury. Biomater Sci 2019; 7:5312-5323. [PMID: 31617509 DOI: 10.1039/c9bm01290g] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Insolubility and toxicity of TP restrict clinical applications in renal diseases. Here, TP-encapsulated mesoscale nanoparticles offer a new therapeutic strategy for renal diseases due to good biocompability, kidney targeting and slow release.
Collapse
|
46
|
Fang H, Lin L, Chen J, Wu J, Tian H, Chen X. Zinc ion coordination significantly improved the transfection efficiency of low molecular weight polyethylenimine. Biomater Sci 2019; 7:1716-1728. [DOI: 10.1039/c9bm00039a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A zinc ion coordination-contained polycationic gene delivery system.
Collapse
Affiliation(s)
- Huapan Fang
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun
- China
| | - Lin Lin
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun
- China
| | - Jie Chen
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun
- China
| | - Jiayan Wu
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun
- China
| | - Huayu Tian
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun
- China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun
- China
| |
Collapse
|
47
|
Xiang J, Liu X, Zhou Z, Zhu D, Zhou Q, Piao Y, Jiang L, Tang J, Liu X, Shen Y. Reactive Oxygen Species (ROS)-Responsive Charge-Switchable Nanocarriers for Gene Therapy of Metastatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2018; 10:43352-43362. [PMID: 30465424 DOI: 10.1021/acsami.8b13291] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The application of nonviral gene vectors has been limited by their insufficient transfection efficiency because of poor serum stability, high endosomal entrapment, limited intracellular release, and low accumulation in the targeted organelle. It is still challenging to design gene carriers with properties that can overcome all of the barriers. We previously developed a reactive oxygen species (ROS)-responsive cationic polymer, poly[(2-acryloyl)ethyl( p-boronic acid benzyl) diethylammonium bromide] (B-PDEAEA), which switches the charge at high concentrations of intracellular ROS to promote intracellular DNA release. However, its gene-delivery efficiency has been limited by serum instability and lysosomal trapping, and coating with an anionic PEGylated lipid only showed mild enhancement. Herein, we coated the ROS-responsive B-PDEAEA polymer with two cationic lipids to form ROS-responsive lipopolyplexes with integrated properties to overcome multiple delivery barriers. The surface cationic lipids endowed the nanocarrier with improved serum stability, effective cellular uptake, and lysosomal evasion. The interior B-PDEAEA/DNA polyplexes, which were highly stable in the extracellular environment, but quickly dissociated, released DNA, promoted nuclei localization, and achieved efficient transcription. The mechanisms of the ROS-responsive and charge-switchable properties of B-PDEAEA were quantitatively studied. The transfection efficiency and antitumor activity of lipopolyplexes were studied in vitro and in vivo. We found that the ROS-responsive lipopolyplexes effectively delivered therapeutic genes into cell nuclei and caused high tumor inhibition in mice bearing peritoneal or lung metastases.
Collapse
Affiliation(s)
| | - Xin Liu
- Center for Stem Cell and Tissue Engineering, School of Medicine , Zhejiang University , Hangzhou 310058 , China
- Zhejiang Xinyue Biotechnology Co. Ltd. , Hangzhou 311121 , China
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ailincai D, Peptanariu D, Pinteala M, Marin L. Dynamic constitutional chemistry towards efficient nonviral vectors. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 94:635-646. [PMID: 30423749 DOI: 10.1016/j.msec.2018.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 09/11/2018] [Accepted: 10/01/2018] [Indexed: 10/28/2022]
Abstract
Dynamic constitutional chemistry has been used to design nonviral vectors for gene transfection. Their design has been thought in order to fulfill ab initio the main requirements for gene therapy. As building blocks were used hyperbranched PEI as hydrophilic part and benzentrialdehyde and a diamine linear siloxane as hydrophobic part, connected through reversible imine linkages. The obtaining of the envisaged structures has been confirmed by NMR and FTIR spectroscopy. The dynamic synthesized amphiphiles proved to be able to self-assemble in nano-sized spherical entities as was demonstrated by TEM and DLS, characterized by a narrow dimensional polydispersity. Agarose gel electrophoresis proved the ability of the synthesized compounds to bind DNA, while TEM revealed the spherical morphology of the formed polyplexes. As a proof of the concept, the nonviral vectors promoted an efficient transfection on HeLa cells, demonstrating that dynamic constitutional chemistry can be an important tool in the development of this domain.
Collapse
Affiliation(s)
- Daniela Ailincai
- Petru Poni Institute of Macromolecular Chemistry, Iasi, Romania.
| | | | | | - Luminita Marin
- Petru Poni Institute of Macromolecular Chemistry, Iasi, Romania
| |
Collapse
|
49
|
Xun MM, Huang Z, Xiao YP, Liu YH, Zhang J, Zhang JH, Yu XQ. Synthesis and Properties of Low-Molecular-Weight PEI-Based Lipopolymers for Delivery of DNA. Polymers (Basel) 2018; 10:E1060. [PMID: 30960985 PMCID: PMC6403936 DOI: 10.3390/polym10101060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/22/2018] [Accepted: 09/22/2018] [Indexed: 01/08/2023] Open
Abstract
Rapid enzymatic degradation and fragmentation during DNA administration can result in limited gene expression, and consequently, poor efficacy. It is necessary to use novel vectors for DNA delivery. Herein, we aimed to design useful carriers for enhancing transfection efficiency (TE). These lipopolymers were prepared through Michael addition reactions from low-molecular-weight (LMW) polyethyleneimine (PEI) and linkers with three kinds of steroids. Agarose gel electrophoresis assay results displayed that the three lipopolymers could condense plasmid DNA well, and the formed polyplexes had appropriate sizes around 200⁻300 nm, and zeta potentials of about +25⁻40 mV. The results of in vitro experiments using HeLa, HEK293, and MCF-7 cells showed that these lipopolymers present higher TE than 25-kDa PEI, both in the absence and presence of 10% serum. Flow cytometry and confocal microscopy studies also demonstrated that these lipopolymer/DNA complexes present higher cellular uptake and intracellular distribution. The measurement of critical micelle concentration (CMC) revealed that these lipopolymers could form micelles, which are suited for drug delivery. All results suggest that the three materials may serve as hopeful candidates for gene and drug delivery in future in vivo applications.
Collapse
Affiliation(s)
- Miao-Miao Xun
- National Demonstration Center for Experimental Chemical Engineering Comprehensive Education, School of Chemical Engineering and Technology, North University of China, Taiyuan 030000, China.
| | - Zheng Huang
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Ya-Ping Xiao
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Yan-Hong Liu
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Ji Zhang
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Ju-Hui Zhang
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Xiao-Qi Yu
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
50
|
Zhou Q, Wang Y, Xiang J, Piao Y, Zhou Z, Tang J, Liu X, Shen Y. Stabilized calcium phosphate hybrid nanocomposite using a benzoxaborole-containing polymer for pH-responsive siRNA delivery. Biomater Sci 2018; 6:3178-3188. [DOI: 10.1039/c8bm00575c] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Herein, we developed a PEG-PBO/siRNA/CaP hybrid nanocomposite with excellent stability and high siRNA loading content for effective pH-responsive siRNA delivery.
Collapse
Affiliation(s)
- Quan Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou 310027
- China
| | - Yue Wang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou 310027
- China
| | - Jiajia Xiang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou 310027
- China
| | - Ying Piao
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou 310027
- China
| | - Zhuxian Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou 310027
- China
| | - Jianbin Tang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou 310027
- China
| | - Xiangrui Liu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou 310027
- China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou 310027
- China
| |
Collapse
|