1
|
Hu X, Wang Y, Wang R, Pu Y, Jin R, Nie Y, Shuai X. The hybrid lipoplex induces cytoskeletal rearrangement via autophagy/RhoA signaling pathway for enhanced anticancer gene therapy. Nat Commun 2025; 16:339. [PMID: 39747218 PMCID: PMC11696071 DOI: 10.1038/s41467-024-55727-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 12/21/2024] [Indexed: 01/04/2025] Open
Abstract
Delivering plasmid DNA (pDNA) to solid tumors remains a significant challenge due to the requirement for multiple transport steps and the need to promote delivery efficiency. Herein, we present a virus-mimicking hybrid lipoplex, composed of an arginine-rich cationic lipid, hyaluronic acid derivatives coated gold nanoparticles, and pDNA. This system induces cytoskeletal rearrangements through "outside-in" mechanical and "inside-out" biochemical signaling, overcoming intra- and intercellular barriers to enhance pDNA delivery. By modulating autophagy, RhoA signaling, and cytoskeletal dynamics, we achieve a 20-fold increase in gene expression with high tissue specificity in solid tumors. Furthermore, the system is applied to co-deliver a p53 plasmid and an MDM2 inhibitor, demonstrating significant synergistic antitumor effects in hepatocellular and lung carcinomas.
Collapse
Affiliation(s)
- Xueyi Hu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China
| | - Yichun Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China
| | - Ruohan Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China
| | - Yiyao Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China.
| | - Yu Nie
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China.
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, P. R. China
| |
Collapse
|
2
|
Tasset A, Bellamkonda A, Wang W, Pyatnitskiy I, Ward D, Peppas N, Wang H. Overcoming barriers in non-viral gene delivery for neurological applications. NANOSCALE 2022; 14:3698-3719. [PMID: 35195645 PMCID: PMC9036591 DOI: 10.1039/d1nr06939j] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Gene therapy for neurological disorders has attracted significant interest as a way to reverse or stop various disease pathologies. Typical gene therapies involving the central and peripheral nervous system make use of adeno-associated viral vectors whose questionable safety and limitations in manufacturing has given rise to extensive research into non-viral vectors. While early research studies have demonstrated limited efficacy with these non-viral vectors, investigation into various vector materials and functionalization methods has provided insight into ways to optimize these non-viral vectors to improve desired characteristics such as improved blood-brain barrier transcytosis, improved perfusion in brain region, enhanced cellular uptake and endosomal escape in neural cells, and nuclear transport of genetic material post- intracellular delivery. Using a combination of various strategies to enhance non-viral vectors, research groups have designed multi-functional vectors that have been successfully used in a variety of pre-clinical applications for the treatment of Parkinson's disease, brain cancers, and cellular reprogramming for neuron replacement. While more work is needed in the design of these multi-functional non-viral vectors for neural applications, much of the groundwork has been done and is reviewed here.
Collapse
Affiliation(s)
- Aaron Tasset
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| | - Arjun Bellamkonda
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| | - Wenliang Wang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| | - Ilya Pyatnitskiy
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| | - Deidra Ward
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| | - Nicholas Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Huiliang Wang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
3
|
Monnery BD. Polycation-Mediated Transfection: Mechanisms of Internalization and Intracellular Trafficking. Biomacromolecules 2021; 22:4060-4083. [PMID: 34498457 DOI: 10.1021/acs.biomac.1c00697] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Polyplex-mediated gene transfection is now in its' fourth decade of serious research, but the promise of polyplex-mediated gene therapy has yet to fully materialize. Only approximately one in a million applied plasmids actually expresses. A large part of this is due to an incomplete understanding of the mechanism of polyplex transfection. There is an assumption that internalization must follow a canonical mechanism of receptor mediated endocytosis. Herein, we present arguments that untargeted (and most targeted) polyplexes do not utilize these routes. By incorporating knowledge of syndecan-polyplex interactions, we can show that syndecans are the "target" for polyplexes. Further, it is known that free polycations (which disrupt cell-membranes by acid-catalyzed hydrolysis of phospholipid esters) are necessary for (untargeted) endocytosis. This can be incorporated into the model to produce a novel mechanism of endocytosis, which fits the observed phenomenology. After membrane translocation, polyplex containing vesicles reach the endosome after diffusing through the actin mesh below the cell membrane. From there, they are acidified and trafficked toward the lysosome. Some polyplexes are capable of escaping the endosome and unpacking, while others are not. Herein, it is argued that for some polycations, as acidification proceeds the polyplexes excluding free polycations, which disrupt the endosomal membrane by acid-catalyzed hydrolysis, allowing the polyplex to escape. The polyplex's internal charge ratio is now insufficient for stability and it releases plasmids which diffuse to the nucleus. A small proportion of these plasmids diffuse through the nuclear pore complex (NPC), with aggregation being the major cause of loss. Those plasmids that have diffused through the NPC will also aggregate, and this appears to be the reason such a small proportion of nuclear plasmids express mRNA. Thus, the structural features which promote unpacking in the endosome and allow for endosomal escape can be determined, and better polycations can be designed.
Collapse
Affiliation(s)
- Bryn D Monnery
- Department of Organic and (Bio)Polymer Chemistry, Hasselt University, Building F, Agoralaan 1, B-3590 Diepenbeek, Belgium
| |
Collapse
|
4
|
Graceffa V. Physical and mechanical cues affecting biomaterial-mediated plasmid DNA delivery: insights into non-viral delivery systems. J Genet Eng Biotechnol 2021; 19:90. [PMID: 34142237 PMCID: PMC8211807 DOI: 10.1186/s43141-021-00194-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Whilst traditional strategies to increase transfection efficiency of non-viral systems aimed at modifying the vector or the polyplexes/lipoplexes, biomaterial-mediated gene delivery has recently sparked increased interest. This review aims at discussing biomaterial properties and unravelling underlying mechanisms of action, for biomaterial-mediated gene delivery. DNA internalisation and cytoplasmic transport are initially discussed. DNA immobilisation, encapsulation and surface-mediated gene delivery (SMD), the role of extracellular matrix (ECM) and topographical cues, biomaterial stiffness and mechanical stimulation are finally outlined. MAIN TEXT Endocytic pathways and mechanisms to escape the lysosomal network are highly variable. They depend on cell and DNA complex types but can be diverted using appropriate biomaterials. 3D scaffolds are generally fabricated via DNA immobilisation or encapsulation. Degradation rate and interaction with the vector affect temporal patterns of DNA release and transgene expression. In SMD, DNA is instead coated on 2D surfaces. SMD allows the incorporation of topographical cues, which, by inducing cytoskeletal re-arrangements, modulate DNA endocytosis. Incorporation of ECM mimetics allows cell type-specific transfection, whereas in spite of discordances in terms of optimal loading regimens, it is recognised that mechanical loading facilitates gene transfection. Finally, stiffer 2D substrates enhance DNA internalisation, whereas in 3D scaffolds, the role of stiffness is still dubious. CONCLUSION Although it is recognised that biomaterials allow the creation of tailored non-viral gene delivery systems, there still are many outstanding questions. A better characterisation of endocytic pathways would allow the diversion of cell adhesion processes and cytoskeletal dynamics, in order to increase cellular transfection. Further research on optimal biomaterial mechanical properties, cell ligand density and loading regimens is limited by the fact that such parameters influence a plethora of other different processes (e.g. cellular adhesion, spreading, migration, infiltration, and proliferation, DNA diffusion and release) which may in turn modulate gene delivery. Only a better understanding of these processes may allow the creation of novel robust engineered systems, potentially opening up a whole new area of biomaterial-guided gene delivery for non-viral systems.
Collapse
Affiliation(s)
- Valeria Graceffa
- Cellular Health and Toxicology Research Group (CHAT), Institute of Technology Sligo, Ash Ln, Bellanode, Sligo, Ireland.
- Department of Life Sciences, Institute of Technology Sligo, Ash Ln, Bellanode, Sligo, Ireland.
| |
Collapse
|
5
|
Abeyratne-Perera HK, Basu S, Chandran PL. Shells of compacted DNA as nanocontainers transporting proteins in multiplexed delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 127:112184. [PMID: 34225845 DOI: 10.1016/j.msec.2021.112184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 11/24/2022]
Abstract
Polyethyleneimine (PEI) polymers are known to compact DNA strands into spheroid, toroid, or rod structures. A formulation with mannose-grafted PEI (PEIm), however, was reported to compact DNA into ~100 nm spheroids that indented like thin-walled pressurized shells. The goal of the study is to understand why mannose bristles divert the traditional pathway of PEI-DNA compaction to produce shell-like structures, and to manipulate the process so that proteins can be packed into the core of the assembling shells for co-delivering DNA and proteins into cells. DLS, AFM, and TEM imaging provide a consistent picture that BSA proteins can be packed into the shells without altering the shell architecture, as long as the proteins were added during the time course of shell assembly. Force spectroscopy studies reveal that DNA shells that buckle also have a rich surface-coating of mannose, indicating that a micelle-like partitioning of hydrophobic and hydrophilic layers governs shell assembly. When HEK293T cells are spiked with BSA-laden DNA shells, co-transfection of DNA and BSA is observed at higher levels than control formulations. Distinct micron-sized features appear having both green fluorescence from BSA-FITC and blue fluorescence from NucBlue DNA stain, suggesting BSA release in nucleus and secretory granules. With DNA nanocontainers, proteins can take advantage of the efficiency of PEI-based DNA transfection for hitchhiking into cells while being shielded from the challenges of the intracellular route. DNA nanocontainers are rapid to assemble, not dependent on the DNA sequence, and can be adapted for different protein types; thereby having potential to serve as a high-throughput platform in scenarios where DNA and protein have to be released at the same site and time within cells (e.g., theranostics, multiplexed co-delivery, gene editing).
Collapse
Affiliation(s)
- Hashanthi K Abeyratne-Perera
- Biochemistry and Molecular Biology Department, College of Medicine, Howard University, Washington, DC, United States of America
| | - Saswati Basu
- Chemical Engineering Department, College of Engineering and Architecture, Howard University, Washington, DC, United States of America
| | - Preethi L Chandran
- Biochemistry and Molecular Biology Department, College of Medicine, Howard University, Washington, DC, United States of America; Chemical Engineering Department, College of Engineering and Architecture, Howard University, Washington, DC, United States of America.
| |
Collapse
|
6
|
Lai WF, Wong WT. Roles of the actin cytoskeleton in aging and age-associated diseases. Ageing Res Rev 2020; 58:101021. [PMID: 31968269 DOI: 10.1016/j.arr.2020.101021] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/06/2020] [Accepted: 01/17/2020] [Indexed: 12/12/2022]
Abstract
The integrity of the cytoskeleton is essential to diverse cellular processes such as phagocytosis and intracellular trafficking. Disruption of the organization and dynamics of the actin cytoskeleton leads to age-associated symptoms and diseases, ranging from cancer to neurodegeneration. In addition, changes in the integrity of the actin cytoskeleton disrupt the functioning of not only somatic and stem cells but also gametes, resulting in aberrant embryonic development. Strategies to preserve the integrity and dynamics of the cytoskeleton are, therefore, potentially therapeutic to age-related disorders. The objective of this article is to revisit the current understanding of the roles played by the actin cytoskeleton in aging, and to review the opportunities and challenges for the transition of basic research into intervention development. It is hoped that, with the snapshot of evidence regarding changes in actin dynamics with advanced age, insights into future research directions can be attained.
Collapse
Affiliation(s)
- Wing-Fu Lai
- School of Pharmaceutical Sciences, Shenzhen University, PR China; School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, PR China; Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, PR China.
| | - Wing-Tak Wong
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, PR China
| |
Collapse
|
7
|
Truong NF, Lesher-Pérez SC, Kurt E, Segura T. Pathways Governing Polyethylenimine Polyplex Transfection in Microporous Annealed Particle Scaffolds. Bioconjug Chem 2019; 30:476-486. [PMID: 30513197 PMCID: PMC7290906 DOI: 10.1021/acs.bioconjchem.8b00696] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Gene delivery using injectable hydrogels can serve as a potential method for regulated tissue regeneration in wound healing. Our microporous annealed particle (MAP) hydrogel has been shown to promote cellular infiltration in both skin and brain wounds, while reducing inflammation. Although the scaffold itself can promote healing, it is likely that other signals will be required to promote healing of hard-to-treat wounds. Gene delivery is one approach to introduce desired bioactive signals. In this study, we investigated how the properties of MAP hydrogels influence non-viral gene delivery of polyethylenimine-condensed plasmid to cells seeded within the MAP gel. From past studies, we found that gene transfer to cells seeded in tissue culture plastic differed from gene transfer to cells seeded inside hydrogel scaffolds. Since MAP scaffolds are generated from hydrogel microparticles that are approximately 100 μm in diameter, they display local characteristics that can be viewed as two-dimensional or three-dimensional to cells. Thus, we sought to study if gene transfer inside MAP scaffolds differed from gene transfer to cells seeded in tissue culture plastic. We sought to understand the roles of the endocytosis pathway, actin and microtubule dynamics, RhoGTPases, and YAP/TAZ on transfection of human fibroblasts.
Collapse
Affiliation(s)
- Norman F Truong
- Department of Chemical and Biomolecular Engineering , University of California , Los Angeles , California 90095 , United States
| | - Sasha Cai Lesher-Pérez
- Department of Chemical and Biomolecular Engineering , University of California , Los Angeles , California 90095 , United States
| | - Evan Kurt
- Department of Biomedical Engineering , Duke University , Durham , North Carolina 27708 , United States
| | - Tatiana Segura
- Department of Chemical and Biomolecular Engineering , University of California , Los Angeles , California 90095 , United States
| |
Collapse
|
8
|
|
9
|
Bus T, Traeger A, Schubert US. The great escape: how cationic polyplexes overcome the endosomal barrier. J Mater Chem B 2018; 6:6904-6918. [DOI: 10.1039/c8tb00967h] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Endo-lysosomal escape strategies of cationic polymer-mediated gene delivery at a glance.
Collapse
Affiliation(s)
- Tanja Bus
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC)
- Friedrich Schiller University Jena
- 07743 Jena
- Germany
- Jena Center for Soft Matter (JCSM)
| | - Anja Traeger
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC)
- Friedrich Schiller University Jena
- 07743 Jena
- Germany
- Jena Center for Soft Matter (JCSM)
| | - Ulrich S. Schubert
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC)
- Friedrich Schiller University Jena
- 07743 Jena
- Germany
- Jena Center for Soft Matter (JCSM)
| |
Collapse
|
10
|
Gonzalez-Fernandez T, Sathy B, Hobbs C, Cunniffe G, McCarthy H, Dunne N, Nicolosi V, O'Brien F, Kelly D. Mesenchymal stem cell fate following non-viral gene transfection strongly depends on the choice of delivery vector. Acta Biomater 2017; 55:226-238. [PMID: 28363788 DOI: 10.1016/j.actbio.2017.03.044] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/06/2017] [Accepted: 03/27/2017] [Indexed: 12/22/2022]
Abstract
Controlling the phenotype of mesenchymal stem cells (MSCs) through the delivery of regulatory genes is a promising strategy in tissue engineering (TE). Essential to effective gene delivery is the choice of gene carrier. Non-viral delivery vectors have been extensively used in TE, however their intrinsic effects on MSC differentiation remain poorly understood. The objective of this study was to investigate the influence of three different classes of non-viral gene delivery vectors: (1) cationic polymers (polyethylenimine, PEI), (2) inorganic nanoparticles (nanohydroxyapatite, nHA) and (3) amphipathic peptides (RALA peptide) on modulating stem cell fate after reporter and therapeutic gene delivery. Despite facilitating similar reporter gene transfection efficiencies, these nanoparticle-based vectors had dramatically different effects on MSC viability, cytoskeletal morphology and differentiation. After reporter gene delivery (pGFP or pLUC), the nHA and RALA vectors supported an elongated MSC morphology, actin stress fibre formation and the development of mature focal adhesions, while cells appeared rounded and less tense following PEI transfection. These changes in MSC morphology correlated with enhanced osteogenesis following nHA and RALA transfection and adipogenesis following PEI transfection. When therapeutic genes encoding for transforming growth factor beta 3 (TGF-β3) and/or bone morphogenic protein 2 (BMP2) were delivered to MSCs, nHA promoted osteogenesis in 2D culture and the development of an endochondral phenotype in 3D culture, while RALA was less osteogenic and appeared to promote a more stable hyaline cartilage-like phenotype. In contrast, PEI failed to induce robust osteogenesis or chondrogenesis of MSCs, despite effective therapeutic protein production. Taken together, these results demonstrate that the differentiation of MSCs through the application of non-viral gene delivery strategies depends not only on the gene delivered, but also on the gene carrier itself. STATEMENT OF SIGNIFICANCE Nanoparticle-based non-viral gene delivery vectors have been extensively used in regenerative medicine, however their intrinsic effects on mesenchymal stem cell (MSC) differentiation remain poorly understood. This paper demonstrates that different classes of commonly used non-viral vectors are not inert and they have a strong effect on cell morphology, stress fiber formation and gene transcription in MSCs, which in turn modulates their capacity to differentiate towards osteogenic, adipogenic and chondrogenic lineages. These results also point to the need for careful and tissue-specific selection of nanoparticle-based delivery vectors to prevent undesired phenotypic changes and off-target effects when delivering therapeutic genes to damaged or diseased tissues.
Collapse
|
11
|
Alves RF, Favaro MT, Balbino TA, de Toledo MA, de la Torre LG, Azzoni AR. Recombinant protein-based nanocarriers and their association with cationic liposomes: Characterization and in vitro evaluation. Colloids Surf A Physicochem Eng Asp 2017. [DOI: 10.1016/j.colsurfa.2016.11.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
12
|
Jumping the nuclear envelope barrier: Improving polyplex-mediated gene transfection efficiency by a selective CDK1 inhibitor RO-3306. J Control Release 2016; 234:90-7. [DOI: 10.1016/j.jconrel.2016.05.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/14/2016] [Accepted: 05/16/2016] [Indexed: 01/02/2023]
|
13
|
Doxorubicin-loaded biodegradable self-assembly zein nanoparticle and its anti-cancer effect: Preparation, in vitro evaluation, and cellular uptake. Colloids Surf B Biointerfaces 2015; 140:324-331. [PMID: 26764113 DOI: 10.1016/j.colsurfb.2015.12.048] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 11/24/2015] [Accepted: 12/24/2015] [Indexed: 01/11/2023]
Abstract
Cancer is one top leading cause of the deaths worldwide. Various anticancer drugs, which can effectively kill cancer cells, have been developed in the last decade. However, the problem is still about the low therapeutic index of the drugs, which means that the effective dose of drugs will cause cytotoxicity to normal cells. A strategy based on drug nano-encapsulation is applied to achieve an effective anti-cancer therapy. In this study, we use zein, which is an amphiphilic protein, to make the anti-cancer drug nano-encapsulation. Doxorubicin (DOX), a popular anti-cancer drug, is selected as the core drug. The results show that DOX could be successfully encapsulated into zein to form spherical nanoparticles. The encapsulation efficiency and loading efficiency could reach as high as 90.06% and 15.01 mg/g, respectively. The cumulative release result showed a desired pH-responsible release behavior: DOX could be released faster in acidic buffer solutions (pH 5.0 and 6.5) than neutral one (pH 7.4). The effects of the nano-encapsulation on the anti-proliferation of HeLa cells were also examined. It indicated that, compared with free DOX, the DOX-loaded zein nanoparticles (DOX-zein-NPs) had a better effect on cancer cell killing at low DOX concentrations. We also investigated the cellular uptake of DOX-zein-NPs using confocal laser scanning microscopy (CLSM), flow cytometry, and transmission electron microscopy (TEM). And the endocytosis mechanism of DOX-zein-NPs entering into HeLa cells was studied using various endocytosis pathway inhibitors.
Collapse
|
14
|
Cervera L, Fuenmayor J, González-Domínguez I, Gutiérrez-Granados S, Segura MM, Gòdia F. Selection and optimization of transfection enhancer additives for increased virus-like particle production in HEK293 suspension cell cultures. Appl Microbiol Biotechnol 2015; 99:9935-49. [PMID: 26278533 DOI: 10.1007/s00253-015-6842-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 07/08/2015] [Accepted: 07/11/2015] [Indexed: 01/09/2023]
Abstract
The manufacturing of biopharmaceuticals in mammalian cells typically relies on the use of stable producer cell lines. However, in recent years, transient gene expression has emerged as a suitable technology for rapid production of biopharmaceuticals. Transient gene expression is particularly well suited for early developmental phases, where several potential therapeutic targets need to be produced and tested in vivo. As a relatively new bioprocessing modality, a number of opportunities exist for improving cell culture productivity upon transient transfection. For instance, several compounds have shown positive effects on transient gene expression. These transfection enhancers either facilitate entry of PEI/DNA transfection complexes into the cell or nucleus or increase levels of gene expression. In this work, the potential of combining transfection enhancers to increase Gag-based virus-like particle production levels upon transfection of suspension-growing HEK 293 cells is evaluated. Using Plackett-Burman design of experiments, it is first tested the effect of eight transfection enhancers: trichostatin A, valproic acid, sodium butyrate, dimethyl sulfoxide (DMSO), lithium acetate, caffeine, hydroxyurea, and nocodazole. An optimal combination of compounds exhibiting the highest effect on gene expression levels was subsequently identified using a surface response experimental design. The optimal consisted on the addition of 20 mM lithium acetate, 3.36 mM valproic acid, and 5.04 mM caffeine which increased VLP production levels 3.8-fold, while maintaining cell culture viability at 94%.
Collapse
Affiliation(s)
- Laura Cervera
- Grup d'Enginyeria Cellular i Bioprocés, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.
| | - Javier Fuenmayor
- Grup d'Enginyeria Cellular i Bioprocés, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Irene González-Domínguez
- Grup d'Enginyeria Cellular i Bioprocés, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Sonia Gutiérrez-Granados
- Grup d'Enginyeria Cellular i Bioprocés, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Maria Mercedes Segura
- Bluebird Bio Pharmaceutical Sciences, Process Development Group, 150 2nd Street, Cambridge, MA, 02141, USA
| | - Francesc Gòdia
- Grup d'Enginyeria Cellular i Bioprocés, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| |
Collapse
|
15
|
ZHOU HAN, MA XIAOFENG, LIU YONGZE, DONG LEI, LUO YI, ZHU GUANGJIE, QIAN XIAOYUN, CHEN JIE, LU LIN, WANG JUNGUO, GAO XIA. Linear polyethylenimine-plasmid DNA nanoparticles are ototoxic to the cultured sensory epithelium of neonatal mice. Mol Med Rep 2015; 11:4381-8. [DOI: 10.3892/mmr.2015.3306] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 01/15/2015] [Indexed: 11/06/2022] Open
|
16
|
New views and insights into intracellular trafficking of drug-delivery systems by fluorescence fluctuation spectroscopy. Ther Deliv 2014; 5:173-88. [DOI: 10.4155/tde.13.148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Biomaterials in the nanometer size range can be engineered for site-specific delivery of drugs after injection into the blood circulation. However, translation of such nanomedicines from the bench to the bedside is still hindered by many extracellular and intracellular barriers. To realize the concept of targeted drug delivery with nanomedicines, research groups are studying intensively the extra- and intra-cellular mechanisms involved as a response to the physicochemical properties of the nanomedicines. In this review, we highlight the contributions of fluorescence fluctuations spectroscopy techniques to better understand, and in turn to bypass, the major hurdles to therapeutic delivery, focusing mostly on the intracellular dynamics of drug-delivery systems.
Collapse
|
17
|
Andersen H, Parhamifar L, Moein Moghimi S. Uptake and Intracellular Trafficking of Nanocarriers. INTRACELLULAR DELIVERY II 2014. [DOI: 10.1007/978-94-017-8896-0_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
18
|
Martin TM, Plautz SA, Pannier AK. Network analysis of endogenous gene expression profiles after polyethyleneimine-mediated DNA delivery. J Gene Med 2013; 15:142-54. [PMID: 23526566 DOI: 10.1002/jgm.2704] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 03/08/2013] [Accepted: 03/15/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND DNA delivery systems, which transport exogenous DNA to cells, have applications that include gene therapy, tissue engineering and medical devices. Although the cationic nonviral DNA carrier polyethyleneimine (PEI) has been widely studied, the molecular factors and pathways underlying PEI-mediated DNA transfer remain largely unknown, preventing the design of more efficient delivery systems. METHODS HEK 293 T cells were treated with polyplexes formed with PEI and pEGFPLuc encoding for green fluorescent protein (GFP). Transfected cells expressing GFP were flow-separated from treated, untransfected cells. Gene expression profiles were obtained using Affymetrix HG-U133 2.0 microarrays and differentially expressed genes were identified using R/Bioconductor. Gene network analysis using EGAN (exploratory gene association network) bioinformatics tools was then used to find interaction among genes and enriched gene ontology (GO) terms related to transfection. Genes identified by this method were perturbed using pharmacologic activators or inhibitors to assess their effect on DNA transfer. RESULTS Microarray analysis comparing transfected cells to untransfected cells revealed 215 genes to be differentially expressed, with the majority enriched to GO processes including metabolism, response to stimulus, cell cycle, biological regulation and cellular component organization or biogenesis pathways. Gene network analysis revealed a coordinated induction of RAP1A, SCG5, PGAP1, ATF3 and NEB genes implicated in cell stress, cell cycle and cytoskeletal processes. Altering pathways with pharmacologic agents confirmed the potential role of RAP1A, SCG5 and ATF3 in transfection. CONCLUSIONS Microarray and gene network analyses of the sorted, transfected cell population can identify potential mediators of transfection, providing a basis for the design of improved delivery systems.
Collapse
Affiliation(s)
- Timothy M Martin
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | | | |
Collapse
|
19
|
Shi J, Chou B, Choi JL, Ta AL, Pun SH. Investigation of Polyethylenimine/DNA Polyplex Transfection to Cultured Cells Using Radiolabeling and Subcellular Fractionation Methods. Mol Pharm 2013; 10:2145-56. [PMID: 23406286 DOI: 10.1021/mp300651q] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Quantitative analysis of the intracellular trafficking of nonviral vectors provides critical information that can guide the rational design of improved cationic systems for gene delivery. Subcellular fractionation methods, combined with radiolabeling, produce quantitative measurements of the intracellular trafficking of nonviral vectors and the therapeutic payload. In this work, differential and density-gradient centrifugation techniques were used to determine the intracellular distribution of radiolabeled 25 kDa branched polyethylenimine (bPEI)/plasmid DNA complexes ("polyplexes") in HeLa cells over time. By differential centrifugation, [(14)C]bPEI was found mostly in the lighter fractions whereas [(3)H]DNA was found mostly in the heavier fractions. A majority of the intracellular polymer (∼60%) and DNA (∼90%) were found in the nuclear fraction. Polymer and DNA also differed in their distribution to heavier and denser organelles (lysosomes, mitochondria) in density-gradient centrifugation studies. An unexpected finding from this study was that between 18 and 50% of the DNA applied to the cells became cell-associated (either with the cell membrane and/or internalized), while only 1-6% of the polymer did so, resulting in an effective N/P ratio of less than 1. These results suggest that a significant amount of cationic polymer is dissociated from the DNA cargo early on in the transfection process.
Collapse
Affiliation(s)
- Julie Shi
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
20
|
DNA delivery with hyperbranched polylysine: a comparative study with linear and dendritic polylysine. J Control Release 2013; 169:276-88. [PMID: 23379996 DOI: 10.1016/j.jconrel.2013.01.019] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 01/13/2013] [Accepted: 01/23/2013] [Indexed: 02/08/2023]
Abstract
PEI and polylysine are among the most investigated synthetic polymeric carriers for DNA delivery. Apart from their practical use, these 2 classes of polymers are also of interest from a fundamental point of view as they both can be prepared in different architectures (linear and branched/dendritic) and in a wide range of molecular weights, which is attractive to establish basic structure-activity relationships. This manuscript reports the results of an extensive study on the influence of molecular weight and architecture of a library of polylysine variants that includes linear, dendritic and hyperbranched polylysine. Hyperbranched polylysine is a new polylysine-based carrier that is structurally related to dendritic polylysine but possesses a randomly branched structure. Hyperbranched polylysine is attractive as it can be prepared in a one-step process on a large scale. The performance of these 3 classes of polylysine analogs was evaluated by assessing eGFP and IgG production in transient gene expression experiments with CHO DG44 cells, which revealed that protein production generally increased with increasing molecular weight and that at comparable molecular weight, the hyperbranched analogs were superior as compared to the dendritic and linear polylysines. To understand the differences between the gene delivery properties of the hyperbranched polylysine analogs on the one hand and the dendritic and linear polylysines on the other hand, the uptake and trafficking of the corresponding polyplexes were investigated. These experiments allowed us to identify (i) polyplex-external cell membrane binding, (ii) free, unbound polylysine coexisting with polyplexes as well as (iii) polymer buffer capacity as three possible factors that may contribute to the superior transfection properties of the hyperbranched polylysines as compared to their linear and dendritic analogs. Altogether, the results of this study indicate that hyperbranched polylysine is an interesting, alternative synthetic gene carrier. Hyperbranched polylysine can be produced at low costs and in large quantities, is partially biodegradable, which may help to prevent cumulative cytotoxicity, and possesses transfection properties that can approach those of PEI.
Collapse
|
21
|
Choi AWT, Louie MW, Li SPY, Liu HW, Chan BTN, Lam TCY, Lin ACC, Cheng SH, Lo KKW. Emissive Behavior, Cytotoxic Activity, Cellular Uptake, and PEGylation Properties of New Luminescent Rhenium(I) Polypyridine Poly(ethylene glycol) Complexes. Inorg Chem 2012. [DOI: 10.1021/ic301948d] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Alex Wing-Tat Choi
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P.R.
China
| | - Man-Wai Louie
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P.R.
China
| | - Steve Po-Yam Li
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P.R.
China
| | - Hua-Wei Liu
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P.R.
China
| | - Bruce Ting-Ngok Chan
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P.R.
China
| | - Tonlex Chun-Ying Lam
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P.R.
China
| | - Alex Chun-Chi Lin
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P.R.
China
| | - Shuk-Han Cheng
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P.R.
China
| | - Kenneth Kam-Wing Lo
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P.R.
China
| |
Collapse
|
22
|
Li SPY, Tang TSM, Yiu KSM, Lo KKW. Cyclometalated Iridium(III)-Polyamine Complexes with Intense and Long-Lived Multicolor Phosphorescence: Synthesis, Crystal Structure, Photophysical Behavior, Cellular Uptake, and Transfection Properties. Chemistry 2012; 18:13342-54. [DOI: 10.1002/chem.201200979] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Indexed: 12/17/2022]
|
23
|
Toledo MA, Janissen R, Favaro MT, Cotta MA, Monteiro GA, Prazeres DMF, Souza AP, Azzoni AR. Development of a recombinant fusion protein based on the dynein light chain LC8 for non-viral gene delivery. J Control Release 2012; 159:222-31. [DOI: 10.1016/j.jconrel.2012.01.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 01/10/2012] [Accepted: 01/12/2012] [Indexed: 01/08/2023]
|
24
|
Multifunctional silica nanotubes for dual-modality gene delivery and MR imaging. Biomaterials 2011; 32:3042-52. [DOI: 10.1016/j.biomaterials.2010.12.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Accepted: 12/13/2010] [Indexed: 11/23/2022]
|
25
|
van Gaal EVB, Oosting RS, van Eijk R, Bakowska M, Feyen D, Kok RJ, Hennink WE, Crommelin DJA, Mastrobattista E. DNA nuclear targeting sequences for non-viral gene delivery. Pharm Res 2011; 28:1707-22. [PMID: 21424159 PMCID: PMC3109246 DOI: 10.1007/s11095-011-0407-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 02/21/2011] [Indexed: 12/02/2022]
Abstract
Purpose To evaluate if introduction of DNA nuclear Targeting Sequences (DTS; i.e. recognition sequences for endogenous DNA-binding proteins) in plasmid DNA (pDNA) leads to increased transfection efficiency of non-viral gene delivery by virtue of enhanced nuclear import of the pDNA. Methods A set of DTS was identified and cloned into EGFP-reporter plasmids controlled by the CMV-promoter. These pDNA constructs were delivered into A431 and HeLa cells using standard electroporation, pEI-based polyfection or lipofection methods. The amount of pDNA delivered into the nucleus was determined by qPCR; transfection efficiency was determined by flow cytometry. Results Neither of these DTS increased transgene expression. We varied several parameters (mitotic activity, applied dose and delivery strategy), but without effect. Although upregulated transgene expression was observed after stimulation with TNF-α, this effect could be ascribed to non-specific upregulation of transcription rather than enhanced nuclear import. Nuclear copy numbers of plasmids containing or lacking a DTS did not differ significantly after lipofectamine-based transfection in dividing and non-dividing cells. Conclusion No beneficial effects of DTS on gene expression or nuclear uptake were observed in this study.
Collapse
Affiliation(s)
- Ethlinn V B van Gaal
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, 3584 CA, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Rao RR, He J, Leach JK. Biomineralized composite substrates increase gene expression with nonviral delivery. J Biomed Mater Res A 2010; 94:344-54. [PMID: 20186740 DOI: 10.1002/jbm.a.32690] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Current strategies to enhance gene transfer have focused on the development of vectors to increase the efficiency of DNA delivery. However, the extracellular matrix and microenvironment have a profound impact on numerous cellular activities including spreading and proliferation; two processes that have been associated with gene transfer efficiency. This study was designed to test the hypothesis that the presence of a biomineralized coating on biodegradable substrates would affect transgene expression following nonviral gene delivery. Thin films were prepared from polymeric microspheres, while biomineralized films were fabricated from microspheres previously soaked in modified simulated body fluid. Mineralized films were significantly more rigid and had widespread mineral coverage compared with nonmineralized substrates. Human mesenchymal stem cells (MSCs) were cultured on biomineralized or nonmineralized films and transfected with plasmid DNA condensed with linear polyethyleneimine (PEI). Compared with cells transfected on nonmineralized films, increases in gene expression were detected in the presence of biomineral at all charge ratios examined. We observed increased uptake of both PEI and DNA by cells on mineralized films. The results of these studies offer an approach to modulate gene delivery and improve the potential benefit of nonviral gene delivery approaches.
Collapse
Affiliation(s)
- Rameshwar R Rao
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA
| | | | | |
Collapse
|
27
|
Synergistic effects between natural histone mixtures and polyethylenimine in non-viral gene delivery in vitro. Int J Pharm 2010; 400:86-95. [PMID: 20816738 DOI: 10.1016/j.ijpharm.2010.08.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 08/18/2010] [Accepted: 08/25/2010] [Indexed: 10/19/2022]
Abstract
Nanoparticles made of plasmid DNA (pDNA) and cationic polymers are promising strategies for non-viral gene delivery. However, many cationic polymers are toxic to cells when used in higher concentrations. Positively charged proteins, such as histones, are biodegradable and a good alternative, especially for potential in vivo applications. It has previously been shown that histones are able to complex DNA and mediate transfection of cells. To investigate possible synergistic effects between the different histone types and to avoid the use of recombinant proteins, we analysed whether natural histone mixtures would be functional as gene carriers. Core and linker histones from calf thymus and from chicken erythrocytes were used to transfect different cell lines. The protein mixtures efficiently complexed the pDNA, and the resulting particles entered the cells. However, only marginal expression of the gene encoded by the pDNA was observed. Transfection rates increased drastically when minimal amounts of the basic polymer polyethylenimine (PEI) were added to the particles. Neither PEI nor histones alone mediated any transfection under the conditions where a combination of both worked efficiently, and the combined particles were well tolerated by the cells. These results demonstrate that histone mixtures from natural sources in combination with minimal amounts of PEI can be used as gene carriers. This might have consequences for the development of novel gene delivery strategies, such as DNA vaccines, with minimal side-effects.
Collapse
|
28
|
Dhaliwal A, Maldonado M, Han Z, Segura T. Differential uptake of DNA-poly(ethylenimine) polyplexes in cells cultured on collagen and fibronectin surfaces. Acta Biomater 2010; 6:3436-47. [PMID: 20371304 DOI: 10.1016/j.actbio.2010.03.038] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 03/27/2010] [Accepted: 03/29/2010] [Indexed: 12/12/2022]
Abstract
Genetically modified bone marrow-derived mesenchymal stem cells (MSCs) have proven to be efficient cell carriers for local or systemic delivery of therapeutics as well as growth factors to augment tissue formation. However, efficient non-viral gene transfer to these cells is limiting their applicability. Although most studies have focused on designing more efficient condensation agents for DNA, our focus in this manuscript is to study the role of two extracellular matrix (ECM) proteins, collagen I (Col I) and fibronectin (Fn), on the ability of MSCs to become transfected. Here we report that plating MSCs on Col I-coated surfaces inhibits transfection, while plating MSCs on Fn-coated surfaces enhances transfection. The mechanism by which these ECM proteins affect non-viral gene transfer involves the endocytosis pathway used for polyplex uptake and intracellular tension. We found that Fn promoted internalization through clathrin-mediated endocytosis and that this pathway resulted in more efficient transfection than caveolae-mediated endocytosis and macropinocytosis. Further, the disruption of actin-myosin interactions resulted in an enhancement of gene transfer for cells plated on Fn-coated surfaces, but not for cells plated on Col I. We believe that the cellular microenvironment can be engineered to enhance the ability of cells to become transfected and that through understanding the mechanisms by which the ECM affects non-viral gene transfer better materials and transfection protocols can be realized.
Collapse
Affiliation(s)
- Anandika Dhaliwal
- Biomedical Engineering Interdepartmental Program, University of California, Los Angeles, CA, USA
| | | | | | | |
Collapse
|
29
|
Chemical vectors for gene delivery: uptake and intracellular trafficking. Curr Opin Biotechnol 2010; 21:640-5. [PMID: 20674331 DOI: 10.1016/j.copbio.2010.07.003] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 07/06/2010] [Indexed: 01/07/2023]
Abstract
Chemical vectors for non-viral gene delivery are based on engineered DNA nanoparticles produced with various range of macromolecules suitable to mimic some viral functions required for gene transfer. Many efforts have been undertaken these past years to identify cellular barriers that have to be passed for this issue. Here, we summarize the current status of knowledge on the uptake mechanism of DNA nanoparticles made with polymers and liposomes, their endosomal escape, cytosolic diffusion, and nuclear import of pDNA. Studies reported these past years regarding pDNA nanoparticles endocytosis indicated that there is no clear evident relationship between the ways of entry and the transfection efficiency. By contrast, the sequestration of pDNA in intracellular vesicles and the low number of pDNA close to the nuclear envelop are identified as the major intracellular barriers. So, intensive investigations to increase the cytosolic delivery of pDNA and its migration toward nuclear pores make sense to bring the transfection efficiency closer to that of viruses.
Collapse
|
30
|
Junk DNA enhances pEI-based non-viral gene delivery. Int J Pharm 2010; 390:76-83. [DOI: 10.1016/j.ijpharm.2009.08.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 05/15/2009] [Accepted: 08/25/2009] [Indexed: 11/21/2022]
|
31
|
Glover DJ, Leyton DL, Moseley GW, Jans DA. The efficiency of nuclear plasmid DNA delivery is a critical determinant of transgene expression at the single cell level. J Gene Med 2010; 12:77-85. [DOI: 10.1002/jgm.1406] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
32
|
Hermanns MI, Kasper J, Dubruel P, Pohl C, Uboldi C, Vermeersch V, Fuchs S, Unger RE, Kirkpatrick CJ. An impaired alveolar-capillary barrier in vitro: effect of proinflammatory cytokines and consequences on nanocarrier interaction. J R Soc Interface 2009; 7 Suppl 1:S41-54. [PMID: 19793744 DOI: 10.1098/rsif.2009.0288.focus] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The alveolar region of the lung is an important target for drug and gene delivery approaches. Treatment with drugs is often necessary under pathophysiological conditions, in which there is acute inflammation of the target organ. Therefore, in vitro models of the alveolar-capillary barrier, which mimic inflammatory conditions in the alveolar region, would be useful to analyse and predict effects of novel drugs on healthy or inflamed tissues. The epithelial cell line H441 was cultivated with primary isolated human pulmonary microvascular endothelial cells (HPMECs) or the endothelial cell line ISO-HAS-1 on opposite sides of a permeable filter support under physiological and inflammatory conditions. Both epithelial and endothelial cell types grew as polarized monolayers in bilayer coculture and were analysed in the presence and absence of the proinflammatory stimuli tumour necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma). In addition, the nanocarrier polyethyleneimine (PEI) was chosen as a model compound to study cell uptake (Oregon Green (OG)-labelled PEI) and gene transfer (PEI-pDNA complex). Upon treatment with TNF-alpha and IFN-gamma, both cocultures exhibited comparable effects on the trans-bilayer electrical resistance, the transport of sodium fluorescein and the increase in secondary cytokine release. Basolateral (endothelial side) exposure to TNF-alpha or simultaneous exposure to TNF-alpha and IFN-gamma generated an alveolar-capillary barrier with inflammation-like characteristics, impaired barrier function and a local disruption of the continuous apical labelling of the tight junction plaque protein zonula occludens-1 (ZO-1). Although transfection rates of 8 per cent were obtained for H441 cells in non-polarized monocultures, apical-basolateral-differentiated (polarized) H441 in coculture could not be transfected. After basolateral cytokine exposure, uptake of fluorescently labelled PEI in polarized H441 was predominantly detected in those areas with a local disruption of ZO-1 expression. Accordingly, transfected cells were only sparsely found in coculture after basolateral costimulation with TNF-alpha and IFN-gamma. We designed a coculture model that mimics both the structural architecture of the alveolar-capillary barrier and inflammatory mechanisms with consequences on barrier characteristics, cytokine production and nanoparticle interaction. Our model will be suitable to systematically study adsorption, uptake and trafficking of newly synthesized nanosized carriers under different physiological conditions.
Collapse
Affiliation(s)
- Maria Iris Hermanns
- Institute of Pathology, Medical University of Mainz, Johannes Gutenberg University, Langenbeckstrasse 1, Mainz 55101, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|