1
|
Kwizera R, Xie J, Nurse N, Yuan C, Kirchmaier AL. Impacts of Nucleosome Positioning Elements and Pre-Assembled Chromatin States on Expression and Retention of Transgenes. Genes (Basel) 2024; 15:1232. [PMID: 39336823 PMCID: PMC11431089 DOI: 10.3390/genes15091232] [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: 08/21/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND/OBJECTIVES Transgene applications, ranging from gene therapy to the development of stable cell lines and organisms, rely on maintaining the expression of transgenes. To date, the use of plasmid-based transgenes has been limited by the loss of their expression shortly after their delivery into the target cells. The short-lived expression of plasmid-based transgenes has been largely attributed to host-cell-mediated degradation and/or silencing of transgenes. The development of chromatin-based strategies for gene delivery has the potential to facilitate defining the requirements for establishing epigenetic states and to enhance transgene expression for numerous applications. METHODS To assess the impact of "priming" plasmid-based transgenes to adopt accessible chromatin states to promote gene expression, nucleosome positioning elements were introduced at promoters of transgenes, and vectors were pre-assembled into nucleosomes containing unmodified histones or mutants mimicking constitutively acetylated states at residues 9 and 14 of histone H3 or residue 16 of histone H4 prior to their introduction into cells, then the transgene expression was monitored over time. RESULTS DNA sequences capable of positioning nucleosomes could positively impact the expression of adjacent transgenes in a distance-dependent manner in the absence of their pre-assembly into chromatin. Intriguingly, the pre-assembly of plasmids into chromatin facilitated the prolonged expression of transgenes relative to plasmids that were not pre-packaged into chromatin. Interactions between pre-assembled chromatin states and nucleosome positioning-derived effects on expression were also assessed and, generally, nucleosome positioning played the predominant role in influencing gene expression relative to priming with hyperacetylated chromatin states. CONCLUSIONS Strategies incorporating nucleosome positioning elements and the pre-assembly of plasmids into chromatin prior to nuclear delivery can modulate the expression of plasmid-based transgenes.
Collapse
Affiliation(s)
- Ronard Kwizera
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Junkai Xie
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Nathan Nurse
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Ann L Kirchmaier
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
2
|
Khairnar P, Kolipaka T, Pandey G, Phatale V, Shah S, Srinivasarao DA, Saraf S, Srivastava S. Nanosponge-mediated oligonucleotide delivery: A cutting-edge technology towards cancer management. J Drug Deliv Sci Technol 2024; 91:105226. [DOI: 10.1016/j.jddst.2023.105226] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Goyal P, Malviya R. Advances in nuclei targeted delivery of nanoparticles for the management of cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188881. [PMID: 36965678 DOI: 10.1016/j.bbcan.2023.188881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/16/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
A carrier is inserted into the appropriate organelles (nucleus) in successful medication transport, crucial to achieving very effective illness treatment. Cell-membrane targeting is the major focus of using nuclei to localize delivery. It has been demonstrated that high quantities of anticancer drugs can be injected directly into the nuclei of cancer cells, causing the cancer cells to die and increasing the effectiveness of chemotherapy. There are several effective ways to functionalize Nanoparticles (NPs), including changing their chemical makeup or attaching functional groups to their surface to increase their ability to target organelles. To cause tumor cells to apoptosis, released medicines must engage with molecular targets on particular organelles when their concentration is high enough. Targeted medication delivery studies will increasingly focus on organelle-specific delivery.
Collapse
Affiliation(s)
- Priyanshi Goyal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India.
| |
Collapse
|
4
|
Vlach M, Coppens-Exandier H, Jamin A, Berchel M, Scaviner J, Chesné C, Montier T, Jaffrès PA, Corlu A, Loyer P. Liposome-Mediated Gene Transfer in Differentiated HepaRG™ Cells: Expression of Liver Specific Functions and Application to the Cytochrome P450 2D6 Expression. Cells 2022; 11:cells11233904. [PMID: 36497165 PMCID: PMC9737581 DOI: 10.3390/cells11233904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/19/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
The goal of this study was to establish a procedure for gene delivery mediated by cationic liposomes in quiescent differentiated HepaRG™ human hepatoma cells. We first identified several cationic lipids promoting efficient gene transfer with low toxicity in actively dividing HepG2, HuH7, BC2 and progenitor HepaRG™ human hepatoma cells. The lipophosphoramidate Syn1-based nanovector, which allowed the highest transfection efficiencies of progenitor HepaRG™ cells, was next used to transfect differentiated HepaRG™ cells. Lipofection of these cells using Syn1-based liposome was poorly efficient most likely because the differentiated HepaRG™ cells are highly quiescent. Thus, we engineered the differentiated HepaRG™ Mitogenic medium supplement (ADD1001) that triggered robust proliferation of differentiated cells. Importantly, we characterized the phenotypical changes occurring during proliferation of differentiated HepaRG™ cells and demonstrated that mitogenic stimulation induced a partial and transient decrease in the expression levels of some liver specific functions followed by a fast recovery of the full differentiation status upon removal of the mitogens. Taking advantage of the proliferation of HepaRG™ cells, we defined lipofection conditions using Syn1-based liposomes allowing transient expression of the cytochrome P450 2D6, a phase I enzyme poorly expressed in HepaRG cells, which opens new means for drug metabolism studies in HepaRG™ cells.
Collapse
Affiliation(s)
- Manuel Vlach
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Institut AGRO Rennes-Angers, F-35042 Rennes, France
| | - Hugo Coppens-Exandier
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Biopredic International, F-35760 Saint Grégoire, France
| | - Agnès Jamin
- Biopredic International, F-35760 Saint Grégoire, France
| | - Mathieu Berchel
- Univ. Brest, CNRS, CEMCA, UMR 6521, F-29238 Brest, France
- Plateforme BiogenOuest SynNanoVect, F-44035 Nantes, France
| | - Julien Scaviner
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Biopredic International, F-35760 Saint Grégoire, France
| | | | - Tristan Montier
- Plateforme BiogenOuest SynNanoVect, F-44035 Nantes, France
- Univ. Brest, INSERM, EFS, UMR 1078, GGB-GTCA, F-29200 Brest, France
| | - Paul-Alain Jaffrès
- Univ. Brest, CNRS, CEMCA, UMR 6521, F-29238 Brest, France
- Plateforme BiogenOuest SynNanoVect, F-44035 Nantes, France
| | - Anne Corlu
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Correspondence: (A.C.); (P.L.); Tel.: +33-(02)-23233873 (P.L.)
| | - Pascal Loyer
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Plateforme BiogenOuest SynNanoVect, F-44035 Nantes, France
- Correspondence: (A.C.); (P.L.); Tel.: +33-(02)-23233873 (P.L.)
| |
Collapse
|
5
|
Yang Y, Liu Z, Ma H, Cao M. Application of Peptides in Construction of Nonviral Vectors for Gene Delivery. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12224076. [PMID: 36432361 PMCID: PMC9693978 DOI: 10.3390/nano12224076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/13/2022] [Accepted: 11/16/2022] [Indexed: 05/29/2023]
Abstract
Gene therapy, which aims to cure diseases by knocking out, editing, correcting or compensating abnormal genes, provides new strategies for the treatment of tumors, genetic diseases and other diseases that are closely related to human gene abnormalities. In order to deliver genes efficiently to abnormal sites in vivo to achieve therapeutic effects, a variety of gene vectors have been designed. Among them, peptide-based vectors show superior advantages because of their ease of design, perfect biocompatibility and safety. Rationally designed peptides can carry nucleic acids into cells to perform therapeutic effects by overcoming a series of biological barriers including cellular uptake, endosomal escape, nuclear entrance and so on. Moreover, peptides can also be incorporated into other delivery systems as functional segments. In this review, we referred to the biological barriers for gene delivery in vivo and discussed several kinds of peptide-based nonviral gene vectors developed for overcoming these barriers. These vectors can deliver different types of genetic materials into targeted cells/tissues individually or in combination by having specific structure-function relationships. Based on the general review of peptide-based gene delivery systems, the current challenges and future perspectives in development of peptidic nonviral vectors for clinical applications were also put forward, with the aim of providing guidance towards the rational design and development of such systems.
Collapse
Affiliation(s)
- Yujie Yang
- State Key Laboratory of Heavy Oil Processing, Department of Biological and Energy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Zhen Liu
- State Key Laboratory of Heavy Oil Processing, Department of Biological and Energy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Hongchao Ma
- State Key Laboratory of Heavy Oil Processing, Department of Biological and Energy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Meiwen Cao
- State Key Laboratory of Heavy Oil Processing, Department of Biological and Energy Chemical Engineering, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| |
Collapse
|
6
|
Winogradoff D, Chou HY, Maffeo C, Aksimentiev A. Percolation transition prescribes protein size-specific barrier to passive transport through the nuclear pore complex. Nat Commun 2022; 13:5138. [PMID: 36050301 PMCID: PMC9437005 DOI: 10.1038/s41467-022-32857-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 08/18/2022] [Indexed: 11/19/2022] Open
Abstract
Nuclear pore complexes (NPCs) control biomolecular transport in and out of the nucleus. Disordered nucleoporins in the complex's pore form a permeation barrier, preventing unassisted transport of large biomolecules. Here, we combine coarse-grained simulations of experimentally derived NPC structures with a theoretical model to determine the microscopic mechanism of passive transport. Brute-force simulations of protein transport reveal telegraph-like behavior, where prolonged diffusion on one side of the NPC is interrupted by rapid crossings to the other. We rationalize this behavior using a theoretical model that reproduces the energetics and kinetics of permeation solely from statistics of transient voids within the disordered mesh. As the protein size increases, the mesh transforms from a soft to a hard barrier, enabling orders-of-magnitude reduction in permeation rate for proteins beyond the percolation size threshold. Our model enables exploration of alternative NPC architectures and sets the stage for uncovering molecular mechanisms of facilitated nuclear transport.
Collapse
Affiliation(s)
- David Winogradoff
- grid.35403.310000 0004 1936 9991Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA ,grid.35403.310000 0004 1936 9991Center for the Physics of Living Cells, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Han-Yi Chou
- grid.35403.310000 0004 1936 9991Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Christopher Maffeo
- grid.35403.310000 0004 1936 9991Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA ,grid.35403.310000 0004 1936 9991Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Aleksei Aksimentiev
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA. .,Center for the Physics of Living Cells, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA. .,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
7
|
Gholami L, Mahmoudi A, Kazemi Oskuee R, Malaekeh-Nikouei B. An overview of polyallylamine applications in gene delivery. Pharm Dev Technol 2022; 27:714-724. [PMID: 35880621 DOI: 10.1080/10837450.2022.2107014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
A chief objective of gene transportation studies is to manipulate clinically accepted carriers that can be utilized to combat incurable diseases. Despite various strategies, efficiency and application of these vectors have been hindered, owing to different obstacles. Polyallylamine (PAA) is a synthetic water-soluble, weak base cationic polymer with different properties that could be administrated as an ideal candidate for biomedical applications such as gene delivery, drug delivery, or even tissue engineering. However, some intrinsic properties of this polymer limit its application. The two associated problems with the use of PAA in gene delivery are low transfection efficiency (because of low buffering capacity) and cytotoxic effects attributed to intense cationic character. Most of the strategies for structural modification of the PAA structure have focused on introducing hydrophobic groups to the polymeric backbone that target both cytotoxicity and transfection. In this perspective, we concentrate on PAA as a gene delivery vehicle and the existing approaches for modification of this cationic polymer to give insight to researchers for exploitation of PAA as an efficient carrier in biomedical applications.
Collapse
Affiliation(s)
- Leila Gholami
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asma Mahmoudi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Kazemi Oskuee
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bizhan Malaekeh-Nikouei
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Mahajan S, Tang T. Polyethylenimine-DNA Nanoparticles under Endosomal Acidification and Implication to Gene Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:8382-8397. [PMID: 35759612 DOI: 10.1021/acs.langmuir.2c00952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Non-viral gene delivery using polyethylenimine (PEI) has shown tremendous promise as a therapeutic technique. Through the formation of nanoparticles (NPs), PEIs protect genetic material such as DNA from degradation. Escape of the NPs from endosomes and lysosomes is facilitated by PEI's buffering capacity over a wide range of pH. However, little is known about the effects of endosomal acidification on the morphology of the NPs. In this work, large-scale coarse-grained simulations performed to mimic endosomal acidification reveal that NPs undergo a resizing process that is highly dependent on the N/P ratio (ratio of PEI nitrogen to DNA phosphate) at which they are prepared. With a low N/P ratio, NPs further aggregate after endosomal acidification, whereas with a high N/P ratio they dissociate. The mechanisms behind such NP resizing and its consequences on endosomal escape and nuclear trafficking are discussed. Based on the findings, suggestions are made on the PEI architecture that may enhance NP dissociation driven by endosomal acidification.
Collapse
Affiliation(s)
- Subhamoy Mahajan
- Department of Mechanical Engineering, University of Alberta, Edmonton T6G 2R3, Alberta, Canada
| | - Tian Tang
- Department of Mechanical Engineering, University of Alberta, Edmonton T6G 2R3, Alberta, Canada
| |
Collapse
|
9
|
Rabiee N, Ghadiri AM, Alinezhad V, Sedaghat A, Ahmadi S, Fatahi Y, Makvandi P, Saeb MR, Bagherzadeh M, Asadnia M, Varma RS, Lima EC. Synthesis of green benzamide-decorated UiO-66-NH 2 for biomedical applications. CHEMOSPHERE 2022; 299:134359. [PMID: 35318020 DOI: 10.1016/j.chemosphere.2022.134359] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/01/2022] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
Metal-organic frameworks (MOFs) biocompatible systems can host enzymes/bacteria/viruses. Herein we synthesized a series of fatty acid amide hydrolase (FAAH)-decorated UiO-66-NH2 based on Citrus tangerine leaf extract for drug delivery and biosensor applications. Five chemically manipulated FAAH-like benzamides were localized on the UiO-66-NH2 surface with physical interactions. Comprehensive cellular and molecular analyses were conducted on HEK-293, HeLa, HepG2, PC12, MCF-7, and HT-29 cell lines (cytotoxicity assessment after 24 and 48 h). MTT results proved above 95 and 50% relative cell viability in the absence and presence of the drug, respectively. A complete targeted drug-releasing capability of nanocarriers was demonstrated after capping with leaf extract from Citrus tangerine, with a stimuli-responsive effect in acidic media. Targeted delivery was complete to the nucleus and cytoplasm of HT-29 cell, but merely to the cytoplasm of HeLa cell lines. Nanocarrier could be targeted for drug delivery to the cytoplasm of the HeLa cell line and to both the nucleus and cytoplasm of HT-29 cell lines. MOF-based nanocarriers proved authentic in vivo towards kidney and liver tissues with targeted cancerous cells efficiently. Besides, FAAH-like molecules revealed optical biosensor potential with high selectivity (even ˂5 nM LOD) towards ssDNA, sgRNA, and Anti-cas9 proteins.
Collapse
Affiliation(s)
- Navid Rabiee
- Department of Physics, Sharif University of Technology, Tehran, P.O. Box 11155-9161, Iran; School of Engineering, Macquarie University, Sydney, New South Wales, 2109, Australia.
| | | | - Vida Alinezhad
- Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | - Anna Sedaghat
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepideh Ahmadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yousef Fatahi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 14155-6451, Iran; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 14155-6451, Iran
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Materials Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, G. Narutowicza 11/12 80-233, Gdańsk, Poland
| | | | - Mohsen Asadnia
- School of Engineering, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Rajender S Varma
- Regional Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacky University, Šlechtitelů 27, 783 71, Olomouc, Czech Republic
| | - Eder C Lima
- Institute of Chemistry, Federal University of Rio Grande Do Sul (UFRGS), Av. Bento Goncalves 9500, Postal Box, 15003, ZIP, 91501-970, Brazil.
| |
Collapse
|
10
|
Wu L, Zhou W, Lin L, Chen A, Feng J, Qu X, Zhang H, Yue J. Delivery of therapeutic oligonucleotides in nanoscale. Bioact Mater 2022; 7:292-323. [PMID: 34466734 PMCID: PMC8379367 DOI: 10.1016/j.bioactmat.2021.05.038] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/28/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023] Open
Abstract
Therapeutic oligonucleotides (TOs) represent one of the most promising drug candidates in the targeted cancer treatment due to their high specificity and capability of modulating cellular pathways that are not readily druggable. However, efficiently delivering of TOs to cancer cellular targets is still the biggest challenge in promoting their clinical translations. Emerging as a significant drug delivery vector, nanoparticles (NPs) can not only protect TOs from nuclease degradation and enhance their tumor accumulation, but also can improve the cell uptake efficiency of TOs as well as the following endosomal escape to increase the therapeutic index. Furthermore, targeted and on-demand drug release of TOs can also be approached to minimize the risk of toxicity towards normal tissues using stimuli-responsive NPs. In the past decades, remarkable progresses have been made on the TOs delivery based on various NPs with specific purposes. In this review, we will first give a brief introduction on the basis of TOs as well as the action mechanisms of several typical TOs, and then describe the obstacles that prevent the clinical translation of TOs, followed by a comprehensive overview of the recent progresses on TOs delivery based on several various types of nanocarriers containing lipid-based nanoparticles, polymeric nanoparticles, gold nanoparticles, porous nanoparticles, DNA/RNA nanoassembly, extracellular vesicles, and imaging-guided drug delivery nanoparticles.
Collapse
Affiliation(s)
- Lei Wu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Wenhui Zhou
- Pharmaceutical Sciences Laboratory and Turku Bioscience Centre, Åbo Akademi University, Turku, 20520, Finland
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Lihua Lin
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Anhong Chen
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Jing Feng
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Xiangmeng Qu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory and Turku Bioscience Centre, Åbo Akademi University, Turku, 20520, Finland
| | - Jun Yue
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| |
Collapse
|
11
|
Nasr SS, Lee S, Thiyagarajan D, Boese A, Loretz B, Lehr CM. Co-Delivery of mRNA and pDNA Using Thermally Stabilized Coacervate-Based Core-Shell Nanosystems. Pharmaceutics 2021; 13:1924. [PMID: 34834339 PMCID: PMC8619316 DOI: 10.3390/pharmaceutics13111924] [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: 09/03/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 11/17/2022] Open
Abstract
Co-delivery of different species of protein-encoding polynucleotides, e.g., messenger RNA (mRNA) and plasmid DNA (pDNA), using the same nanocarrier is an interesting topic that remains scarcely researched in the field of nucleic acid delivery. The current study hence aims to explore the possibility of the simultaneous delivery of mRNA (mCherry) and pDNA (pAmCyan) using a single nanocarrier. The latter is based on gelatin type A, a biocompatible, and biodegradable biopolymer of broad pharmaceutical application. A core-shell nanostructure is designed with a thermally stabilized gelatin-pDNA coacervate in its center. Thermal stabilization enhances the core's colloidal stability and pDNA shielding effect against nucleases as confirmed by nanoparticle tracking analysis and gel electrophoresis, respectively. The stabilized, pDNA-loaded core is coated with the cationic peptide protamine sulfate to enable additional surface-loading with mRNA. The dual-loaded core-shell system transfects murine dendritic cell line DC2.4 with both fluorescent reporter mRNA and pDNA simultaneously, showing a transfection efficiency of 61.4 ± 21.6% for mRNA and 37.6 ± 19.45% for pDNA, 48 h post-treatment, whereas established commercial, experimental, and clinical transfection reagents fail. Hence, the unique co-transfectional capacity and the negligible cytotoxicity of the reported system may hold prospects for vaccination among other downstream applications.
Collapse
Affiliation(s)
- Sarah S. Nasr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, 66123 Saarbrücken, Germany; (S.S.N.); (S.L.); (D.T.); (A.B.); (C.-M.L.)
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Sangeun Lee
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, 66123 Saarbrücken, Germany; (S.S.N.); (S.L.); (D.T.); (A.B.); (C.-M.L.)
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Durairaj Thiyagarajan
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, 66123 Saarbrücken, Germany; (S.S.N.); (S.L.); (D.T.); (A.B.); (C.-M.L.)
| | - Annette Boese
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, 66123 Saarbrücken, Germany; (S.S.N.); (S.L.); (D.T.); (A.B.); (C.-M.L.)
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, 66123 Saarbrücken, Germany; (S.S.N.); (S.L.); (D.T.); (A.B.); (C.-M.L.)
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, 66123 Saarbrücken, Germany; (S.S.N.); (S.L.); (D.T.); (A.B.); (C.-M.L.)
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| |
Collapse
|
12
|
Tarvirdipour S, Skowicki M, Schoenenberger CA, Palivan CG. Peptide-Assisted Nucleic Acid Delivery Systems on the Rise. Int J Mol Sci 2021; 22:9092. [PMID: 34445799 PMCID: PMC8396486 DOI: 10.3390/ijms22169092] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Concerns associated with nanocarriers' therapeutic efficacy and side effects have led to the development of strategies to advance them into targeted and responsive delivery systems. Owing to their bioactivity and biocompatibility, peptides play a key role in these strategies and, thus, have been extensively studied in nanomedicine. Peptide-based nanocarriers, in particular, have burgeoned with advances in purely peptidic structures and in combinations of peptides, both native and modified, with polymers, lipids, and inorganic nanoparticles. In this review, we summarize advances on peptides promoting gene delivery systems. The efficacy of nucleic acid therapies largely depends on cell internalization and the delivery to subcellular organelles. Hence, the review focuses on nanocarriers where peptides are pivotal in ferrying nucleic acids to their site of action, with a special emphasis on peptides that assist anionic, water-soluble nucleic acids in crossing the membrane barriers they encounter on their way to efficient function. In a second part, we address how peptides advance nanoassembly delivery tools, such that they navigate delivery barriers and release their nucleic acid cargo at specific sites in a controlled fashion.
Collapse
Affiliation(s)
- Shabnam Tarvirdipour
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- Department of Biosystem Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Michal Skowicki
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Cora-Ann Schoenenberger
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Cornelia G. Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| |
Collapse
|
13
|
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
|
14
|
Lin G, Revia RA, Zhang M. Inorganic Nanomaterial-Mediated Gene Therapy in Combination with Other Antitumor Treatment Modalities. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007096. [PMID: 34366761 PMCID: PMC8336227 DOI: 10.1002/adfm.202007096] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Indexed: 05/05/2023]
Abstract
Cancer is a genetic disease originating from the accumulation of gene mutations in a cellular subpopulation. Although many therapeutic approaches have been developed to treat cancer, recent studies have revealed an irrefutable challenge that tumors evolve defenses against some therapies. Gene therapy may prove to be the ultimate panacea for cancer by correcting the fundamental genetic errors in tumors. The engineering of nanoscale inorganic carriers of cancer therapeutics has shown promising results in the efficacious and safe delivery of nucleic acids to treat oncological diseases in small-animal models. When these nanocarriers are used for co-delivery of gene therapeutics along with auxiliary treatments, the synergistic combination of therapies often leads to an amplified health benefit. In this review, an overview of the inorganic nanomaterials developed for combinatorial therapies of gene and other treatment modalities is presented. First, the main principles of using nucleic acids as therapeutics, inorganic nanocarriers for medical applications and delivery of gene/drug payloads are introduced. Next, the utility of recently developed inorganic nanomaterials in different combinations of gene therapy with each of chemo, immune, hyperthermal, and radio therapy is examined. Finally, current challenges in the clinical translation of inorganic nanomaterial-mediated therapies are presented and outlooks for the field are provided.
Collapse
Affiliation(s)
- Guanyou Lin
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Richard A Revia
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
15
|
van den Berg AIS, Yun CO, Schiffelers RM, Hennink WE. Polymeric delivery systems for nucleic acid therapeutics: Approaching the clinic. J Control Release 2021; 331:121-141. [PMID: 33453339 DOI: 10.1016/j.jconrel.2021.01.014] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/26/2020] [Accepted: 01/08/2021] [Indexed: 12/20/2022]
Abstract
Gene therapy using nucleic acids has many clinical applications for the treatment of diseases with a genetic origin as well as for the development of innovative vaccine formulations. Since nucleic acids in their free form are rapidly degraded by nucleases present in extracellular matrices, have poor pharmacokinetics and hardly pass cellular membranes, carrier systems are required. Suitable carriers that protect the nucleic acid payload against enzymatic attack, prolong circulation time after systemic administration and assist in cellular binding and internalization are needed to develop nucleic acid based drug products. Viral vectors have been investigated and are also clinically used as delivery vehicles. However, some major drawbacks are associated with their use. Therefore there has been substantial attention on the use of non-viral carrier systems based on cationic lipids and polymers. This review focuses on the properties of polymer-based nucleic acid formulations, also referred as polyplexes. Different polymeric systems are summarized, and the cellular barriers polyplexes encounter and ways to tackle these are discussed. Finally attention is given to the clinical status of non-viral nucleic acid formulations.
Collapse
Affiliation(s)
- Annette I S van den Berg
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, the Netherlands
| | - Chae-Ok Yun
- Institute of Nano Science and Technology, Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Raymond M Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, the Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584, CG, Utrecht, the Netherlands.
| |
Collapse
|
16
|
Fu X, Shi Y, Qi T, Qiu S, Huang Y, Zhao X, Sun Q, Lin G. Precise design strategies of nanomedicine for improving cancer therapeutic efficacy using subcellular targeting. Signal Transduct Target Ther 2020; 5:262. [PMID: 33154350 PMCID: PMC7644763 DOI: 10.1038/s41392-020-00342-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/26/2020] [Accepted: 09/14/2020] [Indexed: 01/10/2023] Open
Abstract
Therapeutic efficacy against cancer relies heavily on the ability of the therapeutic agents to reach their final targets. The optimal targets of most cancer therapeutic agents are usually biological macromolecules at the subcellular level, which play a key role in carcinogenesis. Therefore, to improve the therapeutic efficiency of drugs, researchers need to focus on delivering not only the therapeutic agents to the target tissues and cells but also the drugs to the relevant subcellular structures. In this review, we discuss the most recent construction strategies and release patterns of various cancer cell subcellular-targeting nanoformulations, aiming at providing guidance in the overall design of precise nanomedicine. Additionally, future challenges and potential perspectives are illustrated in the hope of enhancing anticancer efficacy and accelerating the translational progress of precise nanomedicine.
Collapse
Affiliation(s)
- Xianglei Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yanbin Shi
- School of Mechanical and Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, China
| | - Tongtong Qi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Shengnan Qiu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yi Huang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Xiaogang Zhao
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China
| | - Qifeng Sun
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China
| | - Guimei Lin
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
17
|
Li K, Liu CJ, Zhang XZ. Multifunctional peptides for tumor therapy. Adv Drug Deliv Rev 2020; 160:36-51. [PMID: 33080257 DOI: 10.1016/j.addr.2020.10.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022]
Abstract
Controlled nano-systems for drug delivery are designed to deliver therapeutical drugs to desirable sites on demand. Due to the diverse physiological functions of peptides, it is reasonable to introduce peptides into anti-tumor nano-system. The integration of peptides into nanomaterials has complementary advantages, which not only avoids the rapid degradation of peptides in vivo, but also improves the intelligence and functionality of the nano-system. We summarized the functional peptides with targeting and stimulus-responsive properties, and the present review outlined the most relevant and recent developed peptide-based multifunctional nanomaterials for tumor therapy.
Collapse
|
18
|
Chen W, Sun Z, Lu L. Targeted Engineering of Medicinal Chemistry for Cancer Therapy: Recent Advances and Perspectives. Angew Chem Int Ed Engl 2020; 60:5626-5643. [PMID: 32096328 DOI: 10.1002/anie.201914511] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Indexed: 12/13/2022]
Abstract
Severe side effects and poor therapeutic efficacy are the main drawbacks of current anticancer drugs. These problems can be mitigated by targeting, but the targeting efficacy of current drugs is poor and urgently needs improvement. Taking this into consideration, this Review first summarizes the current targeting strategies for cancer therapy in terms of cancer tissue and organelles. Then, we analyse the systematic targeting of anticancer drugs and conclude that a typical journey for a targeted drug administered by intravenous injection is a CTIO cascade of at least four steps. Furthermore, to ensure high overall targeting efficacy, the properties of a targeting drug needed in each step are further analysed, and some guidelines for structure optimization to obtain effective targeting drugs are offered. Finally, some viewpoints highlighting the crucial problems and potential challenges of future research on targeted cancer therapy are presented. This review could actively promote the development of precision medicine against cancer.
Collapse
Affiliation(s)
- Weihua Chen
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, University of Science and Technology of China, Changchun, 130022, China
| | - Zhen Sun
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, University of Science and Technology of China, Changchun, 130022, China
| | - Lehui Lu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, University of Science and Technology of China, Changchun, 130022, China
| |
Collapse
|
19
|
Zielgerichtete Wirkstoffe für die Krebstherapie: Aktuelle Entwicklungen und Perspektiven. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201914511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
20
|
Rueda-Gensini L, Cifuentes J, Castellanos MC, Puentes PR, Serna JA, Muñoz-Camargo C, Cruz JC. Tailoring Iron Oxide Nanoparticles for Efficient Cellular Internalization and Endosomal Escape. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1816. [PMID: 32932957 PMCID: PMC7559083 DOI: 10.3390/nano10091816] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022]
Abstract
Iron oxide nanoparticles (IONs) have been widely explored for biomedical applications due to their high biocompatibility, surface-coating versatility, and superparamagnetic properties. Upon exposure to an external magnetic field, IONs can be precisely directed to a region of interest and serve as exceptional delivery vehicles and cellular markers. However, the design of nanocarriers that achieve an efficient endocytic uptake, escape lysosomal degradation, and perform precise intracellular functions is still a challenge for their application in translational medicine. This review highlights several aspects that mediate the activation of the endosomal pathways, as well as the different properties that govern endosomal escape and nuclear transfection of magnetic IONs. In particular, we review a variety of ION surface modification alternatives that have emerged for facilitating their endocytic uptake and their timely escape from endosomes, with special emphasis on how these can be manipulated for the rational design of cell-penetrating vehicles. Moreover, additional modifications for enhancing nuclear transfection are also included in the design of therapeutic vehicles that must overcome this barrier. Understanding these mechanisms opens new perspectives in the strategic development of vehicles for cell tracking, cell imaging and the targeted intracellular delivery of drugs and gene therapy sequences and vectors.
Collapse
Affiliation(s)
- Laura Rueda-Gensini
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Javier Cifuentes
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Maria Claudia Castellanos
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Paola Ruiz Puentes
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Julian A. Serna
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Carolina Muñoz-Camargo
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Juan C. Cruz
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide 5005, Australia
| |
Collapse
|
21
|
Gigante A, Li M, Junghänel S, Hirschhäuser C, Knauer S, Schmuck C. Non-viral transfection vectors: are hybrid materials the way forward? MEDCHEMCOMM 2019; 10:1692-1718. [PMID: 32180915 PMCID: PMC7053704 DOI: 10.1039/c9md00275h] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/12/2019] [Indexed: 12/18/2022]
Abstract
Transfection is a process by which oligonucleotides (DNA or RNA) are delivered into living cells. This allows the synthesis of target proteins as well as their inhibition (gene silencing). However, oligonucleotides cannot cross the plasma membrane by themselves; therefore, efficient carriers are needed for successful gene delivery. Recombinant viruses are among the earliest described vectors. Unfortunately, they have severe drawbacks such as toxicity and immunogenicity. In this regard, the development of non-viral transfection vectors has attracted increasing interests, and has become an important field of research. In the first part of this review we start with a tutorial introduction into the biological backgrounds of gene transfection followed by the classical non-viral vectors (cationic organic carriers and inorganic nanoparticles). In the second part we highlight selected recent reports, which demonstrate that hybrid vectors that combine key features of classical carriers are a remarkable strategy to address the current challenges in gene delivery.
Collapse
Affiliation(s)
- A Gigante
- Institute of Organic Chemistry , University of Duisburg-Essen , 45141 Essen , Germany .
| | - M Li
- Institute of Organic Chemistry , University of Duisburg-Essen , 45141 Essen , Germany .
| | - S Junghänel
- Institute of Organic Chemistry , University of Duisburg-Essen , 45141 Essen , Germany .
- Biomedical Technology Center of the Medical Faculty , University of Muenster , Muenster , Germany
| | - C Hirschhäuser
- Institute of Organic Chemistry , University of Duisburg-Essen , 45141 Essen , Germany .
| | - S Knauer
- Faculty of Biology , University of Duisburg-Essen , 45141 Essen , Germany
| | - C Schmuck
- Institute of Organic Chemistry , University of Duisburg-Essen , 45141 Essen , Germany .
| |
Collapse
|
22
|
Li Q, Hao X, Wang H, Guo J, Ren XK, Xia S, Zhang W, Feng Y. Multifunctional REDV-G-TAT-G-NLS-Cys peptide sequence conjugated gene carriers to enhance gene transfection efficiency in endothelial cells. Colloids Surf B Biointerfaces 2019; 184:110510. [PMID: 31561046 DOI: 10.1016/j.colsurfb.2019.110510] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/20/2019] [Accepted: 09/15/2019] [Indexed: 01/31/2023]
Abstract
Rapid endothelialization on small diameter artificial blood vessels is an effective strategy to facilitate long-term patency and inhibit thrombosis. The gene delivery can enhance the proliferation and migration of endothelial cells (ECs), which is beneficial for rapid endothelialization. REDV-G-TAT-G-NLS-Cys (abbreviated as TP-G) peptide could weakly condense pEGFP-ZNF580 (pZNF580) and transfect ECs, but its transfection efficiency was still very low because of its low positive charge, low stability and weak endosome escape ability. In order to develop more stable and efficient gene carriers with low cytotoxicity, in the present study, we conjugated different amounts of TP-G peptide onto poly(lactide-co-glycolide)-g-polyethylenimine (PLGA-g-PEI) amphiphilic copolymers via a hetero-poly(ethylene glycol) spacer (OPSS-PEG-NHS). The TP-G peptide and PEI could cooperatively and strongly condense pZNF580. The carrier's cytotoxicity was reduced by the introduction of poly(ethylene glycol) spacer. They condensed pZNF580 to form gene complexes (PPP-TP-G/pZNF580) with suitable size and positive zeta potential for gene delivery. The transfected ECs promoted their migration ability as demonstrated by cell migration assay. The results of cellular uptake and confocal laser scanning microscopy showed significantly high internalization efficiency, endosomal/lysosomal escape and nucleus location of pZNF580 by this multifunctional TP-G peptide sequence conjugated gene delivery system. Furthermore, several inhibitors were used to study the cellular uptake pathways of PPP-TP-G/pZNF580 complexes. The results showed that PPP-TP-G2/Cy5-oligonucleotide complexes exhibited the optimized endocytosis pathways which facilitated for cellular uptake. In conclusion, the multifunctional TP-G peptide conjugated gene carriers could promote the transfection efficiency due to the multifunction of REDV, cell-penetrating peptide and nuclear localization signal in the peptide sequence, which could be a suitable gene carrier for endothelialization.
Collapse
Affiliation(s)
- Qian Li
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
| | - Xuefang Hao
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
| | - Huaning Wang
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
| | - Jintang Guo
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
| | - Xiang-Kui Ren
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China; Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Tianjin 300350, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Shihai Xia
- Department of Hepatopancreatobiliary and Splenic Medicine, Affiliated Hospital, Logistics University of People's Armed Police Force, Tianjin 300162, China.
| | - Wencheng Zhang
- Department of Physiology and Pathophysiology, Logistics University of People's Armed Police Force, Tianjin 300162, China
| | - Yakai Feng
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China; Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Tianjin 300350, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China.
| |
Collapse
|
23
|
Abstract
Gene therapy as a strategy for disease treatment requires safe and efficient gene delivery systems that encapsulate nucleic acids and deliver them to effective sites in the cell.
Collapse
Affiliation(s)
- Ziyao Kang
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| | - Qingbin Meng
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| | - Keliang Liu
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| |
Collapse
|
24
|
Polyester-based nanoparticles for nucleic acid delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:983-994. [DOI: 10.1016/j.msec.2018.07.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 07/05/2018] [Accepted: 07/11/2018] [Indexed: 12/14/2022]
|
25
|
Wang J, Zaidi SSA, Hasnain A, Guo J, Ren X, Xia S, Zhang W, Feng Y. Multitargeting Peptide-Functionalized Star-Shaped Copolymers with Comblike Structure and a POSS-Core To Effectively Transfect Endothelial Cells. ACS Biomater Sci Eng 2018; 4:2155-2168. [DOI: 10.1021/acsbiomaterials.8b00235] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Jun Wang
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
| | - Syed Saqib Ali Zaidi
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
| | - Ali Hasnain
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
| | - Jintang Guo
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Tianjin 300350, China
| | - Xiangkui Ren
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Tianjin 300350, China
| | - Shihai Xia
- Department of Hepatopancreatobiliary and Splenic Medicine, Affiliated Hospital, Logistics University of People’s Armed Police Force, 220 Chenglin Road, Tianjin 300162, China
| | - Wencheng Zhang
- Department of Physiology and Pathophysiology, Logistics University of Chinese People’s Armed Police Force, 220 Chenglin Road, Tianjin 300162, China
| | - Yakai Feng
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Tianjin 300350, China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, P. R. China
| |
Collapse
|
26
|
Mullis AS, Schlichtmann BW, Narasimhan B, Cademartiri R, Mallapragada SK. Ligand-cascading nano-delivery devices to enable multiscale targeting of anti-neurodegenerative therapeutics. Biomed Mater 2018; 13:034102. [DOI: 10.1088/1748-605x/aaa778] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
27
|
Pan L, Liu J, Shi J. Cancer cell nucleus-targeting nanocomposites for advanced tumor therapeutics. Chem Soc Rev 2018; 47:6930-6946. [DOI: 10.1039/c8cs00081f] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent advances in the chemical design and synthesis of nuclear-targeted nanotherapeutics for combating tumors are summarized and highlighted.
Collapse
Affiliation(s)
- Limin Pan
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure
- Shanghai Institute of Ceramics
- Chinese Academy of Sciences
- Shanghai 200050
- China
| | - Jianan Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure
- Shanghai Institute of Ceramics
- Chinese Academy of Sciences
- Shanghai 200050
- China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure
- Shanghai Institute of Ceramics
- Chinese Academy of Sciences
- Shanghai 200050
- China
| |
Collapse
|
28
|
Abstract
Although viral vectors comprise the majority of gene delivery vectors, their various safety, production, and other practical concerns have left a research gap to be addressed. The non-viral vector space encompasses a growing variety of physical and chemical methods capable of gene delivery into the nuclei of target cells. Major physical methods described in this chapter are microinjection, electroporation, and ballistic injection, magnetofection, sonoporation, optical transfection, and localized hyperthermia. Major chemical methods described in this chapter are lipofection, polyfection, gold complexation, and carbon-based methods. Combination approaches to improve transfection efficiency or reduce immunological response have shown great promise in expanding the scope of non-viral gene delivery.
Collapse
Affiliation(s)
- Chi Hong Sum
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| | | | - Shirley Wong
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| | | |
Collapse
|
29
|
Non-Covalent Loading of Anti-Cancer Doxorubicin by Modularizable Peptide Self-Assemblies for a Nanoscale Drug Carrier. Molecules 2017; 22:molecules22111916. [PMID: 29113134 PMCID: PMC6150382 DOI: 10.3390/molecules22111916] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 10/30/2017] [Indexed: 11/16/2022] Open
Abstract
We prepared nanoscale, modularizable, self-assembled peptide nanoarchitectures with diameters less of than 20 nm by combining β-sheet-forming peptides tethering a cell-penetrating peptide or a nuclear localization signal sequence. We also found that doxorubicin (Dox), an anti-cancer drug, was non-covalently accommodated by the assemblies at a ratio of one Dox molecule per ten peptides. The Dox-loaded peptide assemblies facilitated cellular uptake and subsequent nuclear localization in HeLa cells, and induced cell death even at low Dox concentrations. This peptide nanocarrier motif is a promising platform for a biocompatible drug delivery system by altering the targeting head groups of the carrier peptides.
Collapse
|
30
|
Redkiewicz P, Stachyra A, Sawicka RA, Bocian K, Góra-Sochacka A, Kosson P, Sirko A. Immunogenicity of DNA Vaccine against H5N1 Containing Extended Kappa B Site: In Vivo Study in Mice and Chickens. Front Immunol 2017; 8:1012. [PMID: 28883819 PMCID: PMC5573718 DOI: 10.3389/fimmu.2017.01012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/07/2017] [Indexed: 01/21/2023] Open
Abstract
Influenza is one of the most important illnesses in the modern world, causing great public health losses each year due to the lack of medication and broadly protective, long-lasting vaccines. The development of highly immunogenic and safe vaccines is currently one of the major problems encountered in efficient influenza prevention. DNA vaccines represent a novel and powerful alternative to the conventional vaccine approaches. To improve the efficacy of the DNA vaccine against influenza H5N1, we inserted three repeated kappa B (κB) motifs, separated by a 5-bp nucleotide spacer, upstream of the cytomegalovirus promoter and downstream of the SV40 late polyadenylation signal. The κB motif is a specific DNA element (10pb-long) recognized by one of the most important transcription factors NFκB. NFκB is present in almost all animal cell types and upon cell stimulation under a variety of pathogenic conditions. NFκB is released from IκB and translocates to the nucleus and binds to κB sites, thereby leading to enhanced transcription and expression of downstream genes. We tested the variants of DNA vaccine with κB sites flanking the antigen expression cassette and without such sites in two animal models: chickens (broilers and layers) and mice (BALB/c). In chickens, the variant with κB sites stimulated stronger humoral response against the target antigen. In mice, the differences in humoral response were less apparent. Instead, it was possible to spot several gene expression differences in the spleens isolated from mice immunized with both variants. The results of our study indicate that modification of the sequence outside of the sequence encoding the antigen might enhance the immune response to the target but understanding the mechanisms responsible for this process requires further analysis.
Collapse
Affiliation(s)
- Patrycja Redkiewicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Stachyra
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Róz A Sawicka
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | - Anna Góra-Sochacka
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Piotr Kosson
- Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Sirko
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
31
|
Zhou Z, Liu X, Zhu D, Wang Y, Zhang Z, Zhou X, Qiu N, Chen X, Shen Y. Nonviral cancer gene therapy: Delivery cascade and vector nanoproperty integration. Adv Drug Deliv Rev 2017; 115:115-154. [PMID: 28778715 DOI: 10.1016/j.addr.2017.07.021] [Citation(s) in RCA: 282] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 02/07/2023]
Abstract
Gene therapy represents a promising cancer treatment featuring high efficacy and limited side effects, but it is stymied by a lack of safe and efficient gene-delivery vectors. Cationic polymers and lipid-based nonviral gene vectors have many advantages and have been extensively explored for cancer gene delivery, but their low gene-expression efficiencies relative to viral vectors limit their clinical translations. Great efforts have thus been devoted to developing new carrier materials and fabricating functional vectors aimed at improving gene expression, but the overall efficiencies are still more or less at the same level. This review analyzes the cancer gene-delivery cascade and the barriers, the needed nanoproperties and the current strategies for overcoming these barriers, and outlines PEGylation, surface-charge, size, and stability dilemmas in vector nanoproperties to efficiently accomplish the cancer gene-delivery cascade. Stability, surface, and size transitions (3S Transitions) are proposed to resolve those dilemmas and strategies to realize these transitions are comprehensively summarized. The review concludes with a discussion of the future research directions to design high-performance nonviral gene vectors.
Collapse
Affiliation(s)
- Zhuxian Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, 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, Zheda Road 38, 310027 Hangzhou, China
| | - Dingcheng Zhu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, 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, Zheda Road 38, 310027 Hangzhou, China
| | - Zhen Zhang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xuefei Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Nasha Qiu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xuesi Chen
- Changchun Institute of Applied Chemistry, Key Lab of Polymer Ecomaterials, Changchun, 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, Zheda Road 38, 310027 Hangzhou, China.
| |
Collapse
|
32
|
Peng H, Tang J, Zheng R, Guo G, Dong A, Wang Y, Yang W. Nuclear-Targeted Multifunctional Magnetic Nanoparticles for Photothermal Therapy. Adv Healthc Mater 2017; 6. [PMID: 28128891 DOI: 10.1002/adhm.201601289] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/21/2016] [Indexed: 11/11/2022]
Abstract
The pursuit of multifunctional, innovative, more efficient, and safer cancer treatment has gained increasing interest in the research of preclinical nanoparticle-mediated photothermal therapy (PTT). Cell nucleus is recognized as the ideal target for cancer treatment because it plays a central role in genetic information and the transcription machinery reside. In this work, an efficient nuclear-targeted PTT strategy is proposed using transferrin and TAT peptide (TAT: YGRKKRRQRRR) conjugated monodisperse magnetic nanoparticles, which can be readily functionalized and stabilized for potential diagnostic and therapeutic applications. The monodisperse magnetic nanoparticles exhibit high photothermal conversion efficiency (≈37%) and considerable photothermal stability. They also show a high magnetization value and transverse relaxivity (207.1 mm-1 s-1 ), which could be applied for magnetic resonance imaging. The monodisperse magnetic nanoparticles conjugated with TAT peptides can efficiently target the nucleus and achieve the imaging-guided function, efficient cancer cells killing ability. Therefore, this work may present a practicable strategy to develop subcellular organelle targeted PTT agents for simultaneous cancer targeting, imaging, and therapy.
Collapse
Affiliation(s)
- Haibao Peng
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science; Fudan University; Shanghai 200433 China
| | - Jing Tang
- Division of Critical Care Medicine; Boston Children's Hospital; Harvard Medical School; 300 Longwood Avenue Boston MA 02115 USA
| | - Rui Zheng
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science; Fudan University; Shanghai 200433 China
| | - Guannan Guo
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science; Fudan University; Shanghai 200433 China
| | - Angang Dong
- Department of Chemistry and Collaborative Innovation Center of Chemistry for Energy Materials; Fudan University; Shanghai 200433 China
| | - Yajun Wang
- Department of Chemistry and Collaborative Innovation Center of Chemistry for Energy Materials; Fudan University; Shanghai 200433 China
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science; Fudan University; Shanghai 200433 China
| |
Collapse
|
33
|
McErlean EM, McCrudden CM, McCarthy HO. Delivery of nucleic acids for cancer gene therapy: overcoming extra- and intra-cellular barriers. Ther Deliv 2016; 7:619-37. [PMID: 27582234 DOI: 10.4155/tde-2016-0049] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The therapeutic potential of cancer gene therapy has been limited by the difficulty of delivering genetic material to target sites. Various biological and molecular barriers exist which need to be overcome before effective nonviral delivery systems can be applied successfully in oncology. Herein, various barriers are described and strategies to circumvent such obstacles are discussed, considering both the extracellular and intracellular setting. Development of multifunctional delivery systems holds much promise for the progression of gene delivery, and a growing body of evidence supports this approach involving rational design of vectors, with a unique molecular architecture. In addition, the potential application of composite gene delivery platforms is highlighted which may provide an alternative delivery strategy to traditional systemic administration.
Collapse
|
34
|
Xu Y, Liang W, Qiu Y, Cespi M, Palmieri GF, Mason AJ, Lam JKW. Incorporation of a Nuclear Localization Signal in pH Responsive LAH4-L1 Peptide Enhances Transfection and Nuclear Uptake of Plasmid DNA. Mol Pharm 2016; 13:3141-52. [PMID: 27458925 DOI: 10.1021/acs.molpharmaceut.6b00338] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The major intracellular barriers associated with DNA delivery using nonviral vectors are inefficient endosomal/lysosomal escape and poor nuclear uptake. LAH4-L1, a pH responsive cationic amphipathic peptide, is an efficient DNA delivery vector that promotes the release of nucleic acid into cytoplasm through endosomal escape. Here we further enhance the DNA transfection efficiency of LAH4-L1 by incorporating nuclear localizing signal (NLS) to promote nuclear importation. Four NLSs were investigated: Simian virus 40 (SV40) large T-antigen derived NLS, nucleoplasmin targeting signal, M9 sequence, and the reverse SV40 derived NLS. All peptides tested were able to form positively charged nanosized complexes with DNA. Significant improvement in DNA transfection was observed in slow-dividing epithelial cancer cells (Calu-3), macrophages (RAW264.7), dendritic cells (JAWSII), and thymidine-induced growth-arrested cells, but not in rapidly dividing cells (A549). Among the four NLS-modified peptides, PK1 (modified with SV40 derived NLS) and PK2 (modified with reverse SV40 derived NLS) were the most consistent in improving DNA transfection; up to a 10-fold increase in gene expression was observed for PK1 and PK2 over the unmodified LAH4-L1. Additionally PK1 and PK2 were shown to enhance cellular uptake as well as nuclear entry of DNA. Overall, we show that the incorporation of SV40 derived NLS, in particular, to LAH4-L1 is a promising strategy to improve DNA delivery efficiency in slow-dividing cells and dendritic cells, with development potential for in vivo applications and as a DNA vaccine carrier.
Collapse
Affiliation(s)
- Yingying Xu
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong , 21 Sassoon Road, Pokfulam, Hong Kong
| | - Wanling Liang
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong , 21 Sassoon Road, Pokfulam, Hong Kong
| | - Yingshan Qiu
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong , 21 Sassoon Road, Pokfulam, Hong Kong
| | - Marco Cespi
- School of Pharmacy, University of Camerino , Via S. Agostino 1, 62032 Camerino (MC), Italy
| | - Giovanni F Palmieri
- School of Pharmacy, University of Camerino , Via S. Agostino 1, 62032 Camerino (MC), Italy
| | - A James Mason
- Institute of Pharmaceutical Science, King's College London , 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Jenny K W Lam
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong , 21 Sassoon Road, Pokfulam, Hong Kong
| |
Collapse
|
35
|
Samudram A, Mangalassery BM, Kowshik M, Patincharath N, Varier GK. Passive permeability and effective pore size of HeLa cell nuclear membranes. Cell Biol Int 2016; 40:991-8. [PMID: 27338984 DOI: 10.1002/cbin.10640] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/18/2016] [Indexed: 12/12/2022]
Abstract
Nuclear pore complexes in the nuclear membrane act as the sole gateway of transport of molecules from the cytoplasm to the nucleus and vice versa. Studies on biomolecular transport through nuclear membranes provide vital data on the nuclear pore complexes. In this work, we use fluorescein isothiocyanate-labeled dextran molecules as a model system and study the passive nuclear import of biomolecules through nuclear pore complexes in digitonin-permeabilized HeLa cells. Experiments are carried out under transient conditions in the time lapse imaging scheme using an in-house constructed confocal laser scanning microscope. Transport rates of dextran molecules having molecular weights of 4-70 kDa corresponding to Stokes radius of 1.4-6 nm are determined. Analyzing the permeability of the nuclear membrane for different sizes the effective pore radius of HeLa cell nuclear membrane is determined to be 5.3 nm, much larger than the value reported earlier using proteins as probe molecules. The range of values reported for the nuclear pore radius suggest that they may not be rigid structures and it is quite probable that the effective pore size of nuclear pore complexes is critically dependent on the probe molecules and on the environmental factors.
Collapse
Affiliation(s)
- Arunkarthick Samudram
- Department of Physics, Birla Institute of Technology and Science, Pilani-KK Birla Goa Campus, Zuari Nagar, 403 726, Goa, India.,Department of Biological Sciences, Birla Institute of Technology and Science, Pilani-KK Birla Goa Campus, Zuari Nagar, 403 726, Goa, India
| | - Bijeesh M Mangalassery
- Department of Physics, Birla Institute of Technology and Science, Pilani-KK Birla Goa Campus, Zuari Nagar, 403 726, Goa, India
| | - Meenal Kowshik
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani-KK Birla Goa Campus, Zuari Nagar, 403 726, Goa, India
| | - Nandakumar Patincharath
- Department of Physics, Birla Institute of Technology and Science, Pilani-KK Birla Goa Campus, Zuari Nagar, 403 726, Goa, India
| | - Geetha K Varier
- Department of Physics, Birla Institute of Technology and Science, Pilani-KK Birla Goa Campus, Zuari Nagar, 403 726, Goa, India
| |
Collapse
|
36
|
Hu JJ, Xiao D, Zhang XZ. Advances in Peptide Functionalization on Mesoporous Silica Nanoparticles for Controlled Drug Release. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:3344-3359. [PMID: 27152737 DOI: 10.1002/smll.201600325] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 03/22/2016] [Indexed: 06/05/2023]
Abstract
During the last decade, using versatile, promising, and fascinating mesoporous silica nanoparticles (MSNs) as site-specific and stimuli-responsive drug delivery systems (DDSs) has received concentrated research interest. As one of the most attractive surface modification units, peptides have inherent bioactivity, biodegradability and biocompatibility. Recent progresses in the utilization of versatile peptides for surface functionalization of MSNs to achieve cell-specific targeting, fluorescence imaging, and intracellular diagnosis and treatment of tumors are summarized in this review. The various functional peptides decorated on the MSNs are introduced and classified into three types, including targeting peptides, stimuli-responsive peptides and multifunctional chimeric peptides. The limitations and challenges of peptide modified MSNs and their potential applications are further discussed.
Collapse
Affiliation(s)
- Jing-Jing Hu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Institute for Advanced Studies (IAS), Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Dong Xiao
- Key Laboratory of Biomedical Polymers of Ministry of Education, Institute for Advanced Studies (IAS), Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Institute for Advanced Studies (IAS), Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
37
|
Perez Ruiz de Garibay A. Endocytosis in gene therapy with non-viral vectors. Wien Med Wochenschr 2016; 166:227-35. [DOI: 10.1007/s10354-016-0450-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/01/2016] [Indexed: 01/06/2023]
|
38
|
Abstract
Plasmids are currently an indispensable molecular tool in life science research and a central asset for the modern biotechnology industry, supporting its mission to produce pharmaceutical proteins, antibodies, vaccines, industrial enzymes, and molecular diagnostics, to name a few key products. Furthermore, plasmids have gradually stepped up in the past 20 years as useful biopharmaceuticals in the context of gene therapy and DNA vaccination interventions. This review provides a concise coverage of the scientific progress that has been made since the emergence of what are called today plasmid biopharmaceuticals. The most relevant topics are discussed to provide researchers with an updated overview of the field. A brief outline of the initial breakthroughs and innovations is followed by a discussion of the motivation behind the medical uses of plasmids in the context of therapeutic and prophylactic interventions. The molecular characteristics and rationale underlying the design of plasmid vectors as gene transfer agents are described and a description of the most important methods used to deliver plasmid biopharmaceuticals in vivo (gene gun, electroporation, cationic lipids and polymers, and micro- and nanoparticles) is provided. The major safety issues (integration and autoimmunity) surrounding the use of plasmid biopharmaceuticals is discussed next. Aspects related to the large-scale manufacturing are also covered, and reference is made to the plasmid products that have received marketing authorization as of today.
Collapse
|
39
|
Abstract
DNA plasmids can be used to induce a protective (or therapeutic) immune response by delivering genes encoding vaccine antigens. That naked DNA (without the refinement of coat proteins or host evasion systems) can cross from outside the cell into the nucleus and be expressed is particularly remarkable given the sophistication of the immune system in preventing infection by pathogens. As a result of the ease, low cost, and speed of custom gene synthesis, DNA vaccines dangle a tantalizing prospect of the next wave of vaccine technology, promising individual designer vaccines for cancer or mass vaccines with a rapid response time to emerging pandemics. There is considerable enthusiasm for the use of DNA vaccination as an approach, but this enthusiasm should be tempered by the successive failures in clinical trials to induce a potent immune response. The technology is evolving with the development of improved delivery systems that increase expression levels, particularly electroporation and the incorporation of genetically encoded adjuvants. This review will introduce some key concepts in the use of DNA plasmids as vaccines, including how the DNA enters the cell and is expressed, how it induces an immune response, and a summary of clinical trials with DNA vaccines. The review also explores the advances being made in vector design, delivery, formulation, and adjuvants to try to realize the promise of this technology for new vaccines. If the immunogenicity and expression barriers can be cracked, then DNA vaccines may offer a step change in mass vaccination.
Collapse
|
40
|
Delivery of drugs to intracellular organelles using drug delivery systems: Analysis of research trends and targeting efficiencies. Int J Pharm 2015; 496:268-74. [DOI: 10.1016/j.ijpharm.2015.10.053] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 10/12/2015] [Accepted: 10/19/2015] [Indexed: 01/16/2023]
|
41
|
Li Y, Maciel D, Rodrigues J, Shi X, Tomás H. Biodegradable Polymer Nanogels for Drug/Nucleic Acid Delivery. Chem Rev 2015; 115:8564-608. [PMID: 26259712 DOI: 10.1021/cr500131f] [Citation(s) in RCA: 328] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yulin Li
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
- The State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, East China University of Science and Technology , Shanghai 200237, People's Republic of China
| | - Dina Maciel
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
| | - João Rodrigues
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
| | - Xiangyang Shi
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University , Shanghai 201620, People's Republic of China
| | - Helena Tomás
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
| |
Collapse
|
42
|
Chitosan-mediated non-viral gene delivery with improved serum stability and reduced cytotoxicity. BIOTECHNOL BIOPROC E 2015. [DOI: 10.1007/s12257-014-0450-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
43
|
|
44
|
Loughran SP, McCrudden CM, McCarthy HO. Designer peptide delivery systems for gene therapy. EUROPEAN JOURNAL OF NANOMEDICINE 2015. [DOI: 10.1515/ejnm-2014-0037] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
AbstractGene therapy has long been hailed as a revolutionary approach for the treatment of genetic diseases. The enthusiasm that greeted the harnessing of viruses for therapeutic DNA delivery has been tempered by concerns over safety. These concerns led to the development of alternative strategies for nucleic acid delivery to cells. One such strategy is the utilization of cationic peptides for the condensation of therapeutic DNA for delivery to its target. However, success of DNA as a therapy relies on its delivery to the nucleus of target cells, a process that is complicated by the many hurdles encountered following systemic administration. Non-viral peptide gene delivery strategies have sought inspiration from viruses in order to retain DNA delivering potency, but limit virulence. This review summarizes the progression of peptide-based DNA delivery systems, from rudimentary beginnings to the recent development of sophisticated multi-functional vectors that comprise distinct motifs with dedicated barrier evasion functions. The most promising peptides that achieve cell membrane permeabilization, endosomal escape and nuclear delivery are discussed.
Collapse
|
45
|
Gillard M, Jia Z, Hou JJC, Song M, Gray PP, Munro TP, Monteiro MJ. Intracellular Trafficking Pathways for Nuclear Delivery of Plasmid DNA Complexed with Highly Efficient Endosome Escape Polymers. Biomacromolecules 2014; 15:3569-76. [DOI: 10.1021/bm5008376] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Marianne Gillard
- Australian Institute for
Bioengineering and Nanotechnology, The University of Queensland, Brisbane QLD 4072, Australia
| | - Zhongfan Jia
- Australian Institute for
Bioengineering and Nanotechnology, The University of Queensland, Brisbane QLD 4072, Australia
| | - Jeff Jia Cheng Hou
- Australian Institute for
Bioengineering and Nanotechnology, The University of Queensland, Brisbane QLD 4072, Australia
| | - Michael Song
- Australian Institute for
Bioengineering and Nanotechnology, The University of Queensland, Brisbane QLD 4072, Australia
| | - Peter P. Gray
- Australian Institute for
Bioengineering and Nanotechnology, The University of Queensland, Brisbane QLD 4072, Australia
| | - Trent P. Munro
- Australian Institute for
Bioengineering and Nanotechnology, The University of Queensland, Brisbane QLD 4072, Australia
| | - Michael J. Monteiro
- Australian Institute for
Bioengineering and Nanotechnology, The University of Queensland, Brisbane QLD 4072, Australia
| |
Collapse
|
46
|
Luo GF, Chen WH, Liu Y, Lei Q, Zhuo RX, Zhang XZ. Multifunctional enveloped mesoporous silica nanoparticles for subcellular co-delivery of drug and therapeutic peptide. Sci Rep 2014; 4:6064. [PMID: 25317538 PMCID: PMC5377537 DOI: 10.1038/srep06064] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 07/25/2014] [Indexed: 12/22/2022] Open
Abstract
A multifunctional enveloped nanodevice based on mesoporous silica nanoparticle (MSN) was delicately designed for subcellular co-delivery of drug and therapeutic peptide to tumor cells. Mesoporous silica MCM-41 nanoparticles were used as the core for loading antineoplastic drug topotecan (TPT). The surface of nanoparticles was decorated with mitochondria-targeted therapeutic agent (Tpep) containing triphenylphosphonium (TPP) and antibiotic peptide (KLAKLAK)2 via disulfide linkage, followed by coating with a charge reversal polyanion poly(ethylene glycol)-blocked-2,3-dimethylmaleic anhydride-modified poly(L-lysine) (PEG-PLL(DMA)) via electrostatic interaction. It was found that the outer shielding layer could be removed at acidic tumor microenvironment due to the degradation of DMA blocks and the cellular uptake was significantly enhanced by the formation of cationic nanoparticles. After endocytosis, due to the cleavage of disulfide bonds in the presence of intracellular glutathione (GSH), pharmacological agents (Tpep and TPT) could be released from the nanoparticles and subsequently induce specific damage of tumor cell mitochondria and nucleus respectively with remarkable synergistic antitumor effect.
Collapse
Affiliation(s)
- Guo-Feng Luo
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Yun Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Qi Lei
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Ren-Xi Zhuo
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
47
|
Raad MD, Teunissen EA, Mastrobattista E. Peptide vectors for gene delivery: from single peptides to multifunctional peptide nanocarriers. Nanomedicine (Lond) 2014; 9:2217-32. [DOI: 10.2217/nnm.14.90] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The therapeutic use of nucleic acids relies on the availability of sophisticated delivery systems for targeted and intracellular delivery of these molecules. Such a gene delivery should possess essential characteristics to overcome several extracellular and intracellular barriers. Peptides offer an attractive platform for nonviral gene delivery, as several functional peptide classes exist capable of overcoming these barriers. However, none of these functional peptide classes contain all the essential characteristics required to overcome all of the barriers associated with successful gene delivery. Combining functional peptides into multifunctional peptide vectors will be pivotal for improving peptide-based gene delivery systems. By using combinatorial strategies and high-throughput screening, the identification of multifunctional peptide vectors will accelerate the optimization of peptide-based gene delivery systems.
Collapse
Affiliation(s)
- Markus de Raad
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, University of Utrecht, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Erik A Teunissen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, University of Utrecht, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, University of Utrecht, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
48
|
Park K. The mitotic window of opportunity for plasmid DNA delivery. J Control Release 2014; 179:76. [PMID: 24655433 DOI: 10.1016/j.jconrel.2014.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Kinam Park
- Purdue University Departments of Biomedical Engineering and Pharmaceutics West Lafayette, IN, USA.
| |
Collapse
|
49
|
Remaut K, Symens N, Lucas B, Demeester J, De Smedt SC. Cell division responsive peptides for optimized plasmid DNA delivery: the mitotic window of opportunity? J Control Release 2014; 179:1-9. [PMID: 24462902 DOI: 10.1016/j.jconrel.2014.01.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 01/07/2014] [Accepted: 01/13/2014] [Indexed: 10/25/2022]
Abstract
The delivery of plasmid DNA remains hard to achieve, especially due to the presence of the nuclear membrane barrier. During cell division, however, the nuclear membrane is temporarily disassembled. We evaluated two different strategies to optimize plasmid DNA delivery in dividing cells: 1) phosphorylation responsive peptides that release plasmid DNA preferentially during mitosis and 2) chromatin targeting peptides to anchor plasmid DNA in newly formed nuclei upon cell division. Peptide/DNA particles alone were not efficient in penetrating cells. Upon co-delivery with lipid-based carriers, however, transfection efficiency drastically improved when compared to controls. For the phosphorylation responsive peptides, the presence of the phosphorylation sequence slightly increased transfection efficiency. For the chromatin targeting peptides, however, the chromatin targeting sequence did not seem to be the main reason for the improvement of transfection efficiency when applied in living cells. In conclusion, the pre-condensation of plasmid DNA with peptides improves lipid based delivery, but the nature of the peptides (cell responsive or not) does not seem to be the main reason for the improvement. It seems that the nuclear entry of foreign plasmid DNA is still under tight control, even during the mitotic window of opportunity.
Collapse
Affiliation(s)
- K Remaut
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - N Symens
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - B Lucas
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - J Demeester
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | - S C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium.
| |
Collapse
|
50
|
Yu J, Xie X, Xu X, Zhang L, Zhou X, Yu H, Wu P, Wang T, Che X, Hu Z. Development of dual ligand-targeted polymeric micelles as drug carriers for cancer therapy in vitro and in vivo. J Mater Chem B 2014; 2:2114-2126. [DOI: 10.1039/c3tb21539c] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|