1
|
Rajendran AP, Meenakshi Sundaram DN, Morales LC, Kucharski C, Nasrullah M, Bulut B, Tsisar PM, Maguire AD, Kerr BJ, Uludağ H. Lipid-Modified PEI Derivative-Based Binary/Ternary Polyplex Formulations for the Delivery of pDNA and mRNA in Primary Cells. Biomacromolecules 2025; 26:2733-2749. [PMID: 40148262 DOI: 10.1021/acs.biomac.5c00169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
A previous study has demonstrated the benefit of modification of polyethylenimine (PEI1.2k) by lipids through a p-hydroxyphenylacetic acid (PHPA) linker and polyanion (PA), which is now extended in this report to several primary cells. The formulated binary (lipopolymer/NAs) and ternary (lipopolymer/NAs/PA) complexes displayed no significant toxicity (MTT/hemolysis assay). The pDNA/mRNA complexes with PEI1.2k-PHPA-Lin9 and PEI1.2k-PHPA-Lau5-Ole5 lipopolymers showed gene expression levels higher than those of other lipopolymers. The transfection efficiencies of the ternary polyplexes of these lipopolymers possessed higher gene expression than those of the binary polyplexes. The serum-stable ternary polyplexes of PEI1.2k-PHPA-Lau5-Ole5 maintained high levels of mRNA expression in the lungs along with the spleen after intravenous injection. As in in vitro studies, transgene expression was relatively weak with binary complexes in muscle; however, a 10-fold higher efficiency was obtained with ternary complexes. Overall, our results provide improved gene formulations for the transfections of primary cells in vitro, as well as in in vivo animal models.
Collapse
Affiliation(s)
- Amarnath Praphakar Rajendran
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | | | - Luis Carlos Morales
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Cezary Kucharski
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Mohammad Nasrullah
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
| | - Burcak Bulut
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Pavlo Michailo Tsisar
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| | - Aislinn D Maguire
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Bradley J Kerr
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Department of Anesthesiology and Pain Medicine, University of Alberta, Clinical Sciences Building, 2-150, Edmonton, AB T6G 2G3, Canada
- Department of Pharmacology, University of Alberta, Edmonton, AB T6E 2H7, Canada
| | - Hasan Uludağ
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada
- Department of Biomedical Engineering and Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
2
|
Su WC, Lieu R, Fu Y, Kempen T, Yu Z, Zhang K, Chen T, Fan Y. A platform method for simultaneous quantification of lipid and nucleic acid components in lipid nanoparticles. J Chromatogr A 2025; 1746:465788. [PMID: 39987694 DOI: 10.1016/j.chroma.2025.465788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
Nucleic acid-based medicines have achieved significant advancements in recent years, with lipid nanoparticles (LNPs) being a pivotal platform for their delivery. However, the complexity of LNP presents significant challenges, requiring analytical methods to identify and quantify individual components to guide formulation development and ensure quality and safety. Current approaches often perform nucleic acid and lipid analysis separately and focus on a single type of formulation, highlighting the need for a simple platform method that can be applied to diverse formulations. We present a platform ion-pair reversed-phase HPLC method with UV and charged aerosol detection (CAD) to simultaneously separate and quantify lipid and nucleic acid components in LNPs. The method separated and quantified 12 lipid species and three types of nucleic acids (antisense oligonucleotide, single-guide RNA, and mRNA), covering a broad range of therapeutic cargoes. Notably, this can be achieved for the first time by one HPLC run with one-step facile sample preparation. Specifically, we used a simple buffer containing Triton and heparin to enable the single-step, simultaneous extraction of both nucleic acid and lipid components from LNPs, achieving quantification recoveries of 90-110 %. We further applied this method and addressed process and quality control challenges of LNPs, including the recovery rate of individual LNP components after purification and simultaneous quantification of co-loaded, different nucleic acid species for potential gene editing applications. This new platform method offers a robust and widely applicable tool to assess the quality of lipid-based nucleic acid therapies.
Collapse
Affiliation(s)
- Wan-Chih Su
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Raymond Lieu
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yige Fu
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Trevor Kempen
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Zhixin Yu
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kelly Zhang
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Tao Chen
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Yuchen Fan
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
3
|
Montaño-González PA, Bravo-Lozano LM, Chevance S, Dole F, Rosselgong J, Loyer P, Tranchimand S, Chapel JP, Gauffre F, Schatz C, Bravo-Anaya LM. Interactions between PEI and biological polyanions and the ability of glycosaminoglycans in destabilizing PEI/peGFP-C3 polyplexes for genetic material release. Int J Biol Macromol 2025; 301:140351. [PMID: 39880239 DOI: 10.1016/j.ijbiomac.2025.140351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/24/2024] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
The lack of understanding of polyplexes stability and their dissociation mechanisms, allowing the release of DNA, is currently a major limitation in non-viral gene delivery. One proposed mechanism for DNA-based polyplexes dissociation is based on the electrostatic interactions between polycations and biological polyanions, such as glycosaminoglycans (GAGs). This work aimed at investigating whether GAGs such as heparin, chondroitin sulphate and hyaluronic acid promote the dissociation of PEI/DNA polyplexes. We studied the electrostatic complexation between branched poly(ethyleneimine) (b-PEI25) and polyanions (model DNA and GAGs) through conductivity and ζ-potential measurements. The formation of b-PEI25/polyanion polyplexes through electrostatic interactions was analyzed in depth, providing key insights into charge stoichiometry, morphology, thermodynamics and physicochemical characteristics. The stability of polyplexes was tested in the presence of the different GAGs. Heparin was found to be the only polyanion capable of releasing peGFP-C3 plasmid from polyplexes, complexing stoichiometrically with the free b-PEI25 in excess, before releasing the plasmid. The ability of GAGs to disrupt polyplexes and release DNA was correlated with the thermodynamic characteristics of b-PEI25/polyanions complexation. Our findings indicate that heparin's strong interaction with PEI and its high charge density, compared to other GAGs and polyanions, are pivotal in determining complex stability and promoting DNA release.
Collapse
Affiliation(s)
| | | | - Soizic Chevance
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000 Rennes, France
| | - François Dole
- Centre de Recherche Paul Pascal (CRPP), UMR CNRS 5031, Université de Bordeaux, 33600 Pessac, France
| | - Julien Rosselgong
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000 Rennes, France
| | - Pascal Loyer
- Univ Rennes, Inserm, INRAE, Institut NUMECAN, UMR-A 1341, UMR-S 1317, Plateforme SynNanoVect, F-35000 Rennes, France
| | - Sylvain Tranchimand
- Univ Rennes, École Nationale Supérieure de Chimie de Rennes, CNRS, ISCR, UMR 6226, F-35000 Rennes, France
| | - Jean-Paul Chapel
- Centre de Recherche Paul Pascal (CRPP), UMR CNRS 5031, Université de Bordeaux, 33600 Pessac, France
| | - Fabienne Gauffre
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000 Rennes, France
| | - Christophe Schatz
- Univ Bordeaux, Bordeaux INP, LCPO, CNRS, UMR 5629, F-33000 Pessac, France
| | | |
Collapse
|
4
|
Williams-Fegredo T, Davies L, Knevelman C, Mitrophanous K, Miskin J, Rafiq QA. Degradation of specific glycosaminoglycans improves transfection efficiency and vector production in transient lentiviral vector manufacturing processes. Front Bioeng Biotechnol 2024; 12:1409203. [PMID: 38994127 PMCID: PMC11238175 DOI: 10.3389/fbioe.2024.1409203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/04/2024] [Indexed: 07/13/2024] Open
Abstract
Both cell surface and soluble extracellular glycosaminoglycans have been shown to interfere with the exogenous nucleic acid delivery efficiency of non-viral gene delivery, including lipoplex and polyplex-mediated transfection. Most gene therapy viral vectors used commercially and in clinical trials are currently manufactured using transient transfection-based bioprocesses. The growing demand for viral vector products, coupled with a global shortage in production capability, requires improved transfection technologies and processes to maximise process efficiency and productivity. Soluble extracellular glycosaminoglycans were found to accumulate in the conditioned cell culture medium of suspension adapted HEK293T cell cultures, compromising transfection performance and lentiviral vector production. The enzymatic degradation of specific, chondroitin sulphate-based, glycosaminoglycans with chondroitinase ABC was found to significantly enhance transfection performance. Additionally, we report significant improvements in functional lentiviral vector titre when cultivating cells at higher cell densities than those utilised in a control lentiviral vector bioprocess; an improvement that was further enhanced when cultures were supplemented with chondroitinase ABC prior to transfection. A 71.2% increase in functional lentiviral vector titre was calculated when doubling the cell density prior to transfection compared to the existing process and treatment of the high-density cell cultures with 0.1 U/mL chondroitinase ABC resulted in a further 18.6% increase in titre, presenting a method that can effectively enhance transfection performance.
Collapse
Affiliation(s)
- Thomas Williams-Fegredo
- Oxford Biomedica (UK) Limited, Oxford, United Kingdom
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Lee Davies
- Oxford Biomedica (UK) Limited, Oxford, United Kingdom
| | | | | | - James Miskin
- Oxford Biomedica (UK) Limited, Oxford, United Kingdom
| | - Qasim A. Rafiq
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| |
Collapse
|
5
|
Chandra J, Molugulu N, Annadurai S, Wahab S, Karwasra R, Singh S, Shukla R, Kesharwani P. Hyaluronic acid-functionalized lipoplexes and polyplexes as emerging nanocarriers for receptor-targeted cancer therapy. ENVIRONMENTAL RESEARCH 2023; 233:116506. [PMID: 37369307 DOI: 10.1016/j.envres.2023.116506] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 06/29/2023]
Abstract
Cancer is an intricate disease that develops as a response to a combination of hereditary and environmental risk factors, which then result in a variety of changes to the genome. The cluster of differentiation (CD44) is a type of transmembrane glycoprotein that serves as a potential biomarker for cancer stem cells (CSC) and viable targets for therapeutic intervention in the context of cancer therapy. Hyaluronic acid (HA) is a linear polysaccharide that exhibits a notable affinity for the CD44 receptor. This characteristic renders it a promising candidate for therapeutic interventions aimed at selectively targeting CD44-positive cancer cells. Treating cancer via non-viral vector-based gene delivery has changed the notion of curing illness through the incorporation of therapeutic genes into the organism. The objective of this review is to provide an overview of various hyaluronic acid-modified lipoplexes and polyplexes as potential drug delivery methods for specific forms of cancer by effectively targeting CD44.
Collapse
Affiliation(s)
- Jyoti Chandra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Nagashekhara Molugulu
- School of Pharmacy, Monash University, Bandar Sunway, Jalan Lagoon Selatan, 47500, Malaysia
| | - Sivakumar Annadurai
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Ritu Karwasra
- Central Council for Research in Unani Medicine (CCRUM), Ministry of AYUSH, Government of India, Janakpuri, New Delhi 110058, India
| | - Surender Singh
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP, 226002, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
6
|
Supe S, Upadhya A, Tripathi S, Dighe V, Singh K. Liposome-polyethylenimine complexes for the effective delivery of HuR siRNA in the treatment of diabetic retinopathy. Drug Deliv Transl Res 2023; 13:1675-1698. [PMID: 36630075 DOI: 10.1007/s13346-022-01281-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2022] [Indexed: 01/12/2023]
Abstract
Diabetic retinopathy (DR) is a vision-impairing complication of diabetes, damaging the retinal microcirculatory system. Overexpression of VEGF (vascular endothelial growth factor) is implicated in the pathogenesis of DR. Human antigen R (HuR) is an RNA-binding protein that favorably regulates VEGF protein expression by binding to VEGF-encoding mRNA. Downregulating HuR via RNA interference strategies using small interfering RNAs (siRNAs) may constitute a novel therapeutic method for preventing VEGF protein overexpression in DR. Delivery of siRNAs to the cellular cytoplasm can be facilitated by cationic peptides or polymers and lipids. In this study, a cationic polymer (polyethylenimine (PEI)) and lipid nanoparticles (liposomes) were co-formulated with siRNA to form lipopolyplexes (LPPs) for the delivery of HuR siRNA. LPPs-siRNA were analyzed for size, zeta potential, serum stability, RNase stability, heparin stability, toxicity, and siRNA encapsulation efficiency. Cellular uptake, downregulation of the target HuR (mRNA and protein), and associated VEGF protein were used to demonstrate the biological efficacy of the LPPs-HuR siRNA, in vitro (human ARPE-19 cells), and in vivo (Wistar rats). In vivo efficacy study was performed by injecting LPPs-HuR siRNA formulations into the eye of streptozotocin (STZ)-induced diabetic rats after the development of retinopathy. Our findings demonstrated that high retinal HuR and VEGF levels observed in the eyes of untreated STZ rats were lowered after LPPs-HuR siRNA administration. Our observations indicate that intravitreal treatment with HuR siRNA is a promising option for DR using LPPs as delivery agents.
Collapse
Affiliation(s)
- Shibani Supe
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, Maharashtra, 400056, India
| | - Archana Upadhya
- Humera Khan College of Pharmacy, HK College Campus, Oshiwara, Jogeshwari (West), Mumbai, Maharashtra, 400102, India
| | - Santosh Tripathi
- Bombay Veterinary College, Sindhu Nagar, Parel Village, Parel, Mumbai, Maharashtra, 400012, India
| | - Vikas Dighe
- National Centre for Preclinical Reproductive and Genetic Toxicology, ICMR-National Institute for Research in Reproductive and Child Health, J.M.Street, Parel, Mumbai, Maharashtra, 400012, India.
| | - Kavita Singh
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, Maharashtra, 400056, India.
| |
Collapse
|
7
|
Pyreddy S, Poddar A, Carraro F, Polash SA, Dekiwadia C, Murdoch B, Nasa Z, Reddy TS, Falcaro P, Shukla R. Targeting telomerase utilizing zeolitic imidazole frameworks as non-viral gene delivery agents across different cancer cell types. BIOMATERIALS ADVANCES 2023; 149:213420. [PMID: 37062125 DOI: 10.1016/j.bioadv.2023.213420] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/28/2023] [Accepted: 04/02/2023] [Indexed: 04/18/2023]
Abstract
Telomerase, a ribonucleoprotein coded by the hTERT gene, plays an important role in cellular immortalization and carcinogenesis. hTERT is a suitable target for cancer therapeutics as its activity is highly upregulated in most of cancer cells but absent in normal somatic cells. Here, by employing the two Metal-Organic Frameworks (MOFs), viz. ZIF-C and ZIF-8, based biomineralization we encapsulate Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)/Cas9 plasmid system that targets hTERT gene (CrhTERT) in cancer cells. When comparing the two biocomposites, ZIF-C shows the better loading capacity and cell viability. The loaded plasmid in ZIF-C is highly protected against enzymatic degradation. CrhTERT@ZIF-C is efficiently endocytosed by cancer cells and the subcellular release of CrhTERT leads to telomerase knockdown. The resultant inhibition of hTERT expression decreases cellular proliferation and causing cancer cell death. Furthermore, hTERT knockdown shows a significant reduction in tumour metastasis and alters protein expression. Collectively we show the high potential of ZIF-C-based biocomposites as a promising general tool for gene therapy of different types of cancers.
Collapse
Affiliation(s)
- Suneela Pyreddy
- NanoBiotechnology Research Laboratory, Centre for Advanced Materials & Industrial Chemistry, RMIT University, Melbourne, Victoria 3001, Australia; School of Science, RMIT University, Melbourne, Victoria 3001, Australia
| | - Arpita Poddar
- NanoBiotechnology Research Laboratory, Centre for Advanced Materials & Industrial Chemistry, RMIT University, Melbourne, Victoria 3001, Australia; School of Science, RMIT University, Melbourne, Victoria 3001, Australia; Fiona Elsey Cancer Research Institute, Ballarat, Victoria 3350, Australia
| | - Francesco Carraro
- Institute of Physical and Theoretical Chemistry, Graz University of Technology, Graz 8010, Austria
| | - Shakil Ahmed Polash
- NanoBiotechnology Research Laboratory, Centre for Advanced Materials & Industrial Chemistry, RMIT University, Melbourne, Victoria 3001, Australia; School of Science, RMIT University, Melbourne, Victoria 3001, Australia
| | | | - Billy Murdoch
- School of Science, RMIT University, Melbourne, Victoria 3001, Australia
| | - Zeyad Nasa
- School of Science, RMIT University, Melbourne, Victoria 3001, Australia
| | - T Srinivasa Reddy
- NanoBiotechnology Research Laboratory, Centre for Advanced Materials & Industrial Chemistry, RMIT University, Melbourne, Victoria 3001, Australia; School of Science, RMIT University, Melbourne, Victoria 3001, Australia
| | - Paolo Falcaro
- Institute of Physical and Theoretical Chemistry, Graz University of Technology, Graz 8010, Austria.
| | - Ravi Shukla
- NanoBiotechnology Research Laboratory, Centre for Advanced Materials & Industrial Chemistry, RMIT University, Melbourne, Victoria 3001, Australia; School of Science, RMIT University, Melbourne, Victoria 3001, Australia.
| |
Collapse
|
8
|
Yang W, Miyazaki T, Nakagawa Y, Boonstra E, Masuda K, Nakashima Y, Chen P, Mixich L, Barthelmes K, Matsumoto A, Mi P, Uchida S, Cabral H. Block catiomers with flanking hydrolyzable tyrosinate groups enhance in vivo mRNA delivery via π-π stacking-assisted micellar assembly. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2023; 24:2170164. [PMID: 36950277 PMCID: PMC10026751 DOI: 10.1080/14686996.2023.2170164] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 06/18/2023]
Abstract
Messenger RNA (mRNA) therapeutics have recently demonstrated high clinical potential with the accelerated approval of SARS-CoV-2 vaccines. To fulfill the promise of unprecedented mRNA-based treatments, the development of safe and efficient carriers is still necessary to achieve effective delivery of mRNA. Herein, we prepared mRNA-loaded nanocarriers for enhanced in vivo delivery using biocompatible block copolymers having functional amino acid moieties for tunable interaction with mRNA. The block copolymers were based on flexible poly(ethylene glycol)-poly(glycerol) (PEG-PG) modified with glycine (Gly), leucine (Leu) or tyrosine (Tyr) via ester bonds to generate block catiomers. Moreover, the amino acids can be gradually detached from the block copolymers after ester bond hydrolyzation, avoiding cytotoxic effects. When mixed with mRNA, the block catiomers formed narrowly distributed polymeric micelles with high stability and enhanced delivery efficiency. Particularly, the micelles based on tyrosine-modified PEG-PG (PEG-PGTyr), which formed a polyion complex (PIC) and π-π stacking with mRNA, displayed excellent stability against polyanions and promoted mRNA integrity in serum. PEG-PGTyr-based micelles also increased the cellular uptake and the endosomal escape, promoting high protein expression both in vitro and in vivo. Furthermore, the PEG-PGTyr-based micelles significantly extended the half-life of the loaded mRNA after intravenous injection. Our results highlight the potential of PEG-PGTyr-based micelles as safe and effective carriers for mRNA, expediting the rational design of polymeric materials for enhanced mRNA delivery.
Collapse
Affiliation(s)
- Wenqian Yang
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Takuya Miyazaki
- Kanagawa Institute of Industrial Science and Technology, Ebina, Japan
| | - Yasuhiro Nakagawa
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Eger Boonstra
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Keita Masuda
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Yuki Nakashima
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Pengwen Chen
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Lucas Mixich
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Kevin Barthelmes
- Kanagawa Institute of Industrial Science and Technology, Ebina, Japan
| | - Akira Matsumoto
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Peng Mi
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Satoshi Uchida
- Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
9
|
Zhang YQ, Guo RR, Chen YH, Li TC, Du WZ, Xiang RW, Guan JB, Li YP, Huang YY, Yu ZQ, Cai Y, Zhang P, Ling GX. Ionizable drug delivery systems for efficient and selective gene therapy. Mil Med Res 2023; 10:9. [PMID: 36843103 PMCID: PMC9968649 DOI: 10.1186/s40779-023-00445-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 02/01/2023] [Indexed: 02/28/2023] Open
Abstract
Gene therapy has shown great potential to treat various diseases by repairing the abnormal gene function. However, a great challenge in bringing the nucleic acid formulations to the market is the safe and effective delivery to the specific tissues and cells. To be excited, the development of ionizable drug delivery systems (IDDSs) has promoted a great breakthrough as evidenced by the approval of the BNT162b2 vaccine for prevention of coronavirus disease 2019 (COVID-19) in 2021. Compared with conventional cationic gene vectors, IDDSs can decrease the toxicity of carriers to cell membranes, and increase cellular uptake and endosomal escape of nucleic acids by their unique pH-responsive structures. Despite the progress, there remain necessary requirements for designing more efficient IDDSs for precise gene therapy. Herein, we systematically classify the IDDSs and summarize the characteristics and advantages of IDDSs in order to explore the underlying design mechanisms. The delivery mechanisms and therapeutic applications of IDDSs are comprehensively reviewed for the delivery of pDNA and four kinds of RNA. In particular, organ selecting considerations and high-throughput screening are highlighted to explore efficiently multifunctional ionizable nanomaterials with superior gene delivery capacity. We anticipate providing references for researchers to rationally design more efficient and accurate targeted gene delivery systems in the future, and indicate ideas for developing next generation gene vectors.
Collapse
Affiliation(s)
- Yu-Qi Zhang
- Faculty of Medical Device, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Ran-Ran Guo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Yong-Hu Chen
- School of Pharmacy, Yanbian University, Yanji, 133002, Jilin, China
| | - Tian-Cheng Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Wen-Zhen Du
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Rong-Wu Xiang
- Faculty of Medical Device, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Ji-Bin Guan
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yu-Peng Li
- Masonic Cancer Center and Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yuan-Yu Huang
- Advanced Research Institute of Multidisciplinary Science; School of Life Science; School of Medical Technology; Key Laboratory of Molecular Medicine and Biotherapy; Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhi-Qiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, Guangdong, China
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China.
| | - Gui-Xia Ling
- Faculty of Medical Device, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China.
| |
Collapse
|
10
|
Karamali F, Behtaj S, Babaei-Abraki S, Hadady H, Atefi A, Savoj S, Soroushzadeh S, Najafian S, Nasr Esfahani MH, Klassen H. Potential therapeutic strategies for photoreceptor degeneration: the path to restore vision. J Transl Med 2022; 20:572. [PMID: 36476500 PMCID: PMC9727916 DOI: 10.1186/s12967-022-03738-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/29/2022] [Indexed: 12/12/2022] Open
Abstract
Photoreceptors (PRs), as the most abundant and light-sensing cells of the neuroretina, are responsible for converting light into electrical signals that can be interpreted by the brain. PR degeneration, including morphological and functional impairment of these cells, causes significant diminution of the retina's ability to detect light, with consequent loss of vision. Recent findings in ocular regenerative medicine have opened promising avenues to apply neuroprotective therapy, gene therapy, cell replacement therapy, and visual prostheses to the challenge of restoring vision. However, successful visual restoration in the clinical setting requires application of these therapeutic approaches at the appropriate stage of the retinal degeneration. In this review, firstly, we discuss the mechanisms of PR degeneration by focusing on the molecular mechanisms underlying cell death. Subsequently, innovations, recent developments, and promising treatments based on the stage of disorder progression are further explored. Then, the challenges to be addressed before implementation of these therapies in clinical practice are considered. Finally, potential solutions to overcome the current limitations of this growing research area are suggested. Overall, the majority of current treatment modalities are still at an early stage of development and require extensive additional studies, both pre-clinical and clinical, before full restoration of visual function in PR degeneration diseases can be realized.
Collapse
Affiliation(s)
- Fereshteh Karamali
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Sanaz Behtaj
- grid.1022.10000 0004 0437 5432Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Queensland, Australia ,grid.1022.10000 0004 0437 5432Menzies Health Institute Queensland, Griffith University, Southport, QLD 4222 Australia
| | - Shahnaz Babaei-Abraki
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Hanieh Hadady
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Atefeh Atefi
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Soraya Savoj
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Sareh Soroushzadeh
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Samaneh Najafian
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr Esfahani
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Henry Klassen
- grid.266093.80000 0001 0668 7243Gavin Herbert Eye Institute, Irvine, CA USA
| |
Collapse
|
11
|
Chien Y, Hsiao YJ, Chou SJ, Lin TY, Yarmishyn AA, Lai WY, Lee MS, Lin YY, Lin TW, Hwang DK, Lin TC, Chiou SH, Chen SJ, Yang YP. Nanoparticles-mediated CRISPR-Cas9 gene therapy in inherited retinal diseases: applications, challenges, and emerging opportunities. J Nanobiotechnology 2022; 20:511. [DOI: 10.1186/s12951-022-01717-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/23/2022] [Indexed: 12/04/2022] Open
Abstract
AbstractInherited Retinal Diseases (IRDs) are considered one of the leading causes of blindness worldwide. However, the majority of them still lack a safe and effective treatment due to their complexity and genetic heterogeneity. Recently, gene therapy is gaining importance as an efficient strategy to address IRDs which were previously considered incurable. The development of the clustered regularly-interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) system has strongly empowered the field of gene therapy. However, successful gene modifications rely on the efficient delivery of CRISPR-Cas9 components into the complex three-dimensional (3D) architecture of the human retinal tissue. Intriguing findings in the field of nanoparticles (NPs) meet all the criteria required for CRISPR-Cas9 delivery and have made a great contribution toward its therapeutic applications. In addition, exploiting induced pluripotent stem cell (iPSC) technology and in vitro 3D retinal organoids paved the way for prospective clinical trials of the CRISPR-Cas9 system in treating IRDs. This review highlights important advances in NP-based gene therapy, the CRISPR-Cas9 system, and iPSC-derived retinal organoids with a focus on IRDs. Collectively, these studies establish a multidisciplinary approach by integrating nanomedicine and stem cell technologies and demonstrate the utility of retina organoids in developing effective therapies for IRDs.
Collapse
|
12
|
Peptide-Based Nanoparticles for αvβ3 Integrin-Targeted DNA Delivery to Cancer and Uterine Leiomyoma Cells. Molecules 2022; 27:molecules27238363. [PMID: 36500454 PMCID: PMC9741203 DOI: 10.3390/molecules27238363] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Uterine leiomyoma is the most common benign tumor of the reproductive system. Current therapeutic options do not simultaneously meet the requirements of long-term efficiency and fertility preservation. Suicide gene delivery can be proposed as a novel approach to uterine leiomyoma therapy. Non-viral vehicles are an attractive approach to DNA delivery for gene therapy of both malignant and benign tumors. Peptide-based vectors are among the most promising candidates for the development of artificial viruses, being able to efficiently cross barriers of DNA transport to cells. Here we described nanoparticles composed of cysteine-crosslinked polymer and histidine-arginine-rich peptide modified with iRGD moiety and characterized them as vehicles for plasmid DNA delivery to pancreatic cancer PANC-1 cells and the uterine leiomyoma cell model. Several variants of nanoparticles were formulated with different targeting ligand content. The physicochemical properties that were studied included DNA binding and protection, interaction with polyanions and reducing agents, size, structure and zeta-potential of the peptide-based nanoparticles. Cytotoxicity, cell uptake and gene transfection efficiency were assessed in PANC-1 cells with GFP and LacZ-encoding plasmids. The specificity of gene transfection via αvβ3 integrin binding was proved in competitive transfection. The therapeutic potential was evaluated in a uterine leiomyoma cell model using the suicide gene therapy approach. The optimal formulation was found to be at the polyplex with the highest iRGD moiety content being able to transfect cells more efficiently than control PEI. Suicide gene therapy using the best formulation resulted in a significant decrease of uterine leiomyoma cells after ganciclovir treatment. It can be concluded that the application of iRGD-modified peptide-based nanoparticles has a high potential for cellular delivery of DNA therapeutics in favor of uterine leiomyoma gene therapy.
Collapse
|
13
|
Yamasaki Y, Kumekawa D, Yamauchi S, Omuro H. Re-examination of Peptide-Sequence-Dependent Gene Expression of Cysteine-Installed Pegylated Oligolysine/DNA Complexes. ACS OMEGA 2022; 7:15478-15487. [PMID: 35571853 PMCID: PMC9096824 DOI: 10.1021/acsomega.2c00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/08/2022] [Indexed: 06/15/2023]
Abstract
We previously synthesized cysteine-installed C-terminally PEGylated oligolysines with 20 amino acid residues to form cross-linked polymeric micelles (PMs) with luciferase-coding plasmid DNA as a candidate for artificial gene vectors. Luciferase gene expression in HeLa cells mediated by PEG-CK18C, PEG-CK9CK9, and PEG-K9CK9C was reported to be 35-, 5.4-, and 1.3-fold higher than that mediated by cysteine-uninstalled PEGylated oligolysine PEG-K20, respectively. However, after the publication, the survival rate of HeLa cells used in the previous study was found to be lower than usual when subcutaneously implanted into mice to create a xenograft model. In this study, to re-examine the peptide sequence-dependent gene expression, gene expression efficacy mediated by PEG-peptide PMs was compared with the PM cellular uptake results using newly obtained HeLa cell lines and the additional cell lines Huh-7, PANC-1, and BxPC3. As a result, PEG-K9CK9C PMs mediated the maximum gene expression in all cell lines, and the corresponding cellular uptake was also obtained. Therefore, we concluded that our previous results were erroneously obtained due to normality-depleted HeLa cells. A comparison of physicochemical characterizations, gene expression efficacy, and cellular uptake of PEG-peptide PMs is discussed in detail.
Collapse
|
14
|
Srikantha N, Teijeiro-Gonzalez Y, Simpson A, Elsaid N, Somavarapu S, Suhling K, Jackson TL. Determining vitreous viscosity using fluorescence recovery after photobleaching. PLoS One 2022; 17:e0261925. [PMID: 35143514 PMCID: PMC8830689 DOI: 10.1371/journal.pone.0261925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 12/14/2021] [Indexed: 01/22/2023] Open
Abstract
PURPOSE Vitreous humor is a complex biofluid whose composition determines its structure and function. Vitreous viscosity will affect the delivery, distribution, and half-life of intraocular drugs, and key physiological molecules. The central pig vitreous is thought to closely match human vitreous viscosity. Diffusion is inversely related to viscosity, and diffusion is of fundamental importance for all biochemical reactions. Fluorescence Recovery After Photobleaching (FRAP) may provide a novel means of measuring intravitreal diffusion that could be applied to drugs and physiological macromolecules. It would also provide information about vitreous viscosity, which is relevant to drug elimination, and delivery. METHODS Vitreous viscosity and intravitreal macromolecular diffusion of fluorescently labelled macromolecules were investigated in porcine eyes using fluorescence recovery after photobleaching (FRAP). Fluorescein isothiocyanate conjugated (FITC) dextrans and ficolls of varying molecular weights (MWs), and FITC-bovine serum albumin (BSA) were employed using FRAP bleach areas of different diameters. RESULTS The mean (±standard deviation) viscosity of porcine vitreous using dextran, ficoll and BSA were 3.54 ± 1.40, 2.86 ± 1.13 and 4.54 ± 0.13 cP respectively, with an average of 3.65 ± 0.60 cP. CONCLUSIONS FRAP is a feasible and practical optical method to quantify the diffusion of macromolecules through vitreous.
Collapse
Affiliation(s)
- Nishanthan Srikantha
- School of Medicine, King’s College London, London, United Kingdom
- Department of Ophthalmology, King’s College Hospital, London, United Kingdom
- * E-mail:
| | | | - Andrew Simpson
- School of Medicine, King’s College London, London, United Kingdom
- Department of Ophthalmology, King’s College Hospital, London, United Kingdom
| | - Naba Elsaid
- Anglia Ruskin University, Bishop Hall Lane, Chelmsford, United Kingdom
| | - Satyanarayana Somavarapu
- Department of Pharmaceutics, University College London School of Pharmacy, London, United Kingdom
| | - Klaus Suhling
- Department of Physics, King’s College London, Strand, London, United Kingdom
| | - Timothy L. Jackson
- School of Medicine, King’s College London, London, United Kingdom
- Department of Ophthalmology, King’s College Hospital, London, United Kingdom
| |
Collapse
|
15
|
Egorova A, Shtykalova S, Maretina M, Selutin A, Shved N, Deviatkin D, Selkov S, Baranov V, Kiselev A. Polycondensed Peptide Carriers Modified with Cyclic RGD Ligand for Targeted Suicide Gene Delivery to Uterine Fibroid Cells. Int J Mol Sci 2022; 23:1164. [PMID: 35163086 PMCID: PMC8835468 DOI: 10.3390/ijms23031164] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/04/2023] Open
Abstract
Suicide gene therapy was suggested as a possible strategy for the treatment of uterine fibroids (UFs), which are the most common benign tumors inwomen of reproductive age. For successful suicide gene therapy, DNAtherapeutics should be specifically delivered to UF cells. Peptide carriers are promising non-viral gene delivery systems that can be easily modified with ligands and other biomolecules to overcome DNA transfer barriers. Here we designed polycondensed peptide carriers modified with a cyclic RGD moiety for targeted DNA delivery to UF cells. Molecular weights of the resultant polymers were determined, and inclusion of the ligand was confirmed by MALDI-TOF. The physicochemical properties of the polyplexes, as well as cellular DNA transport, toxicity, and transfection efficiency were studied, and the specificity of αvβ3 integrin-expressing cell transfection was proved. The modification with the ligand resulted in a three-fold increase of transfection efficiency. Modeling of the suicide gene therapy by transferring the HSV-TK suicide gene to primary cells obtained from myomatous nodes of uterine leiomyoma patients was carried out. We observed up to a 2.3-fold decrease in proliferative activity after ganciclovir treatment of the transfected cells. Pro- and anti-apoptotic gene expression analysis confirmed our findings that the developed polyplexes stimulate UF cell death in a suicide-specific manner.
Collapse
Affiliation(s)
- Anna Egorova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| | - Sofia Shtykalova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| | - Marianna Maretina
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| | - Alexander Selutin
- Department of Immunology and Intercellular Interactions, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.S.); (S.S.)
| | - Natalia Shved
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| | - Dmitriy Deviatkin
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| | - Sergey Selkov
- Department of Immunology and Intercellular Interactions, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.S.); (S.S.)
| | - Vladislav Baranov
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| | - Anton Kiselev
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| |
Collapse
|
16
|
Arora S, Sharma D, Layek B, Singh J. A Review of Brain-Targeted Nonviral Gene-Based Therapies for the Treatment of Alzheimer's Disease. Mol Pharm 2021; 18:4237-4255. [PMID: 34705472 DOI: 10.1021/acs.molpharmaceut.1c00611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diseases of the central nervous system (CNS) are difficult to treat owing to the complexity of the brain and the presence of a natural blood-brain-barrier (BBB). Alzheimer's disease (AD) is one of the major progressive and currently incurable neurodegenerative disorders of the CNS, which accounts for 60-80% of cases of dementia. The pathophysiology of AD involves the accumulation of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain. Additionally, synaptic loss and imbalance of neuronal signaling molecules are characterized as important markers of AD. Existing treatments of AD help in the management of its symptoms and aim toward the maintenance of cognitive functions, behavior, and attenuation of gradual memory loss. Over the past decade, nonviral gene therapy has attracted increasing interest due to its various advantages over its viral counterparts. Moreover, advancements in nonviral gene technology have led to their increasing contributions in clinical trials. However, brain-targeted nonviral gene delivery vectors come across various extracellular and intracellular barriers, limiting their ability to transfer the therapeutic gene into the target cells. Chief barriers to nonviral gene therapy have been discussed briefly in this review. We have also highlighted the rapid advancement of several nonviral gene therapies for AD, which are broadly categorized into physical and chemical methods. These methods aim to modulate Aβ, beta-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), apolipoprotein E, or neurotrophic factors' expression in the CNS. Overall, this review discusses challenges and recent advancements of nonviral gene therapy for AD.
Collapse
Affiliation(s)
- Sanjay Arora
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Divya Sharma
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Buddhadev Layek
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| |
Collapse
|
17
|
Diltemiz SE, Tavafoghi PhD M, Roberto de Barros N, Kanada M, Heinamaki J, Contag C, Seidlits S, Ashammakhi N. USE OF ARTIFICIAL CELLS AS DRUG CARRIERS. MATERIALS CHEMISTRY FRONTIERS 2021; 5:6672-6692. [PMID: 38344270 PMCID: PMC10857888 DOI: 10.1039/d1qm00717c] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Cells are the fundamental functional units of biological systems and mimicking their size, function and complexity is a primary goal in the development of new therapeutic strategies. Recent advances in chemistry, synthetic biology and material science have enabled the development of cell membrane-based drug delivery systems (DDSs), often referred to as "artificial cells" or protocells. Artificial cells can be made by removing functions from natural systems in a top-down manner, or assembly from synthetic, organic or inorganic materials, through a bottom-up approach where simple units are integrated to form more complex structures. This review covers the latest advances in the development of artificial cells as DDSs, highlighting how their designs have been inspired by natural cells or cell membranes. Advancement of artificial cell technologies has led to a set of drug carriers with effective and controlled release of a variety of therapeutics for a range of diseases, and with increasing complexity they will have a greater impact on therapeutic designs.
Collapse
Affiliation(s)
- Sibel Emir Diltemiz
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
- Department of Chemistry, Eskisehir Technical University, Eskisehir, Turkey
| | - Maryam Tavafoghi PhD
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
| | - Natan Roberto de Barros
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
- Department of Bioprocess and Biotechnology Engineering, São Paulo State University (Unesp), School of Pharmaceutical Sciences, Araraquara, São Paulo, Brazil
| | - Masamitsu Kanada
- Institute for Quantitative Health Science and Engineering (IQ), Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Jyrki Heinamaki
- Institute of Pharmacy, Faculty of Medicine, University of Tartu, Nooruse Str. 1, EE-50411 Tartu, Estonia
| | - Christopher Contag
- Institute for Quantitative Health Science and Engineering (IQ) and Departments of Biomedical Engineering (BME), and Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Stephanie Seidlits
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
| | - Nureddin Ashammakhi
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
18
|
Sharma D, Arora S, Singh J, Layek B. A review of the tortuous path of nonviral gene delivery and recent progress. Int J Biol Macromol 2021; 183:2055-2073. [PMID: 34087309 PMCID: PMC8266766 DOI: 10.1016/j.ijbiomac.2021.05.192] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/24/2021] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Gene therapy encompasses the transfer of exogenous genetic materials into the patient's target cells to treat or prevent diseases. Nevertheless, the transfer of genetic material into desired cells is challenging and often requires specialized tools or delivery systems. For the past 40 years, scientists are mainly pursuing various viruses as gene delivery vectors, and the overall progress has been slow and far from the expectation. As an alternative, nonviral vectors have gained substantial attention due to their several advantages, including superior safety profile, enhanced payload capacity, and stealth abilities. Since nonviral vectors encounter multiple extra- and intra-cellular barriers limiting the transfer of genetic payload into the target cell nucleus, we have discussed these barriers in detail for this review. A direct approach, utilizing physical methods like electroporation, sonoporation, gene gun, eliminate the requirement for a specific carrier for gene delivery. In contrast, chemical methods of gene transfer exploit natural or synthetic compounds as carriers to increase cellular targeting and gene therapy effectiveness. We have also emphasized the recent advancements aimed at enhancing the current nonviral approaches. Therefore, in this review, we have focused on discussing the current evolving state of nonviral gene delivery systems and their future perspectives.
Collapse
Affiliation(s)
- Divya Sharma
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Sanjay Arora
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Buddhadev Layek
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA.
| |
Collapse
|
19
|
Yoshinaga N, Naito M, Tachihara Y, Boonstra E, Osada K, Cabral H, Uchida S. PEGylation of mRNA by Hybridization of Complementary PEG-RNA Oligonucleotides Stabilizes mRNA without Using Cationic Materials. Pharmaceutics 2021; 13:800. [PMID: 34071840 PMCID: PMC8227728 DOI: 10.3390/pharmaceutics13060800] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/19/2022] Open
Abstract
Messenger RNA (mRNA) delivery strategies are required to protect biologically fragile mRNA from ribonuclease (RNase) attacks to achieve efficient therapeutic protein expression. To tackle this issue, most mRNA delivery systems have used cationic components, which form electrostatically driven complexes with mRNA and shield encapsulated mRNA strands. However, cationic materials interact with anionic biomacromolecules in physiological environments, which leads to unspecific reactions and toxicities. To circumvent this issue of cation-based approaches, herein, we propose a cation-free delivery strategy by hybridization of PEGylated RNA oligonucleotides with mRNA. The PEG strands on the mRNA sterically and electrostatically shielded the mRNA, improving mRNA nuclease stability 15-fold after serum incubation compared with unhybridized mRNA. Eventually, the PEGylated mRNA induced nearly 20-fold higher efficiency of reporter protein expression than unhybridized mRNA in cultured cells. This study provides a platform to establish a safe and efficient cation-free mRNA delivery system.
Collapse
Grants
- 20H04524 the Ministry of Education, Culture, Sports, Science and Technology, Japan (MEXT)
- 18K03529 the Ministry of Education, Culture, Sports, Science and Technology, Japan (MEXT)
- JP18K19901 the Ministry of Education, Culture, Sports, Science and Technology, Japan (MEXT)
- JP18ae0201009 Japan Agency for Medical Research and Development
Collapse
Affiliation(s)
- Naoto Yoshinaga
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; (N.Y.); (Y.T.); (E.B.)
- RIKEN Center for Sustainable Resource Science, Wako 351-0198, Saitama, Japan
| | - Mitsuru Naito
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan;
| | - Yoshihiro Tachihara
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; (N.Y.); (Y.T.); (E.B.)
| | - Eger Boonstra
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; (N.Y.); (Y.T.); (E.B.)
| | - Kensuke Osada
- National Institute of Radiological Science, 4-9-1 Anagawa, Inage-ku, Chiba-shi 236-8555, Chiba, Japan;
| | - Horacio Cabral
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; (N.Y.); (Y.T.); (E.B.)
| | - Satoshi Uchida
- Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; (N.Y.); (Y.T.); (E.B.)
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Inamori Memorial Building, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| |
Collapse
|
20
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 194] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
21
|
Zeng K, Ma L, Yang W, Lei S, Wang M, You Y, Zhao Y, Ge X. Biodegradable nano-organosilica gene carrier for high-efficiency gene transfection. J Mater Chem B 2021; 8:2483-2494. [PMID: 32110782 DOI: 10.1039/d0tb00108b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Finding and exploiting safe and high-efficiency gene carriers have always been critical tasks for gene therapy. In this work, novel GSH-triggered degradable organosilica nanoparticles grafted with guanidinated-fluorinated α-polylysine (o-SiNP-GF) are prepared to be studied as gene carriers. The organosilica matrix of o-SiNP-GF is synthesized through the hydrolysis and condensation of 1,2-bis(triethoxysilyl)ethane (BTSE) and bis[3-(triethoxysilyl)propyl]tetrasulfide (BTSPTS). The o-SiNP-GF nanoparticles have a size of about 20 nm. They possess a positive zeta potential of 42 mV in PBS (pH 7.4) and can be disintegrated in the presence of GSH. The cytotoxicity and DNA-binding ability of o-SiNP-GF, as well as in vitro gene transfection performance of DNA/o-SiNP-GF complexes, have been investigated using enhanced green fluorescent protein plasmid (pEGFP) as the DNA model. MTT assay shows that the cytotoxicity of o-SiNP-GF is very low even at a concentration up to 800 μg mL-1. The o-SiNP-GF nanoparticles can effectively bind to pEGFP through a complex coacervation method. The in vitro transfection efficiency of pEGFP/o-SiNP-GF complexes in 293T cells is up to 94.7% at the N/P ratio of 10, much higher than that of pEGFP/PEI complexes. Luciferase gene and fibroblast growth factor (FGF2) gene are also used as the DNA models to study the in vivo gene transfection performance of the o-SiNP-GF carrier by bioluminescence imaging and the evaluation of the healing rate of a mouse wound, respectively. Compared with naked DNA and DNA/PEI complexes, DNA/o-SiNP-GF complexes show much higher in vivo transfection efficiency. This work not only provides a way to prepare novel GSH-triggered degradable organosilica nanoparticles of size less than 50 nm, but also proves that the modification of guanidinated-fluorinated α-polylysine is an effective method to improve the efficiency of gene carriers.
Collapse
Affiliation(s)
- Kun Zeng
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Li Ma
- Department of Plastic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Wenxiu Yang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Shan Lei
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Mozhen Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Yezi You
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Yu Zhao
- Department of Plastic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Xuewu Ge
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China.
| |
Collapse
|
22
|
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: 110] [Impact Index Per Article: 27.5] [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
|
23
|
Pérez-Alfonso D, López-López M, López-Cornejo P, Romero-Azogil L, Benito E, García-Martín MDG, García-Calderón CB, Rosado IV, Balestra FR, Huertas P, García-Calderón M, Moyá ML. Properties of polyplexes formed between a cationic polymer derived from l-arabinitol and nucleic acids. NEW J CHEM 2021. [DOI: 10.1039/d1nj00606a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Polyplexes formed between a cationic polymer, PUArab, and both linear and plasmid DNA were studied. The transfection efficiency of PURarab/pDNA was investigated.
Collapse
|
24
|
Wang TX, Li XX, Chen L, Li L, Janaswamy S. Carriers Based on Zein-Dextran Sulfate Sodium Binary Complex for the Sustained Delivery of Quercetin. Front Chem 2020; 8:662. [PMID: 33195002 PMCID: PMC7555997 DOI: 10.3389/fchem.2020.00662] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/26/2020] [Indexed: 12/19/2022] Open
Abstract
Herein, a self-assembly formulation of Zein and dextran sulfate sodium (DSS) binary complex has been developed for the quercetin (Que) delivery. The prepared particles display a smooth sphere in the range of 180 ~ 250 nm. The addition of DSS shields the Trp residues of Zein that were located on the hydrophilic exterior and in-turn reduces the surface hydrophobicity of the nanoparticles. The presence of DSS, indeed, increases the encapsulation efficiency of Que from the initial 45.9 in the Zein to 72.6% in the Zein/DSS binary complex. A significant reduction of Que diffusion in the simulated intestinal conditions has been observed with the addition of DSS on the nanoparticles, which also improves Que bioavailability. The release mechanism of Que-loaded Zein/DSS composites is in accordance with the Higuchi model (Q = 0.0913t0.5+0.1652, R2 = 0.953). Overall, nanoparticles based on Zein-DSS complexes stand out as an attractive carrier system of quercetin and the outcome could be extended to several bioactive compounds.
Collapse
Affiliation(s)
- Tian-Xing Wang
- Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Xiao-Xi Li
- Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Ling Chen
- Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Lin Li
- Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Srinivas Janaswamy
- Department of Dairy and Food Science, South Dakota State University, Brookings, SD, United States
| |
Collapse
|
25
|
Osada K. Structural Polymorphism of Single pDNA Condensates Elicited by Cationic Block Polyelectrolytes. Polymers (Basel) 2020; 12:polym12071603. [PMID: 32707655 PMCID: PMC7408586 DOI: 10.3390/polym12071603] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
DNA folding is a core phenomenon in genome packaging within a nucleus. Such a phenomenon is induced by polyelectrolyte complexation between anionic DNA and cationic proteins of histones. In this regard, complexes formed between DNA and cationic polyelectrolytes have been investigated as models to gain insight into genome packaging. Upon complexation, DNA undergoes folding to reduce its occupied volume, which often results in multi-complex associated aggregates. However, when cationic copolymers comprising a polycation block and a neutral hydrophilic polymer block are used instead, DNA undergoes folding as a single molecule within a spontaneously formed polyplex micelle (PM), thereby allowing the observation of the higher-order structures that DNA forms. The DNA complex forms polymorphic structures, including globular, rod-shaped, and ring-shaped (toroidal) structures. This review focuses on the polymorphism of DNA, particularly, to elucidate when, how, and why DNA organizes into these structures with cationic copolymers. The interactions between DNA and the copolymers, and the specific nature of DNA in rigidity; i.e., rigid but foldable, play significant roles in the observed polymorphism. Moreover, PMs serve as potential gene vectors for systemic application. The significance of the controlled DNA folding for such an application is addressed briefly in the last part.
Collapse
Affiliation(s)
- Kensuke Osada
- Quantum Medical Science Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
| |
Collapse
|
26
|
Ramsay E, Raviña M, Sarkhel S, Hehir S, Cameron NR, Ilmarinen T, Skottman H, Kjems J, Urtti A, Ruponen M, Subrizi A. Avoiding the Pitfalls of siRNA Delivery to the Retinal Pigment Epithelium with Physiologically Relevant Cell Models. Pharmaceutics 2020; 12:pharmaceutics12070667. [PMID: 32708811 PMCID: PMC7407886 DOI: 10.3390/pharmaceutics12070667] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/03/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022] Open
Abstract
Inflammation is involved in the pathogenesis of several age-related ocular diseases, such as macular degeneration (AMD), diabetic retinopathy, and glaucoma. The delivery of anti-inflammatory siRNA to the retinal pigment epithelium (RPE) may become a promising therapeutic option for the treatment of inflammation, if the efficient delivery of siRNA to target cells is accomplished. Unfortunately, so far, the siRNA delivery system selection performed in dividing RPE cells in vitro has been a poor predictor of the in vivo efficacy. Our study evaluates the silencing efficiency of polyplexes, lipoplexes, and lipidoid-siRNA complexes in dividing RPE cells as well as in physiologically relevant RPE cell models. We find that RPE cell differentiation alters their endocytic activity and causes a decrease in the uptake of siRNA complexes. In addition, we determine that melanosomal sequestration is another significant and previously unexplored barrier to gene silencing in pigmented cells. In summary, this study highlights the importance of choosing a physiologically relevant RPE cell model for the selection of siRNA delivery systems. Such cell models are expected to enable the identification of carriers with a high probability of success in vivo, and thus propel the development of siRNA therapeutics for ocular disease.
Collapse
Affiliation(s)
- Eva Ramsay
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; (E.R.); (M.R.); (S.S.); (A.U.)
| | - Manuela Raviña
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; (E.R.); (M.R.); (S.S.); (A.U.)
| | - Sanjay Sarkhel
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; (E.R.); (M.R.); (S.S.); (A.U.)
| | - Sarah Hehir
- Department of Life Sciences, Institute of Technology Sligo, F91 YW50 Sligo, Ireland;
| | - Neil R. Cameron
- School of Engineering, University of Warwick, Coventry CV4 7AL, UK;
- Department of Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Tanja Ilmarinen
- Faculty of Medicine and Health Technology, BioMediTech, Tampere University, 33014 Tampere, Finland; (T.I.); (H.S.)
| | - Heli Skottman
- Faculty of Medicine and Health Technology, BioMediTech, Tampere University, 33014 Tampere, Finland; (T.I.); (H.S.)
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, 8000 Aarhus C, Denmark;
| | - Arto Urtti
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; (E.R.); (M.R.); (S.S.); (A.U.)
- Laboratory of Biohybrid Technologies, Institute of Chemistry, St. Petersburg State University, 198504 Peterhoff, Russia
- School of Pharmacy, University of Eastern Finland, 70210 Kuopio, Finland;
| | - Marika Ruponen
- School of Pharmacy, University of Eastern Finland, 70210 Kuopio, Finland;
| | - Astrid Subrizi
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, 8000 Aarhus C, Denmark;
- School of Pharmacy, University of Eastern Finland, 70210 Kuopio, Finland;
- Correspondence: ; Tel.: +358-40-016-3407
| |
Collapse
|
27
|
Photosensitive Poly-l-lysine/Heparin Interpolyelectrolyte Complexes for Delivery of Genetic Drugs. Polymers (Basel) 2020; 12:polym12051077. [PMID: 32397208 PMCID: PMC7285230 DOI: 10.3390/polym12051077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Photo-triggered release of biopharmaceutical drugs inside the cells is a challenging direction of modern science, which requires obtaining new polymeric systems. The interpolyelectrolyte complexes (IPECs) of poly-l-lysine with heparin capable of encapsulation of genetic constructions—such as model oligonucleotide, siRNA, and pDNA—were obtained. Poly-l-lysine to heparin ratios were optimized to provide the appropriate release kinetics of genetic material from the polyplex. In order to impart the obtained IPEC with photosensitive properties, the linker was synthesized as based on 4-brommethyl-3-nitrobenzoic acid. The conditions and kinetics of photosensitive linker destruction were carefully studied. The colloid particles of IPEC were modified with Cy3 probe and their cellular internalization was investigated by flow cytometry method. The efficacy of photosensitive IPECs as siRNA and pDNA delivery system was evaluated.
Collapse
|
28
|
Peeler DJ, Luera N, Horner PJ, Pun SH, Sellers DL. Polyplex transfection from intracerebroventricular delivery is not significantly affected by traumatic brain injury. J Control Release 2020; 322:149-156. [PMID: 32198024 DOI: 10.1016/j.jconrel.2020.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/09/2020] [Accepted: 03/16/2020] [Indexed: 10/24/2022]
Abstract
Traumatic brain injury (TBI) is largely non-preventable and often kills or permanently disables its victims. Because current treatments for TBI merely ameliorate secondary effects of the initial injury like swelling and hemorrhaging, strategies for the induction of neuronal regeneration are desperately needed. Recent discoveries regarding the TBI-responsive migratory behavior and differentiation potential of neural progenitor cells (NPCs) found in the subventricular zone (SVZ) have prompted strategies targeting gene therapies to these cells to enhance neurogenesis after TBI. We have previously shown that plasmid polyplexes can non-virally transfect SVZ NPCs when directly injected in the lateral ventricles of uninjured mice. We describe the first reported intracerebroventricular transfections mediated by polymeric gene carriers in a murine TBI model and investigate the anatomical parameters that dictate transfection through this route of administration. Using both luciferase and GFP plasmid transfections, we show that the time delay between injury and polyplex injection directly impacts the magnitude of transfection efficiency, but that overall trends in the location of transfection are not affected by injury. Confocal microscopy of quantum dot-labeled plasmid uptake in vivo reveals association between our polymers and negatively charged NG2 chondroitin sulfate proteoglycans of the SVZ extracellular matrix. We further validate that glycosaminoglycans but not sulfate groups are required for polyplex uptake and transfection in vitro. These studies demonstrate that non-viral gene delivery is impacted by proteoglycan interactions and suggest the need for improved polyplex targeting materials that penetrate brain extracellular matrix to increase transfection efficiency in vivo.
Collapse
Affiliation(s)
- David J Peeler
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, United States
| | - Nicholas Luera
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, United States
| | - Philip J Horner
- Center for Neuroregeneration and Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, United States
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, United States.
| | - Drew L Sellers
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
29
|
An Engineered Gene Nanovehicle Developed for Smart Gene Therapy to Selectively Inhibit Smooth Muscle Cells: An In Vitro Study. Int J Mol Sci 2020; 21:ijms21041530. [PMID: 32102332 PMCID: PMC7073206 DOI: 10.3390/ijms21041530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/03/2022] Open
Abstract
In-stent restenosis is a serious concern for patients treated through the stenting procedure, although this can be solved using drug-eluting stents and/or drug-eluting balloon catheters. However, the chemical agents released from the drug-eluting layer for inhibiting smooth muscle cell (SMC) migration are inevitably associated with damage to vascular endothelial cell (ECs). The present in vitro study used a distinct strategy, in which a smart gene (phEGR1-PKCδ, an engineered plasmid consists of an SMC-specific promoter (human early growth response 1, hEGR1 promoter) ligated with a gene encoding apoptosis-inducing protein (protein kinase C-delta, PKCδ) was incorporated into a novel gene vehicle (Au cluster-incorporated polyethylenimine/carboxymethyl hexanoyl chitosan, PEI-Au/CHC) to form the PEI-Au/CHC/phEGR1-PKCδ complex, which was proposed for the selective inhibition of SMC proliferation. It was found that the cell viability of SMCs receiving the PEI-Au/CHC/phEGR1-PKCδ complex under simulated inflammation conditions was significantly lower than that of the ECs receiving the same treatment. In addition, the PEI-Au/CHC/phEGR1-PKCδ complex did not demonstrate an inhibitory effect on EC proliferation and migration under simulated inflammation conditions. Finally, the PEI-Au/CHC/phEGR1-PKCδ complexes coated onto a balloon catheter used in percutaneous transluminal coronary angioplasty (PTCA) could be transferred to both the ECs and the SMC layer of Sprague Dawley (SD) rat aortas ex vivo. These preliminary in vitro results suggest that the newly developed approach proposed in the present study might be a potential treatment for reducing the incidence rate of in-stent restenosis and late thrombosis in the future.
Collapse
|
30
|
Abstract
Liposomes are one of the most widely investigated carriers for CRISPR/Cas9 delivery. The surface properties of liposomal carriers, including the surface charge, PEGylation, and ligand modification can significantly affect the gene silencing efficiency. Three barriers of systemic CRISPR/Cas9 delivery (long blood circulation, efficient tumor penetration, and efficient cellular uptake/endosomal escape) are analyzed on liposomal carriers with different surface charges, PEGylations, and ligand modifications. Cationic formulations dominate CRISPR/Cas9 delivery and neutral formulations also have good performance while anionic formulations are generally not proper for CRISPR/Cas9 delivery. The PEG dilemma (prolonged blood circulation vs. reduced cellular uptake/endosomal escape) and the side effect of repeated PEGylated formulation (accelerated blood clearance) were discussed. Effects of ligand modification on cationic and neutral formulations were analyzed. Finally, we summarized the achievements in liposomal CRISPR/Cas9 delivery, outlined existing problems, and provided some future perspectives. Liposomes are one of the most widely investigated carriers for CRISPR/Cas9 delivery. The surface properties of liposomal carriers, including the surface charge, PEGylation, and ligand modification can significantly affect the gene silencing efficiency. Three barriers of systemic siRNA delivery (long blood circulation, efficient tumor penetration, and efficient cellular uptake/endosomal escape) are analyzed on liposomal carriers with different surface charges, PEGylations, and ligand modifications. Cationic formulations dominate CRISPR/Cas9 delivery and neutral formulations also have good performance while anionic formulations are generally not proper for CRISPR/Cas9 delivery. The PEG dilemma (prolonged blood circulation vs. reduced cellular uptake/endosomal escape) and the side effect of repeated PEGylated formulation (accelerated blood clearance) were discussed. Effects of ligand modification on cationic and neutral formulations were analyzed. Finally, we summarized the achievements in liposomal CRISPR/Cas9 delivery, outlined existing problems, and provided some future perspectives.
Collapse
|
31
|
He Y, Liu Y, Sun Z, Han F, Tang JZ, Gao R, Wang G. The proper strategy to compress and protect plasmid DNA in the Pluronic L64-electropulse system for enhanced intramuscular gene delivery. Regen Biomater 2019; 6:289-298. [PMID: 31616566 PMCID: PMC6783702 DOI: 10.1093/rb/rby028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/30/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022] Open
Abstract
Intramuscular expression of functional proteins is a promising strategy for therapeutic purposes. Previously, we developed an intramuscular gene delivery method by combining Pluronic L64 and optimized electropulse, which is among the most efficient methods to date. However, plasmid DNAs (pDNAs) in this method were not compressed, making them unstable and inefficient in vivo. We considered that a proper compression of pDNAs by an appropriate material should facilitate gene expression in this L64-electropulse system. Here, we reported our finding of such a material, Epigallocatechin gallate (EGCG), a natural compound in green teas, which could compress and protect pDNAs and significantly increase intramuscular gene expression in the L64-electropulse system. Meanwhile, we found that polyethylenimine (PEI) could also slightly improve exogenous gene expression in the optimal procedure. By analysing the characteristic differences between EGCG and PEI, we concluded that negatively charged materials with strong affinity to nucleic acids and/or other properties suitable for gene delivery, such as EGCG, are better alternatives than cationic materials (like PEI) for muscle-based gene delivery. The results revealed that a critical principle for material/pDNA complex benefitting intramuscular gene delivery/expression is to keep the complex negatively charged. This proof-of-concept study displays the breakthrough in compressing pDNAs and provides a principle and strategy to develop more efficient intramuscular gene delivery systems for therapeutic applications.
Collapse
Affiliation(s)
- Yutong He
- National Engineering Research Center for Biomaterials
| | - Yili Liu
- National Engineering Research Center for Biomaterials
- Key Laboratory for Bio-Resource and Eco-Environment of Ministry Education, College of Life Science, Sichuan University, No. 29, Wangjiang Road, Chengdu, Sichuan, P.R. China
| | - Zhe Sun
- National Engineering Research Center for Biomaterials
| | - Fei Han
- National Engineering Research Center for Biomaterials
| | - James Zhenggui Tang
- Research Institute in Healthcare Science, Faculty of Science and Engineering, School of Pharmacy, University of Wolverhampton, Wolverhampton, UK
| | - Rong Gao
- Key Laboratory for Bio-Resource and Eco-Environment of Ministry Education, College of Life Science, Sichuan University, No. 29, Wangjiang Road, Chengdu, Sichuan, P.R. China
| | - Gang Wang
- National Engineering Research Center for Biomaterials
| |
Collapse
|
32
|
Controlling complexation/decomplexation and sizes of polymer-based electrostatic pDNA polyplexes is one of the key factors in effective transfection. Colloids Surf B Biointerfaces 2019; 184:110497. [PMID: 31536938 DOI: 10.1016/j.colsurfb.2019.110497] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/22/2019] [Accepted: 09/07/2019] [Indexed: 12/20/2022]
Abstract
The delivery of plasmid DNA (pDNA) using polycations has been investigated for several decades; however, obstacles that limit efficient gene delivery still hinder the clinical application of gene therapy. One of the major limiting factors is controlling pDNA binding affinity with polymers to control the complexation and decomplexation of polyplexes. To address this challenge, polycations of α-poly(L-lysine) (APL) and ε-poly(L-lysine) (EPL) were used to prepare variable complexation/decomplexation polyplexes with binding affinities ranging from too tight to too loose and sizes ranging from small to large. APL-EPL/ATP-pDNA polyplexes were also prepared to compare the effects of endosomolytic ATP on complexation/decomplexation and the sizes of polyplexes. The results showed that smaller and tighter polyplexes delivered more pDNA into the cells and into the nucleus than the larger and looser polyplexes. Larger polyplexes exhibited slower cytosolic transport and consequently less nuclear delivery of pDNA than smaller polyplexes. Tighter polyplexes exhibited poor pDNA release in the nucleus, leading to no improvement in transfection efficiency. Thus, polyplexes should maintain a balance between complexation and decomplexation and should have optimal sizes for effective cellular uptake, cytosolic transport, nuclear import, and gene expression. Understanding the effects of complexation/decomplexation and size is important when designing effective polymer-based electrostatic gene carriers.
Collapse
|
33
|
BioMed Research International. Retracted: Recent Trends of Polymer Mediated Liposomal Gene Delivery System. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8195729. [PMID: 31485446 PMCID: PMC6702818 DOI: 10.1155/2019/8195729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 07/03/2019] [Indexed: 11/28/2022]
Abstract
[This retracts the article DOI: 10.1155/2014/934605.].
Collapse
|
34
|
The Impact of Nylon-3 Copolymer Composition on the Efficiency of siRNA Delivery to Glioblastoma Cells. NANOMATERIALS 2019; 9:nano9070986. [PMID: 31288448 PMCID: PMC6669510 DOI: 10.3390/nano9070986] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 02/06/2023]
Abstract
Glioblastoma multiforme is a devastating disease that has attracted enormous attention due to poor prognosis and high recurrence. Small interfering RNA (siRNA) in principle offers a promising therapeutic approach by the downregulation of disease-related genes via RNA interference. For efficient siRNA delivery to target sites, cationic polymers are often used in preclinical studies for the protection of siRNA and complex formation based on electrostatic interactions. In an effort to develop biocompatible and efficient nanocarriers with a translational outlook for optimal gene silencing at reduced toxicity, we synthesized two sets of nylon-3 copolymers with variable cationic content (DM or NM monomer) and hydrophobic subunits (CP monomer) and evaluated their suitability for in vitro siRNA delivery into glioblastoma cells. DM0.4/CP0.6 and NM0.4/CP0.6 polymers with similar subunit ratios were synthesized to compare the effect of different cationic subunits. Additionally, we utilized NM0.2/CP0.8 polymers to evaluate the impact of the different hydrophobic content in the polymer chain. The siRNA condensation ability and polymer–siRNA complex stability was evaluated by unmodified and modified SYBR gold assays, respectively. Further physicochemical characteristics, e.g., particle size and surface charge, were evaluated by dynamic light scattering and laser Doppler anemometry, whereas a relatively new method for polyplex size distribution analysis—tunable resistive pulse sensing—was additionally developed and compared to DLS measurements. Transfection efficiencies, the route of cell internalization, and protein knockdown abilities in glioblastoma cells were investigated by flow cytometry. Furthermore, cellular tolerability was evaluated by MTT and LDH assays. All the polymers efficiently condensed siRNA at N/P ratios of three, whereas polymers with NM cationic subunits demonstrated smaller particle size and lower polyplex stability. Furthermore, NM0.2/CP0.8 polyplexes with the highest hydrophobic content displayed significantly higher cellular internalization in comparison to more cationic formulations and successful knockdown capabilities. Detailed investigations of the cellular uptake route demonstrated that these polyplexes mainly follow clathrin-mediated endocytotic uptake mechanisms, implying high interaction capacity with cellular membranes. Taken together with conducive toxicity profiles, highly hydrophobic nylon-3 polymers provide an appropriate siRNA delivery agent for the potential treatment of glioblastoma.
Collapse
|
35
|
Subia B, Reinisalo M, Dey N, Tavakoli S, Subrizi A, Ganguli M, Ruponen M. Nucleic acid delivery to differentiated retinal pigment epithelial cells using cell-penetrating peptide as a carrier. Eur J Pharm Biopharm 2019; 140:91-99. [PMID: 31085311 DOI: 10.1016/j.ejpb.2019.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/16/2019] [Accepted: 05/08/2019] [Indexed: 10/26/2022]
Abstract
Nucleic acid delivery to the eye is a promising treatment strategy for many retinal disorders. In this manuscript, retinal gene delivery with non-coated and chondroitin sulphate (CS) coated amphipathic and cationic peptides was tested. The transfection and gene knockdown efficiencies were evaluated in different retinal pigment epithelial (RPE) cell models including both dividing and differentiated cells. In addition, the mobility of peptide-based gene delivery systems was examined in porcine vitreous by particle tracking analysis. The results indicate that amphipathic and cationic peptides are safe in vitro and are capable of high transgene expression and gene knockdown in dividing cells. We further demonstrate that incorporation of CS improves the efficiency of gene delivery of peptide-based systems. Most importantly, the transgene expression mediated by both non-coated and CS coated peptides was high in differentiated as well as in human primary RPE cells which are typically difficult to transfect. Coating of peptide-based gene delivery systems with CS improved diffusion in the vitreous and enhanced the stability of the polyplexes. The results indicate that a peptide-based system can be fine-tuned as a promising approach for retinal gene delivery.
Collapse
Affiliation(s)
- Bano Subia
- School of Pharmacy, University of Eastern Finland, Kuopio 70211, Finland.
| | - Mika Reinisalo
- School of Pharmacy, University of Eastern Finland, Kuopio 70211, Finland
| | - Namit Dey
- Delhi Technological University, Delhi 110042, India
| | | | - Astrid Subrizi
- School of Pharmacy, University of Eastern Finland, Kuopio 70211, Finland; Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C 800, Denmark
| | - Munia Ganguli
- CSIR-Institute of Genomics and Integrative Biology, New Delhi 110021, India
| | - Marika Ruponen
- School of Pharmacy, University of Eastern Finland, Kuopio 70211, Finland
| |
Collapse
|
36
|
Phillips HR, Tolstyka ZP, Hall BC, Hexum JK, Hackett PB, Reineke TM. Glycopolycation–DNA Polyplex Formulation N/P Ratio Affects Stability, Hemocompatibility, and in Vivo Biodistribution. Biomacromolecules 2019; 20:1530-1544. [DOI: 10.1021/acs.biomac.8b01704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Haley R. Phillips
- Center for Genome Engineering and Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Zachary P. Tolstyka
- Center for Genome Engineering and Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Bryan C. Hall
- Center for Genome Engineering and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K. Hexum
- Center for Genome Engineering and Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Perry B. Hackett
- Center for Genome Engineering and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M. Reineke
- Center for Genome Engineering and Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
37
|
Uchida S, Kataoka K. Design concepts of polyplex micelles for in vivo therapeutic delivery of plasmid DNA and messenger RNA. J Biomed Mater Res A 2019; 107:978-990. [PMID: 30665262 DOI: 10.1002/jbm.a.36614] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 11/27/2018] [Indexed: 12/20/2022]
Abstract
Nonviral delivery of plasmid (p)DNA or messenger (m)RNA is a safe and promising therapeutic option to continuously supply therapeutic proteins into diseased tissues. In most cases of in vivo pDNA and mRNA delivery, these nucleic acids are loaded into carriers based on cationic polymers and/or lipids to prevent nuclease-mediated degradation before reaching target cells. The carriers should also evade host clearance mechanisms, including uptake by scavenger cells and filtration in the spleen. Installation of ligands onto the carriers can facilitate their rapid uptake into target cells. Meanwhile, carrier toxicity should be minimized not only for preventing undesirable adverse responses in patients, but also for preserving the function of transfected cells to exert therapeutic effects. Long-term progressive improvement of platform technologies has helped overcome most of these issues, though some still remain hindering the widespread clinical application of nonviral pDNA and mRNA delivery. This review discusses design concepts of nonviral carriers for in vivo delivery and the issues to be overcome, focusing especially on our own efforts using polyplex micelles. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 978-990, 2019.
Collapse
Affiliation(s)
- Satoshi Uchida
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo 113-8656, Japan.,Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan.,Policy Alternatives Research Institute, The University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
| |
Collapse
|
38
|
Kasai H, Inoue K, Imamura K, Yuvienco C, Montclare JK, Yamano S. Efficient siRNA delivery and gene silencing using a lipopolypeptide hybrid vector mediated by a caveolae-mediated and temperature-dependent endocytic pathway. J Nanobiotechnology 2019; 17:11. [PMID: 30670041 PMCID: PMC6341701 DOI: 10.1186/s12951-019-0444-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/07/2019] [Indexed: 12/15/2022] Open
Abstract
Background We developed a non-viral vector, a combination of HIV-1 Tat peptide modified with histidine and cysteine (mTat) and polyethylenimine, jetPEI (PEI), displaying the high efficiency of plasmid DNA transfection with little toxicity. Since the highest efficiency of INTERFERin (INT), a cationic amphiphilic lipid-based reagent, for small interfering RNA (siRNA) transfection among six commercial reagents was shown, we hypothesized that combining mTat/PEI with INT would improve transfection efficiency of siRNA delivery. To elucidate the efficacy of the hybrid vector for siRNA silencing, β-actin expression was measured after siRNA β-actin was transfected with mTat/PEI/INT or other vectors in HSC-3 human oral squamous carcinoma cells. Results mTat/PEI/INT/siRNA produced significant improvement in transfection efficiency with little cytotoxicity compared to other vectors and achieved ≈ 100% knockdown of β-actin expression compared to non-treated cells. The electric charge of mTat/PEI/INT/siRNA was significantly higher than INT/siRNA. The particle size of mTat/PEI/INT/siRNA was significantly smaller than INT/siRNA. Filipin III and β-cyclodextrin, an inhibitor of caveolae-mediated endocytosis, significantly inhibited mTat/PEI/INT/siRNA transfection, while chlorpromazine, an inhibitor of clathrin-mediated endocytosis, did not inhibit mTat/PEI/INT/siRNA transfection. Furthermore, the transfection efficiency of mTat/PEI/INT at 4 °C was significantly lower than 37 °C. Conclusions These findings demonstrated the feasibility of using mTat/PEI/INT as a potentially attractive non-viral vector for siRNA delivery.
Collapse
Affiliation(s)
- Hironori Kasai
- Department of Prosthodontics, New York University College of Dentistry, New York, NY, 10010, USA
| | - Kenji Inoue
- Department of Prosthodontics, New York University College of Dentistry, New York, NY, 10010, USA
| | - Kentaro Imamura
- Department of Prosthodontics, New York University College of Dentistry, New York, NY, 10010, USA.,Department of Periodontology, Tokyo Dental College, Tokyo, Japan
| | - Carlo Yuvienco
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY, 11201, USA
| | - Jin K Montclare
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY, 11201, USA.,Department of Chemistry, New York University, New York, NY, 10003, USA.,Department of Biomaterials, New York University College of Dentistry, New York, NY, 10010, USA.,Department of Radiology, New York University School of Medicine, New York, NY, 10010, USA
| | - Seiichi Yamano
- Department of Prosthodontics, New York University College of Dentistry, New York, NY, 10010, USA.
| |
Collapse
|
39
|
Zou Y, Wan L, Blacklock J, Xie L, Carroll S, Oupicky D, Mao G. In Situ AFM Analysis Investigating Disassembly of DNA Nanoparticles and Nanofilms. Methods Mol Biol 2019; 1943:199-209. [PMID: 30838618 DOI: 10.1007/978-1-4939-9092-4_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Synthetic vector-based gene delivery continues to gain strength as viable alternatives to viral vectors due to safety and other concerns. DNA release dynamics is key to the understanding and control of gene delivery from nanosystems. Here we describe atomic force microscope (AFM) application to the understanding of DNA release dynamics from bioreducible polycation-based nanosystems. The two nanosystems are polyplex nanoparticles and layer-by-layer (LbL) films.
Collapse
Affiliation(s)
- Yi Zou
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Lei Wan
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Jenifer Blacklock
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, USA
| | - Lingxiao Xie
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Sean Carroll
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, USA
| | - David Oupicky
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
- University of Nebraska Medical Center, Omaha, NE, USA
| | - Guangzhao Mao
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
40
|
Makyła-Juzak K, Chachaj-Brekiesz A, Dynarowicz-Latka P, Dąbczyński P, Zemla J. The Effect of Dextran Sulfate-as Model Glycosaminoglycan Analogue-on Membrane Lipids: DPPC, Cholesterol, and DPPC-Cholesterol Mixture. The Monolayer Study. J Membr Biol 2018; 251:641-651. [PMID: 30030544 PMCID: PMC6244761 DOI: 10.1007/s00232-018-0041-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/09/2018] [Indexed: 01/04/2023]
Abstract
Glycosaminoglycans (GAGs) are essential components of the extracellular matrices (ECMs) located on the outer surface of cellular membranes. They belong to the group of polysaccharides involved in diverse biological processes acting on the surface and across natural lipid membranes. Recently, particular attention has been focused on possible role of GAGs in the amyloid deposits. The amyloid formation is related to a disorder in protein folding, causing that soluble-in normal conditions-peptides become deposited extracellularly as insoluble fibrils, impairing tissue structure and its function. One of the hypothesis holds that GAGs may inhibit amyloid formation by interacting with the lipid membrane by blocking the accumulation of protein aggregates on the membrane surface. Although the biophysical properties of GAGs are described rather well, little is known about the nature of association between these polysaccharides and components of natural cell membranes. Therefore, a study of GAGs influence on membrane lipids is of particular importance. The aim of the present work is to get insight into the effect of hydrophilic dextran sulfate (DS)-that can be considered as GAG analogue-on membrane lipids organization. This study was based on examining interactions between DS sodium salt of molecular weight equal to about 40 kDa (DS40), dissolved in water subphase, and a model membrane, mimicked as Langmuir monolayer, formed by representative natural membrane lipids: cholesterol and 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) as well as their mixtures. Due to the fact that calcium ions in excess may accumulate in the lipid membrane, attracting high molecular weight molecules to their surface, the influence of calcium ions present in the subphase on the DS40 activity has also been examined. It has been found that negatively charged DS, forming a sublayer underneath the monolayer, barely interacts with membrane lipids; however, in the presence of calcium ions the electrostatic interactions between DS40 and lipid membrane are significantly enhanced, leading to the formation of network-like crystalline structures at the surface of model membrane, which can prevent incorporation and interaction with other extracellular molecules, e.g., proteins.
Collapse
Affiliation(s)
- Katarzyna Makyła-Juzak
- Department of General Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Kraków, Poland.
| | - Anna Chachaj-Brekiesz
- Department of General Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Kraków, Poland
| | - Patrycja Dynarowicz-Latka
- Department of General Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Kraków, Poland
| | - Paweł Dąbczyński
- Institute of Physics, Jagiellonian University, Łojasiewicza 11, 30-348, Kraków, Poland
| | - Joanna Zemla
- Institute of Nuclear Physics, Polish Academy of Sciences, Radzikowskiego 152, 31-342, Kraków, Poland
| |
Collapse
|
41
|
González-Paredes A, Sitia L, Ruyra A, Morris CJ, Wheeler GN, McArthur M, Gasco P. Solid lipid nanoparticles for the delivery of anti-microbial oligonucleotides. Eur J Pharm Biopharm 2018; 134:166-177. [PMID: 30468838 DOI: 10.1016/j.ejpb.2018.11.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 11/18/2018] [Accepted: 11/19/2018] [Indexed: 12/29/2022]
Abstract
Novel alternatives to antibiotics are urgently needed for the successful treatment of antimicrobial resistant (AMR) infections. Experimental antibacterial oligonucleotide therapeutics, such as transcription factor decoys (TFD), are a promising approach to circumvent AMR. However, the therapeutic potential of TFD is contingent upon the development of carriers that afford efficient DNA protection against nucleases and delivery of DNA to the target infection site. As a carrier for TFD, here we present three prototypes of anionic solid lipid nanoparticles that were coated with either the cationic bolaamphiphile 12-bis-tetrahydroacridinium or with protamine. Both compounds switched particles zeta potential to positive values, showing efficient complexation with TFD and demonstrable protection from deoxyribonuclease. The effective delivery of TFD into bacteria was confirmed by confocal microscopy while SLN-bacteria interactions were studied by flow cytometry. Antibacterial efficacy was confirmed using a model TFD targeting the Fur iron uptake pathway in E. coli under microaerobic conditions. Biocompatibility of TFD-SLN was assessed using in vitro epithelial cell and in vivo Xenopus laevis embryo models. Taken together these results indicate that TFD-SLN complex can offer preferential accumulation of TFD in bacteria and represent a promising class of carriers for this experimental approach to tackling the worldwide AMR crisis.
Collapse
Affiliation(s)
| | - Leopoldo Sitia
- Procarta Biosystems Ltd., Innovation Centre, Norwich Research Park, Norwich NR4 7UH, UK; Norwich Medical School, University of East Anglia, Norwich NR4 7UQ, UK
| | - Angels Ruyra
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | | | - Grant N Wheeler
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Michael McArthur
- Procarta Biosystems Ltd., Innovation Centre, Norwich Research Park, Norwich NR4 7UH, UK; Norwich Medical School, University of East Anglia, Norwich NR4 7UQ, UK
| | - Paolo Gasco
- Nanovector Srl., Via Livorno 60, 10144 Turin, Italy
| |
Collapse
|
42
|
Feng G, Zha Z, Huang Y, Li J, Wang Y, Ke W, Chen H, Liu L, Song Y, Ge Z. Sustained and Bioresponsive Two-Stage Delivery of Therapeutic miRNA via Polyplex Micelle-Loaded Injectable Hydrogels for Inhibition of Intervertebral Disc Fibrosis. Adv Healthc Mater 2018; 7:e1800623. [PMID: 30296017 DOI: 10.1002/adhm.201800623] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 08/09/2018] [Indexed: 02/05/2023]
Abstract
Intervertebral disc degeneration (IDD) is frequently caused by gradual pathological changes inside intervertebral discs (IVDs) and progressive fibrosis. MicroRNA-29 (miR-29) family possesses potent fibrosis suppression capability, but their application for treatment of chronic IDD is limited due to lack of suitable local delivery systems. In this report, given various overexpressed matrix metalloproteinases (MMPs) during IDD, injectable MMP-degradable hydrogels encapsulating MMP-responsive polyplex micelles are developed for sustained and bioresponsive delivery of miR-29a into nucleus pulposus cells via a two-stage process. Cationic block copolymers are designed to complex miR-29a, and subsequently mixed with the poly(ethylene glycol) (PEG) gelation precursors and MMP-cleavable peptide cross-linkers for in situ formation of polyplex micelle-encapsulated hydrogels in the diseased IVDs. In the presence of MMPs, the polyplex micelles are first released by MMP cleavage of the hydrogels, and subsequently, MMPs-responsive detachment of PEG shells from polyplex micelles contributes to efficient cellular uptake and endosomal escape. MiR-29a is demonstrated to effectively silence the expression of MMP-2, inhibit the fibrosis process, and reverse IDD in animal models through blocking the β-catenin translocation pathway from the cytoplasm to the nucleus. This two-stage bioresponsive local miRNA delivery system represents a novel and promising strategy for the treatment of chronic IDD.
Collapse
Affiliation(s)
- Ganjun Feng
- Department of Orthopedic Surgery; West China Hospital; Sichuan University; Chengdu 610041 Sichuan China
| | - Zengshi Zha
- CAS Key Laboratory of Soft Matter Chemistry; Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei 230026 Anhui China
| | - Yong Huang
- Department of Orthopedic Surgery; West China Hospital; Sichuan University; Chengdu 610041 Sichuan China
| | - Junjie Li
- CAS Key Laboratory of Soft Matter Chemistry; Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei 230026 Anhui China
| | - Yuheng Wang
- CAS Key Laboratory of Soft Matter Chemistry; Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei 230026 Anhui China
| | - Wendong Ke
- CAS Key Laboratory of Soft Matter Chemistry; Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei 230026 Anhui China
| | - Hongying Chen
- Technology Center for Public Research; West China Hospital; Sichuan University; Chengdu 610041 Sichuan China
| | - Limin Liu
- Department of Orthopedic Surgery; West China Hospital; Sichuan University; Chengdu 610041 Sichuan China
| | - Yueming Song
- Department of Orthopedic Surgery; West China Hospital; Sichuan University; Chengdu 610041 Sichuan China
| | - Zhishen Ge
- CAS Key Laboratory of Soft Matter Chemistry; Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei 230026 Anhui China
| |
Collapse
|
43
|
Munsell EV, Fang B, Sullivan MO. Histone-Mimetic Gold Nanoparticles as Versatile Scaffolds for Gene Transfer and Chromatin Analysis. Bioconjug Chem 2018; 29:3691-3704. [PMID: 30350573 DOI: 10.1021/acs.bioconjchem.8b00611] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Histone-inspired polymer assemblies (polyplexes) can regulate gene expression and subcellular transport in plasmids by harnessing the cellular machinery normally used for histone proteins. When grafted to polyplexes, histone tails promote nuclear accumulation, trigger plasmid DNA (pDNA) release, and enhance transcription. Herein, we developed multifunctional gold nanoparticles (AuNPs) decorated by histone motifs as histone-inspired scaffolds with improved pDNA binding, easy bioimaging, and increased potential for gene delivery and chromatin analysis applications. We hypothesized that polycationic AuNPs coupled to histone motifs would mimic the native presentation of these sequences on the histone octamer and thereby create structures with the capacity to both engage native histone effectors and condense pDNA into nucleosome-inspired nanostructures. AuNPs bearing ∼2 nm cores were prepared based on the well-established Brust-Schiffrin two-phase method involving tetrachloroaurate reduction in the presence of 1-pentanethiol. Solid phase peptide synthesis was employed to generate thiolated polycationic ligands and histone tail motifs, and the AuNPs and peptide ligands were combined in a two-step Murray place exchange reaction at various ratios to produce a collection of polycationic AuNPs modified with varying amounts of histone tails. Electron microscopy and thermal analyses demonstrated that these modified AuNPs exhibited tunable biochemical and biophysical properties that closely mimicked the properties of native histones. The histone-mimetic nanoscaffolds efficiently and sequence-specifically engaged histone effectors responsible for activating transcription. In addition, the nanoscaffolds condensed pDNA into complexes with high stability in the presence of physiological concentrations of heparin, a common extracellular polyanion. These combined properties of histone engagement and high stability led to a ∼6-fold enhancement in transfection efficiency as compared with typical polymeric transfection reagents, with the increased transfection efficiency correlated to the presence and amount of histone tails displayed on the surface of the nanoscaffolds. These findings demonstrate the utility of employing a biomimetic materials design approach to develop more effective and stable delivery vehicles for gene transfer and chromatin analysis applications.
Collapse
Affiliation(s)
- Erik V Munsell
- Department of Chemical and Biomolecular Engineering , University of Delaware , 150 Academy Street , Newark , Delaware 19716 , United States
| | - Bing Fang
- Department of Chemical and Biomolecular Engineering , University of Delaware , 150 Academy Street , Newark , Delaware 19716 , United States
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering , University of Delaware , 150 Academy Street , Newark , Delaware 19716 , United States
| |
Collapse
|
44
|
Abstract
The genetic modification of human T lymphocytes with established non-viral methods is inefficient. Linear polyethylenimine (l-PEI), one of the most popular non-viral transfection agents for mammalian cells in general, only achieves transfection rates in the single digit percentage range for these cells. Here, a well-defined 24-armed poly(2-dimethylamino) ethyl methacrylate (PDMAEMA) nanostar (number average of the molecular weight: 755 kDa, polydispersity: <1.21) synthesized via atom transfer radical polymerization (ATRP) from a silsesquioxane initiator core is proposed as alternative. The agent is used to prepare polyplexes with plasmid DNA (pDNA). Under optimal conditions these polyplexes reproducibly transfect >80% of the cells from a human T-cell leukemia cell line (Jurkat cells) at viabilities close to 90%. The agent also promotes pDNA uptake when simply added to a mixture of cells and pDNA. This constitutes a particular promising approach for efficient transient transfection at large scale. Finally, preliminary experiments were carried out with primary T cells from two different donors. Results were again significantly better than for l-PEI, although further research into the response of individual T cells to the transfection agent will be necessary, before either method can be used to routinely transfect primary T lymphocytes.
Collapse
|
45
|
Muro S. Alterations in Cellular Processes Involving Vesicular Trafficking and Implications in Drug Delivery. Biomimetics (Basel) 2018; 3:biomimetics3030019. [PMID: 31105241 PMCID: PMC6352689 DOI: 10.3390/biomimetics3030019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 12/31/2022] Open
Abstract
Endocytosis and vesicular trafficking are cellular processes that regulate numerous functions required to sustain life. From a translational perspective, they offer avenues to improve the access of therapeutic drugs across cellular barriers that separate body compartments and into diseased cells. However, the fact that many factors have the potential to alter these routes, impacting our ability to effectively exploit them, is often overlooked. Altered vesicular transport may arise from the molecular defects underlying the pathological syndrome which we aim to treat, the activity of the drugs being used, or side effects derived from the drug carriers employed. In addition, most cellular models currently available do not properly reflect key physiological parameters of the biological environment in the body, hindering translational progress. This article offers a critical overview of these topics, discussing current achievements, limitations and future perspectives on the use of vesicular transport for drug delivery applications.
Collapse
Affiliation(s)
- Silvia Muro
- Institute for Bioscience and Biotechnology Research and Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA.
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain.
- Institute for Bioengineering of Catalonia (IBEC) of the Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain.
| |
Collapse
|
46
|
Ho JK, White PJ, Pouton CW. Self-Crosslinking Lipopeptide/DNA/PEGylated Particles: A New Platform for DNA Vaccination Designed for Assembly in Aqueous Solution. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:504-517. [PMID: 30195787 PMCID: PMC6077166 DOI: 10.1016/j.omtn.2018.05.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 05/20/2018] [Accepted: 05/30/2018] [Indexed: 12/31/2022]
Abstract
Delivery of plasmids for gene expression in vivo is an inefficient process that requires improvement and optimization to unlock the clinical potential of DNA vaccines. With ease of manufacture and biocompatibility in mind, we explored condensation of DNA in aqueous solution with a self-crosslinking, endosome-escaping lipopeptide (LP), stearoyl-Cys-His-His-Lys-Lys-Lys-amide (stearoyl-CH2K3), to produce cationic LP/DNA complexes. To test whether poly(ethylene glycol) (PEG)-ylation of these cationic complexes to neutralize the surface charge would improve the distribution, gene expression, and immune responses poly(ethylene glycol), these LP/DNA complexes were combined with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (DSPE-PEG2000). Fluorescence imaging illustrated that the cationic complexes exhibited the highest degree of localization and lowest degree of dispersion throughout the injected muscle, suggesting impaired mobility of cationic particles upon administration. Nanoluciferase reporter assays over a 90-day period demonstrated that gene expression levels in muscle were highest for PEGylated particles, with over a 200-fold higher level of expression than the cationic particles observed at 30 days. Humoral and cell-mediated immune responses were evaluated in vivo after injection of an ovalbumin expression plasmid. PEGylation improved both immune responses to the DNA complexes in mice. Overall, this suggests that PEGylation of cationic lipopeptide complexes can significantly improve both the transgene expression and immunogenicity of intramuscular DNA vaccines.
Collapse
Affiliation(s)
- Joan K Ho
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, VIC, Australia
| | - Paul J White
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, VIC, Australia
| | - Colin W Pouton
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, VIC, Australia.
| |
Collapse
|
47
|
Cabral H, Miyata K, Osada K, Kataoka K. Block Copolymer Micelles in Nanomedicine Applications. Chem Rev 2018; 118:6844-6892. [PMID: 29957926 DOI: 10.1021/acs.chemrev.8b00199] [Citation(s) in RCA: 838] [Impact Index Per Article: 119.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Polymeric micelles are demonstrating high potential as nanomedicines capable of controlling the distribution and function of loaded bioactive agents in the body, effectively overcoming biological barriers, and various formulations are engaged in intensive preclinical and clinical testing. This Review focuses on polymeric micelles assembled through multimolecular interactions between block copolymers and the loaded drugs, proteins, or nucleic acids as translationable nanomedicines. The aspects involved in the design of successful micellar carriers are described in detail on the basis of the type of polymer/payload interaction, as well as the interplay of micelles with the biological interface, emphasizing on the chemistry and engineering of the block copolymers. By shaping these features, polymeric micelles have been propitious for delivering a wide range of therapeutics through effective sensing of targets in the body and adjustment of their properties in response to particular stimuli, modulating the activity of the loaded drugs at the targeted sites, even at the subcellular level. Finally, the future perspectives and imminent challenges for polymeric micelles as nanomedicines are discussed, anticipating to spur further innovations.
Collapse
Affiliation(s)
| | | | | | - Kazunori Kataoka
- Innovation Center of NanoMedicine , Kawasaki Institute of Industrial Promotion , 3-25-14, Tonomachi , Kawasaki-ku , Kawasaki 210-0821 , Japan.,Policy Alternatives Research Institute , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku , Tokyo 113-0033 , Japan
| |
Collapse
|
48
|
Nisakar D, Vij M, Pandey T, Natarajan P, Sharma R, Mishra S, Ganguli M. Deciphering the Role of Chondroitin Sulfate in Increasing the Transfection Efficiency of Amphipathic Peptide-Based Nanocomplexes. ACS Biomater Sci Eng 2018; 5:45-55. [PMID: 33405865 DOI: 10.1021/acsbiomaterials.8b00069] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Glycosaminoglycans, both cell-surface and exogenous, can interfere with DNA delivery efficiency of nonviral carrier systems. In this work, we report an extensive comparative study to explore the effect of exogenously added chondroitin sulfate on biophysical characteristics, cellular uptake, transfection efficiency, and intracellular trafficking of nanocomplexes formed using primary and secondary amphipathic peptides developed in our laboratory. Our results indicate that the presence of exogenous chondroitin sulfate exhibits differential enhancement in transfection efficiency of the amphipathic peptides depending upon their chemical nature. The enhancement was more pronounced in primary amphipathic peptide-based nanocomplexes as compared to the secondary counterpart. This difference can be attributed to possible alteration of the intracellular entry pathway in addition to increased extracellular stability, less cellular toxicity, and assistance in nuclear accumulation. These results imply potential use of glycosaminoglycans such as chondroitin sulfate to improve the transfection efficiency of primary amphipathic peptides for possible in vivo applications.
Collapse
Affiliation(s)
- Daniel Nisakar
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India
| | - Manika Vij
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India.,Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India
| | - Tanuja Pandey
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India
| | - Poornemaa Natarajan
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India
| | - Rajpal Sharma
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India
| | - Sarita Mishra
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India.,Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India
| | - Munia Ganguli
- CSIR-Institute of Genomics and Integrative Biology, South Campus, Mathura Road, Opp: Sukhdev Vihar Bus Depot, New Delhi 110020, India.,Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India
| |
Collapse
|
49
|
Deshpande S, Singh N. Probing the nanoparticle-AGO2 interaction for enhanced gene knockdown. SOFT MATTER 2018; 14:4169-4177. [PMID: 29687822 DOI: 10.1039/c8sm00534f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
RNAi is emerging as a promising technology for treatment of various diseases due to its ability to silence specific target genes. To date, a number of nanoparticle based formulations have been reported for the delivery of small interfering RNA (siRNA), with continuous modifications in the nanoparticle design for enhancing their efficiency. While majority of the design aspects are focused on avoiding or overcoming endosomal entrapment, limited studies are available that address the role of interaction of nanoparticles with the RNA induced silencing complex (RISC) machinery, which is a crucial aspect deciding the outcome. Here, we systematically probed the effect of steric hindrance of nanoparticles on RISC interaction, by modulating two parameters, nanoparticle size and hardness. An assay was developed for quantifying the extent of RISC interaction of different nanoparticles in vitro, which was then correlated with their gene knockdown efficiency. The results suggest that the soft and small nanoparticles were most efficacious in knocking down polo-like-kinase 1 (PLK1) siRNA, a gene overexpressed in a variety of cancer types.
Collapse
Affiliation(s)
- Sonal Deshpande
- Centre for Biomedical Engineering, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi-110016, India. sneetu.iitd.ac.in
| | | |
Collapse
|
50
|
Abstract
Immune-mediated diseases are emerging as a major healthcare concern in the present era. TNF-α, a proinflammatory cytokine, plays a major role in the manifestation of these diseases by mediating different pathways and inducing the expression of other cytokines. In last decades, monoclonal antibodies and extracellular portion of human TNF-α receptors are explored in this area; however, the risk of immunological response and undesired effects urge a need to develop more effective therapies to control TNF-α levels. siRNA therapeutic strategies are emerging for the treatment of myriad of diseases, but the delivery challenges associated with siRNA require the development of suitable delivery vectors. For delivery of TNF-α siRNA, both viral and nonviral vectors are explored. This review attempts to describe different delivery approaches for TNF-α siRNA with special focus on nonviral delivery vectors.
Collapse
|