1
|
Zhang H, Gao X, Sun Q, Dong X, Zhu Z, Yang C. Incorporation of poly(γ-glutamic acid) in lipid nanoparticles for enhanced mRNA delivery efficiency in vitro and in vivo. Acta Biomater 2024; 177:361-376. [PMID: 38342193 DOI: 10.1016/j.actbio.2024.02.004] [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: 09/09/2023] [Revised: 01/26/2024] [Accepted: 02/04/2024] [Indexed: 02/13/2024]
Abstract
Messenger RNA (mRNA)-based therapy shows immense potential for broad biomedical applications. However, the development of safe and efficacious mRNA delivery vectors remains challenging due to delivery barriers and inefficient intracellular payload release. Herein, we presented a simple strategy to boost the mRNA intracellular release by incorporation of anionic poly(γ-glutamic acid) (PGA) into an ionizable lipid-based LNP/mRNA. We systematically investigated the impact of PGA incorporation on mRNA transfection both in vitro and in vivo. The molecular weights and formulation ratios of PGA greatly affected the transfection efficacy of LNP/mRNA. From in vitro study, the optimized LNP/mRNA/PGA was formulated by incorporation of PGA with the molecular weight of 80 kDa or 200 kDa and the charge ratio (N/P/C) of 25/1/1. The optimized formulation achieved around 3-fold mRNA expression in HeLa cells compared to the bare LNP/mRNA. The intracellular releasing study using specific DNA probe revealed that this enhancement of transfection efficacy was attributed to the elevated mRNA release into cytoplasm. Moreover, the optimized LNP/mRNA/PGA achieved up to 5-fold or 3-fold increase of luciferase mRNA expression in vivo after being injected into mice systematically or intramuscularly, respectively. In addition, the incorporation of PGA did not significantly alter the biodistribution profile of the complexes on both organ and cellular levels. Therefore, our work provides a simple strategy to boost mRNA delivery, which holds great promise to improve the efficacy of mRNA therapeutics for various biomedical applications. STATEMENT OF SIGNIFICANCE: The process of designing and screening potent mRNA carriers is complicated and time-consuming, while the efficacy is not always satisfying due to the delivery barriers and inefficient mRNA release. This work presented an alternative strategy to boost the mRNA delivery efficacy by incorporating an anionic natural polymer poly(γ-glutamic acid) (PGA) into LNP/mRNA complexes. The optimized LNP/mRNA/PGA achieved up to 3-fold and 5-fold increase in transfection efficacy in vitro and in vivo, respectively. Intracellular releasing analysis revealed that the enhancement of transfection efficacy was mainly attributed to the elevated intracellular release of mRNA. In addition, the incorporation of PGA did not alter the biodistribution or the biosafety profile of the complexes. These findings indicate that PGA incorporation is a promising strategy to improve the efficacy of mRNA therapeutics.
Collapse
Affiliation(s)
- Hongqian Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and School of Chemistry and Chemical Engineering, Shandong University, Jinan 25010, China
| | - Xue Gao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and School of Chemistry and Chemical Engineering, Shandong University, Jinan 25010, China
| | - Qian Sun
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and School of Chemistry and Chemical Engineering, Shandong University, Jinan 25010, China
| | - Xiaoxue Dong
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and School of Chemistry and Chemical Engineering, Shandong University, Jinan 25010, China
| | - Zongwei Zhu
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and School of Chemistry and Chemical Engineering, Shandong University, Jinan 25010, China
| | - Chuanxu Yang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and School of Chemistry and Chemical Engineering, Shandong University, Jinan 25010, China.
| |
Collapse
|
2
|
Okuyama R. mRNA and Adenoviral Vector Vaccine Platforms Utilized in COVID-19 Vaccines: Technologies, Ecosystem, and Future Directions. Vaccines (Basel) 2023; 11:1737. [PMID: 38140142 PMCID: PMC10748114 DOI: 10.3390/vaccines11121737] [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: 10/27/2023] [Revised: 11/12/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
New technological platforms, such as mRNA and adenoviral vector vaccines, have been utilized to develop coronavirus disease 2019 (COVID-19) vaccines. These new modalities enable rapid and flexible vaccine design and cost-effective and swift manufacturing, effectively combating pandemics caused by mutating viruses. Innovation ecosystems, including universities, startups, investors, and governments are crucial for developing these cutting-edge technologies. This review summarizes the research and development trajectory of these vaccine technologies, their investments, and the support surrounding them, in addition to the technological details of each technology. In addition, this study examines the importance of an innovation ecosystem in developing novel technologies, comparing it with the case of Japan, which has lagged behind in COVID-19 vaccine development. It also explores the direction of vaccine development in the post-COVID-19 era.
Collapse
Affiliation(s)
- Ryo Okuyama
- College of International Management, Ritsumeikan Asia Pacific University, Beppu 874-8577, Japan
| |
Collapse
|
3
|
Kimura S, Harashima H. On the mechanism of tissue-selective gene delivery by lipid nanoparticles. J Control Release 2023; 362:797-811. [PMID: 37004796 DOI: 10.1016/j.jconrel.2023.03.052] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/04/2023]
Abstract
The era of nucleic acid nanomedicine has arrived, as evidenced by Patisiran, a small interfering RNA (siRNA) encapsulated lipid nanoparticle (LNP), and mRNA-loaded LNPs used in COVID-19 vaccines. The diversity of nano-designs for delivering nucleic acid molecules tested in Phase II/III clinical trials reflects the potential of these technologies. These breakthroughs in non-viral gene delivery, including the use of LNPs, have attracted substantial interest worldwide for developing more effective drugs. A next step in this field is to target tissues other than the liver, which requires significant research efforts and material development. However, mechanistic studies in this area are lacking. This study compares two types of LNPs with different tissue-selectivity for delivering plasmid DNA (pDNA), one being liver-selective and the other spleen-selective, in an effort to understand the mechanisms responsible for differences in gene expression of delivered genes. We observed little difference in the biodistribution of these two LNPs despite the 100-1000-fold differences in gene expression. We then quantified the amount of delivered pDNA and mRNA expression in each tissue by quantitative real-time PCR (qPCR) to evaluate various intracellular processes, such as nuclear delivery, transcription and translation. The results showed a >100-fold difference in the translation step but there were little differences in amount of pDNA delivered to the nucleus or the amount of mRNA expression for the two LNP deliveries. Our findings suggest that endogenous factors affect gene expression efficiency not the extent of biodistribution.
Collapse
Affiliation(s)
- Seigo Kimura
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan; Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan; Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
4
|
Hu D, Fumoto S, Yoshikawa N, Peng J, Miyamoto H, Tanaka M, Nishida K. Diffusion coefficient of cationic liposomes during lipoplex formation determines transfection efficiency in HepG2 cells. Int J Pharm 2023; 637:122881. [PMID: 36963641 DOI: 10.1016/j.ijpharm.2023.122881] [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: 12/02/2022] [Revised: 02/21/2023] [Accepted: 03/19/2023] [Indexed: 03/26/2023]
Abstract
Cationic lipid-based lipoplexes are well-known for gene delivery. To determine the relationship between physicochemical characteristics and transfection efficiency, cationic liposomes of different sizes were prepared and incubated with plasmid DNA at different temperatures to form lipoplexes. We found that the liposome diffusion coefficient during lipoplex formation strongly correlated with the physicochemical characteristics of lipoplexes, accessibility of plasmid DNA in lipoplexes, and logarithm of gene expression per metabolic activity. Clathrin-mediated endocytosis was the major route for lipoplexes comprising 100 nm-liposomes, as reported previously. As liposome size increased, the major route shifted to lipid raft-mediated endocytosis. In addition, macropinocytosis was observed for all liposome sizes. The role of reactive oxygen species might depend on liposome size and endocytosis. Information from this study would be useful for understanding cationic lipoplex-mediated transfection.
Collapse
Affiliation(s)
- Die Hu
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Shintaro Fumoto
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan.
| | - Naoki Yoshikawa
- Department of Pharmacy, University of Miyazaki Hospital, 5200 Kihara, Kiyotake-Cho, Miyazaki 889-1692, Japan
| | - Jianqing Peng
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang, 550025, China
| | - Hirotaka Miyamoto
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Masakazu Tanaka
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Koyo Nishida
- Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| |
Collapse
|
5
|
Harashima H. Innovative System for Delivering Nucleic Acids/Genes Based on Controlled Intracellular Trafficking as Well as Controlled Biodistribution for Nanomedicines. Biol Pharm Bull 2023; 46:1648-1660. [PMID: 38044089 DOI: 10.1248/bpb.b23-00634] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
This review paper summarizes progress that has been made in the new field of "Controlled Intracellular Trafficking." This involves the development of new systems for delivering plasmid DNA (pDNA), small interfering RNA (siRNA), mRNA, proteins, their escape from endosomes, the mechanism for how they enter the nucleus, how they enter mithochondria and how materials subsequently function within a cell. In addition, strategies for delivering these materials to a selective tissue after intravenous administration was also intensively investigated not only to the liver but also to tumors, lungs, adipose tissue and the spleen. In 2020, a new mRNA vaccine was developed against coronavirus disease 2019 (COVID-19), where ionizable cationic lipids were used as a delivery system. Our strategy to identify an efficient ionizable cationic lipids (iCL) based on a lipid library as well as their applications concerning the delivery of siRNA/mRNA/pDNA is also described.
Collapse
Affiliation(s)
- Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University
| |
Collapse
|
6
|
Wang J, Du L, Chen X. Oncolytic virus: A catalyst for the treatment of gastric cancer. Front Oncol 2022; 12:1017692. [PMID: 36505792 PMCID: PMC9731121 DOI: 10.3389/fonc.2022.1017692] [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: 08/12/2022] [Accepted: 11/02/2022] [Indexed: 11/25/2022] Open
Abstract
Gastric cancer (GC) is a leading contributor to global cancer incidence and mortality. According to the GLOBOCAN 2020 estimates of incidence and mortality for 36 cancers in 185 countries produced by the International Agency for Research on Cancer (IARC), GC ranks fifth and fourth, respectively, and seriously threatens the survival and health of people all over the world. Therefore, how to effectively treat GC has become an urgent problem for medical personnel and scientific workers at this stage. Due to the unobvious early symptoms and the influence of some adverse factors such as tumor heterogeneity and low immunogenicity, patients with advanced gastric cancer (AGC) cannot benefit significantly from treatments such as radical surgical resection, radiotherapy, chemotherapy, and targeted therapy. As an emerging cancer immunotherapy, oncolytic virotherapies (OVTs) can not only selectively lyse cancer cells, but also induce a systemic antitumor immune response. This unique ability to turn unresponsive 'cold' tumors into responsive 'hot' tumors gives them great potential in GC therapy. This review integrates most experimental studies and clinical trials of various oncolytic viruses (OVs) in the diagnosis and treatment of GC. It also exhaustively introduces the concrete mechanism of invading GC cells and the viral genome composition of adenovirus and herpes simplex virus type 1 (HSV-1). At the end of the article, some prospects are put forward to determine the developmental directions of OVTs for GC in the future.
Collapse
Affiliation(s)
- Junqing Wang
- School of the 1st Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linyong Du
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China,*Correspondence: Xiangjian Chen, ; Linyong Du,
| | - Xiangjian Chen
- School of the 1st Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China,*Correspondence: Xiangjian Chen, ; Linyong Du,
| |
Collapse
|
7
|
Akita H. Development of an SS-Cleavable pH-Activated Lipid-Like Material (ssPalm) as a Nucleic Acid Delivery Device. Biol Pharm Bull 2021; 43:1617-1625. [PMID: 33132308 DOI: 10.1248/bpb.b20-00534] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Gene and nucleic acid-based medication is an ultimate strategy in the field of personalized medicine. A gene or short interference RNA (siRNA) molecule needs to be delivered to the appropriate organelle (i.e., nucleus and cytoplasm, respectively). We recently focused on improving the intrinsic activity of my original material (ssPalm) in terms of endosomal/lysosomal membrane destabilization activity by chemically modifying the tertiary amine structure. In parallel, I have been expanding the range of applications of ssPalms. The first application is a DNA or RNA vaccine. My crucial finding is that the vitamin E-scaffold ssPalm (ssPalmE) is highly immune-stimulative when combined with DNA. Thereafter, I redesigned the hydrophobic scaffold structure, and found that an oleic acid-scaffold ssPalm (ssPalmO) can confer anti-inflammatory characteristics. Based on this result, I further upgraded the ssPalmO, by inserting a newly designed linker with self-degradable properties.
Collapse
Affiliation(s)
- Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University
| |
Collapse
|
8
|
Somiya M, Sakaeda K, Ishii Y, Kuroda S. Cytoplasmic delivery of small interfering RNA by photoresponsive non-cationic liposomes. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
9
|
Berger S, Krhač Levačić A, Hörterer E, Wilk U, Benli-Hoppe T, Wang Y, Öztürk Ö, Luo J, Wagner E. Optimizing pDNA Lipo-polyplexes: A Balancing Act between Stability and Cargo Release. Biomacromolecules 2021; 22:1282-1296. [PMID: 33616407 DOI: 10.1021/acs.biomac.0c01779] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
When optimizing nanocarriers, structural motifs that are beneficial for the respective type of cargo need to be identified. Here, succinoyl tetraethylene pentamine (Stp)-based lipo-oligoaminoamides (OAAs) were optimized for the delivery of plasmid DNA (pDNA). Structural variations comprised saturated fatty acids with chain lengths between C2 and C18 and terminal cysteines as units promoting nanoparticle stabilization, histidines for endosomal buffering, and disulfide building blocks for redox-sensitive release. Biophysical and tumor cell culture screening established clear-cut relationships between lipo-OAAs and characteristics of the formed pDNA complexes. Based on the optimized alternating Stp-histidine backbones, lipo-OAAs containing fatty acids with chain lengths around C6 to C10 displayed maximum gene transfer with around 500-fold higher gene expression than that of C18 lipo-OAA analogues. Promising lipo-OAAs, however, showed only moderate in vivo efficiency. In vitro testing in 90% full serum, revealing considerable inhibition of lytic and gene-transfer activity, was found as a new screening model predictive for intravenous applications in vivo.
Collapse
Affiliation(s)
- Simone Berger
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Ana Krhač Levačić
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Elisa Hörterer
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Ulrich Wilk
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Teoman Benli-Hoppe
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Yanfang Wang
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Özgür Öztürk
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Jie Luo
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich 81377, Germany
| |
Collapse
|
10
|
Fumoto S, Yamamoto T, Okami K, Maemura Y, Terada C, Yamayoshi A, Nishida K. Understanding In Vivo Fate of Nucleic Acid and Gene Medicines for the Rational Design of Drugs. Pharmaceutics 2021; 13:159. [PMID: 33530309 PMCID: PMC7911509 DOI: 10.3390/pharmaceutics13020159] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
Nucleic acid and genetic medicines are increasingly being developed, owing to their potential to treat a variety of intractable diseases. A comprehensive understanding of the in vivo fate of these agents is vital for the rational design, discovery, and fast and straightforward development of the drugs. In case of intravascular administration of nucleic acids and genetic medicines, interaction with blood components, especially plasma proteins, is unavoidable. However, on the flip side, such interaction can be utilized wisely to manipulate the pharmacokinetics of the agents. In other words, plasma protein binding can help in suppressing the elimination of nucleic acids from the blood stream and deliver naked oligonucleotides and gene carriers into target cells. To control the distribution of these agents in the body, the ligand conjugation method is widely applied. It is also important to understand intracellular localization. In this context, endocytosis pathway, endosomal escape, and nuclear transport should be considered and discussed. Encapsulated nucleic acids and genes must be dissociated from the carriers to exert their activity. In this review, we summarize the in vivo fate of nucleic acid and gene medicines and provide guidelines for the rational design of drugs.
Collapse
Affiliation(s)
- Shintaro Fumoto
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan; (T.Y.); (K.O.); (Y.M.); (C.T.); (A.Y.); (K.N.)
| | | | | | | | | | | | | |
Collapse
|
11
|
Hagino Y, Khalil IA, Kimura S, Kusumoto K, Harashima H. GALA-Modified Lipid Nanoparticles for the Targeted Delivery of Plasmid DNA to the Lungs. Mol Pharm 2021; 18:878-888. [PMID: 33492961 DOI: 10.1021/acs.molpharmaceut.0c00854] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This study describes the development of lipid nanoparticles (LNPs) for the efficient and selective delivery of plasmid DNA (pDNA) to the lungs. The GALA peptide was used as a ligand to target the lung endothelium and as an endosomal escape device. Transfection activity in the lungs was significantly improved when pDNA was encapsulated in double-coated LNPs. The inner coat was composed of dioleoylphsophoethanolamine and a stearylated octaarginine (STR-R8) peptide, while the outer coat was largely a cationic lipid, di-octadecenyl-trimethylammonium propane, mixed with YSK05, a pH-sensitive lipid, and cholesterol. Optimized amounts of YSK05 and GALA were used to achieve an efficient and lung-selective system. The optimized system produced a high gene expression level in the lungs (>107 RLU/mg protein) with high lung/liver and lung/spleen ratios. GALA/R8 modification and the double-coating design were indispensable for efficient gene expression in the lungs. Despite the fact that NPs prepared with 1-step or 2-step coating have the same lipid amount and composition and the same pDNA dose, the transfection activity was dramatically higher in the lungs in the case of 2-step coating. Surprisingly, 1-step or 2-step coatings had no effect on the amount of nanoparticles that were delivered to the lungs, suggesting that the double-coating strategy substantially improved the efficiency of gene expression at the intracellular level.
Collapse
Affiliation(s)
- Yuta Hagino
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Ikramy A Khalil
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.,Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Seigo Kimura
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Kenji Kusumoto
- Formulation Research Laboratory, Taiho Pharmaceutical Co., Ltd., 224-2, Ebisuno, Hiraishi, Kawauchi-cho, Tokushima 771-0194, Japan
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.,Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| |
Collapse
|
12
|
Current Status and Challenges Associated with CNS-Targeted Gene Delivery across the BBB. Pharmaceutics 2020; 12:pharmaceutics12121216. [PMID: 33334049 PMCID: PMC7765480 DOI: 10.3390/pharmaceutics12121216] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/19/2020] [Accepted: 12/11/2020] [Indexed: 12/21/2022] Open
Abstract
The era of the aging society has arrived, and this is accompanied by an increase in the absolute numbers of patients with neurological disorders, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). Such neurological disorders are serious costly diseases that have a significant impact on society, both globally and socially. Gene therapy has great promise for the treatment of neurological disorders, but only a few gene therapy drugs are currently available. Delivery to the brain is the biggest hurdle in developing new drugs for the central nervous system (CNS) diseases and this is especially true in the case of gene delivery. Nanotechnologies such as viral and non-viral vectors allow efficient brain-targeted gene delivery systems to be created. The purpose of this review is to provide a comprehensive review of the current status of the development of successful drug delivery to the CNS for the treatment of CNS-related disorders especially by gene therapy. We mainly address three aspects of this situation: (1) blood-brain barrier (BBB) functions; (2) adeno-associated viral (AAV) vectors, currently the most advanced gene delivery vector; (3) non-viral brain targeting by non-invasive methods.
Collapse
|
13
|
The use of a MITO-Porter to deliver exogenous therapeutic RNA to a mitochondrial disease's cell with a A1555G mutation in the mitochondrial 12S rRNA gene results in an increase in mitochondrial respiratory activity. Mitochondrion 2020; 55:134-144. [PMID: 33035688 DOI: 10.1016/j.mito.2020.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/01/2020] [Accepted: 09/28/2020] [Indexed: 01/02/2023]
Abstract
We report on validating a mitochondrial gene therapeutic strategy using fibroblasts derived from patients with an A1555G point mutation in mitochondrial DNA coding 12S ribosomal RNA (rRNA (12S)). Wild-type rRNA (12S) as a therapeutic RNA was encapsulated in a mitochondrial targeting liposome, a MITO-Porter (rRNA-MITO-Porter), and an attempt was made to deliver the MITO-Porter to mitochondria of the diseased cells. It was confirmed that the rRNA-MITO-Porter treatment significantly decreased the ratio of the mutant rRNA content. Moreover, it was shown that the mitochondrial respiratory activities of the diseased cells were improved as the result of the mitochondrial transfection of the rRNA-MITO-Porter.
Collapse
|
14
|
Tanaka H, Sakurai Y, Anindita J, Akita H. Development of lipid-like materials for RNA delivery based on intracellular environment-responsive membrane destabilization and spontaneous collapse. Adv Drug Deliv Rev 2020; 154-155:210-226. [PMID: 32650040 DOI: 10.1016/j.addr.2020.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/26/2020] [Accepted: 07/02/2020] [Indexed: 01/01/2023]
Abstract
Messenger RNA and small interfering RNA are attractive modalities for curing diseases by complementation or knock-down of proteins. For success of these RNAs, a drug delivery system (DDS) is required to control a pharmacokinetics, to enhance cellular uptake, to overcome biological membranes, and to release the cargo into the cytoplasm. Based on past research, developing nanoparticles that are neutrally charged have been the mainstream of their development. Also, the materials are further mounted with pH- and/or reducing environment-responsive units. In this review, we summarize progress made in the molecular design of these materials. We also focus on the importance of the hydrophobic scaffold for tissue/cell targeting, intracellular trafficking, and immune responses. As a practical example, the design concept of the SS-cleavable and pH-activated lipid-like material (ssPalm) and subsequent molecular modification tailored to the RNA-based medical application is discussed.
Collapse
|
15
|
Pied N, Wodrich H. Imaging the adenovirus infection cycle. FEBS Lett 2019; 593:3419-3448. [PMID: 31758703 DOI: 10.1002/1873-3468.13690] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022]
Abstract
Incoming adenoviruses seize control of cytosolic transport mechanisms to relocate their genome from the cell periphery to specialized sites in the nucleoplasm. The nucleus is the site for viral gene expression, genome replication, and the production of progeny for the next round of infection. By taking control of the cell, adenoviruses also suppress cell-autonomous immunity responses. To succeed in their production cycle, adenoviruses rely on well-coordinated steps, facilitated by interactions between viral proteins and cellular factors. Interactions between virus and host can impose remarkable morphological changes in the infected cell. Imaging adenoviruses has tremendously influenced how we delineate individual steps in the viral life cycle, because it allowed the development of specific optical markers to label these morphological changes in space and time. As technology advances, innovative imaging techniques and novel tools for specimen labeling keep uncovering previously unseen facets of adenovirus biology emphasizing why imaging adenoviruses is as attractive today as it was in the past. This review will summarize past achievements and present developments in adenovirus imaging centered on fluorescence microscopy approaches.
Collapse
Affiliation(s)
- Noémie Pied
- CNRS UMR 5234, Microbiologie Fondamentale et Pathogénicité, Université de Bordeaux, France
| | - Harald Wodrich
- CNRS UMR 5234, Microbiologie Fondamentale et Pathogénicité, Université de Bordeaux, France
| |
Collapse
|
16
|
Dahiya U, Mishra S, Chattopadhyay S, Kumari A, Gangal A, Ganguli M. Role of Cellular Retention and Intracellular State in Controlling Gene Delivery Efficiency of Multiple Nonviral Carriers. ACS OMEGA 2019; 4:20547-20557. [PMID: 31858039 PMCID: PMC6906788 DOI: 10.1021/acsomega.9b02401] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/06/2019] [Indexed: 06/10/2023]
Abstract
Nonviral gene delivery has seen major progress in the last two decades owing to facile synthesis, low toxicity, and ease of modification of nanocarriers that take nucleic acids to cells and tissues. Gene delivery nanocomplexes need to reach the target locations in significant amounts by overcoming multiple barriers. While the importance of nanocomplex stability, cellular uptake, intracellular trafficking, and nuclear localization has been studied extensively, the role of cellular retention and recycling of these nanocomplexes is less understood in the context of gene delivery. In this study, we used different DNA carriers and made efforts to understand the role played by cellular retention in determining their gene delivery efficiency across multiple cell lines. In addition, we also analyzed whether state of complexation and localization of the nanocomplexes play a role in conjunction with cellular retention. We observed higher transfection efficiencies for nanocomplexes showing better retention, lower unpackaging, and low recycling. Our data also suggests that nanocomplexes made of peptides with terminal cysteine modification show enhanced retention and transfection efficiency compared to their counterparts with no terminal cysteine. Overall, the work highlights myriad of factors to be considered for improving gene delivery efficiency of nanocomplexes.
Collapse
Affiliation(s)
- Ujjwal
Ranjan Dahiya
- CSIR—Institute
of Genomics and Integrative Biology, Mathura Road, New Delhi 110020, India
- Academy
of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India
| | - Sarita Mishra
- CSIR—Institute
of Genomics and Integrative Biology, Mathura Road, New Delhi 110020, India
- Academy
of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India
| | | | - Anupama Kumari
- CSIR—Institute
of Genomics and Integrative Biology, Mathura Road, New Delhi 110020, India
- Academy
of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India
| | - Apurva Gangal
- CSIR—Institute
of Genomics and Integrative Biology, Mathura Road, New Delhi 110020, India
| | - Munia Ganguli
- CSIR—Institute
of Genomics and Integrative Biology, Mathura Road, New Delhi 110020, India
- Academy
of Scientific and Innovative Research (AcSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi 110001, India
| |
Collapse
|
17
|
Tateshita N, Miura N, Tanaka H, Masuda T, Ohtsuki S, Tange K, Nakai Y, Yoshioka H, Akita H. Development of a lipoplex-type mRNA carrier composed of an ionizable lipid with a vitamin E scaffold and the KALA peptide for use as an ex vivo dendritic cell-based cancer vaccine. J Control Release 2019; 310:36-46. [PMID: 31386869 DOI: 10.1016/j.jconrel.2019.08.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 12/31/2022]
Abstract
A dendritic cells (DCs)-based vaccine (DC-vaccine) system is an attractive technology for eliciting antigen-specific immune responses that can protect subjects from infectious diseases and for curing various types of cancers. For the insertion of a foreign antigen to DCs, the transfection of an antigen-coding mRNA to the cells is a promising approach. In order to introduce an antigen, a carrier for mRNA transfection is required, since the mRNA molecule per se is unstable in serum-containing medium. We previously reported on an ionizable lipid-like material with vitamin E-scaffolds (ssPalmE) as a material for a lipid nanoparticle (LNP)-based carrier for nucleic acids. In the present study, we report on the development of a lipoplex-type mRNA carrier for use as a DC-vaccine by using a combination of an ssPalmE-LNP and an α-helical cationic peptide "KALA" (ssPalmE-KALA). The transfection of mRNAs complexed with the ssPalmE-KALA achieved a significantly higher protein expression and the production of proinflammatory cytokines from murine bone marrow derived DCs (BMDCs) in comparison with a lipoplex that was prepared with an ssPalm with fatty acid-scaffolds (myristic acid; ssPalmM-KALA). A cellular uptake process and a pH-responsive membrane-destabilization activity cannot explain the preferred protein expression and immune-stimulation caused by the ssPalmE-KALA. Proteomic analyses suggest that transfection with the ssPalmM-KALA stimulates a down-regulatory pathway of translation, while the transfection with the ssPalmE-KALA does not stimulate it. In the vaccination with the BMDCs that were preliminarily transfected with an ovalbumin (OVA)-encoding mRNA elicited the induction OVA specific cytotoxic T-lymphocyte activity in vivo. In parallel, the vaccination induced significant prophylactic anti-tumor effects against a model tumor that stably expressed the OVA protein. Based on the above findings, the ssPalmE-KALA appears to be a potent ex vivo DCs-based RNA vaccine platform.
Collapse
Affiliation(s)
- Naho Tateshita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba, Japan
| | - Naoya Miura
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba, Japan
| | - Hiroki Tanaka
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba, Japan.
| | - Takeshi Masuda
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto City, Kumamoto, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto City, Kumamoto, Japan
| | - Kota Tange
- DDS Research Laboratory, DDS Development Division, NOF Corporation, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki 210-0865, Japan
| | - Yuta Nakai
- DDS Research Laboratory, DDS Development Division, NOF Corporation, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki 210-0865, Japan
| | - Hiroki Yoshioka
- DDS Research Laboratory, DDS Development Division, NOF Corporation, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki 210-0865, Japan
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba, Japan.
| |
Collapse
|
18
|
Liu H, Yang Z, Xun Z, Gao Z, Sun Y, Yu J, Yang T, Zhao X, Cai C, Ding P. Nuclear delivery of plasmid DNA determines the efficiency of gene expression. Cell Biol Int 2019; 43:789-798. [PMID: 31042002 DOI: 10.1002/cbin.11155] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 04/24/2019] [Accepted: 04/29/2019] [Indexed: 01/21/2023]
Abstract
As a cationic non-viral gene delivery vector, poly(agmatine/ N, N'-cystamine-bis-acrylamide) (AGM-CBA) showed significantly higher plasmid DNA (pDNA) transfection ability than polyethylenimine (PEI) in NIH/3T3 cells. The transfection expression of AGM-CBA/pDNA polyplexes was found to have a non-linear relationship with AGM-CBA/pDNA weight ratios. To further investigate the mechanism involved in the transfection process of poly(AGM-CBA), we used pGL3-control luciferase reporter gene (pLUC) as a reporter pDNA in this study. The distribution of pLUC in NIH/3T3 cells and nuclei after AGM-CBA/pLUC and PEI/pLUC transfection were determined by quantitative polymerase chain reaction (qPCR) analysis. The intracellular trafficking of the polyplexes was evaluated by cellular uptake and nuclei delivery of pLUC, and the intracellular availability was evaluated by the ratio of transfection expression to the numbers of pLUC delivered in nuclei. It was found that pLUC intracellular trafficking did not have any correlation with the transfection expression, while an excellent correlation was found between the nuclei pLUC availability and transfection expression. These results suggested that the intracellular availability of pLUC in nuclei was the rate-limiting step for pLUC transfection expression. Further optimization of the non-viral gene delivery system can be focused on the improvement of gene intracellular availability.
Collapse
Affiliation(s)
- Hui Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Zhen Yang
- Research and development Center, CR Sanjiu Medical & Pharmaceutical Co., Ltd, Shenzhen, 518110, China
| | - Zhe Xun
- Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, 110122, China
| | - Zihao Gao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yanping Sun
- School of Pharmacy, Hebei University of Science and Technology, Shijiazhuang, 050018, China
| | - Jiankun Yu
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME, 04401, USA
| | - Xiaoyun Zhao
- Department of Microbiology and Cell Biology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Cuifang Cai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| |
Collapse
|
19
|
Low Electric Treatment activates Rho GTPase via Heat Shock Protein 90 and Protein Kinase C for Intracellular Delivery of siRNA. Sci Rep 2019; 9:4114. [PMID: 30858501 PMCID: PMC6412017 DOI: 10.1038/s41598-019-40904-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 01/28/2019] [Indexed: 11/08/2022] Open
Abstract
Low electric treatment (LET) promotes intracellular delivery of naked siRNA by altering cellular physiology. However, which signaling molecules and cellular events contribute to LET-mediated siRNA uptake are unclear. Here, we used isobaric tags in relative and absolute quantification (iTRAQ) proteomic analysis to identify changes in the levels of phosphorylated proteins that occur during cellular uptake of siRNA promoted by LET. iTRAQ analysis revealed that heat shock protein 90 (Hsp90)α and myristoylated alanine-rich C-kinase substrate (Marcks) were highly phosphorylated following LET of NIH 3T3 cells, but not untreated cells. Furthermore, the levels of phosphorylated Hsp90α and protein kinase C (PKC)γ were increased by LET both with siRNA and liposomes having various physicochemical properties used as model macromolecules, suggesting that PKCγ activated partly by Ca2+ influx as well as Hsp90 chaperone function were involved in LET-mediated cellular siRNA uptake. Furthermore, LET with siRNA induced activation of Rho GTPase via Hsp90 and PKC, which could contribute to cellular siRNA uptake accompanied by actin cytoskeleton remodeling. Collectively, our results suggested that LET-induced Rho GTPase activation via Hsp90 and PKC would participate in actin-dependent cellular uptake of siRNA.
Collapse
|
20
|
Ito T, Okuda T, Takashima Y, Okamoto H. Naked pDNA Inhalation Powder Composed of Hyaluronic Acid Exhibits High Gene Expression in the Lungs. Mol Pharm 2019; 16:489-497. [PMID: 30092131 DOI: 10.1021/acs.molpharmaceut.8b00502] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Gene therapy is a breakthrough treatment strategy against several intractable and lethal diseases that previously lacked established treatments. Viral and nonviral vectors have been studied to realize higher gene transfection efficiencies and to suppress the degradation of gene by nucleolytic enzymes in vivo. However, it is often the case that the addition of a vector results in adverse effects. In this study, we identified formulations of dry naked plasmid DNA (pDNA) powders with no vector showing significantly higher gene expression than pDNA solutions including vectors such as polyethylenimine (PEI) in the lungs of mice. We prepared the naked pDNA powders by spray-freeze-drying with various excipients. The gene expression of naked pDNA powders exceeded those of pDNA solutions containing PEI, naked pDNA solution, and reconstituted pDNA powder. Gene expression of each naked pDNA powder was dependent on the composition of excipients. Among them, the mice that were administered the pDNA powder composed of low-molecular-weight hyaluronic acid (LHA) as an excipient showed the highest gene expression. The lactate dehydrogenase activity and concentration of inflammatory cytokines in bronchoalveolar lavage fluid were comparable to those caused by ultrapure water. The results suggest that useful dry naked nucleic acid powders for inhalation could be created by optimizing the excipients, offering new insights into the development of pulmonary gene therapy.
Collapse
Affiliation(s)
- Takaaki Ito
- Department of Drug Delivery Research, Faculty of Pharmacy , Meijo University , 150 Yagotoyama , Tempaku-ku, Nagoya 468-8503 , Japan
| | - Tomoyuki Okuda
- Department of Drug Delivery Research, Faculty of Pharmacy , Meijo University , 150 Yagotoyama , Tempaku-ku, Nagoya 468-8503 , Japan
| | - Yoshimasa Takashima
- Department of Drug Delivery Research, Faculty of Pharmacy , Meijo University , 150 Yagotoyama , Tempaku-ku, Nagoya 468-8503 , Japan
| | - Hirokazu Okamoto
- Department of Drug Delivery Research, Faculty of Pharmacy , Meijo University , 150 Yagotoyama , Tempaku-ku, Nagoya 468-8503 , Japan
| |
Collapse
|
21
|
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: 780] [Impact Index Per Article: 130.0] [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
|
22
|
Togashi R, Tanaka H, Nakamura S, Yokota H, Tange K, Nakai Y, Yoshioka H, Harashima H, Akita H. A hepatic pDNA delivery system based on an intracellular environment sensitive vitamin E-scaffold lipid-like material with the aid of an anti-inflammatory drug. J Control Release 2018; 279:262-270. [DOI: 10.1016/j.jconrel.2018.04.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/29/2018] [Accepted: 04/11/2018] [Indexed: 12/16/2022]
|
23
|
Tanaka H, Nakatani T, Furihata T, Tange K, Nakai Y, Yoshioka H, Harashima H, Akita H. In Vivo Introduction of mRNA Encapsulated in Lipid Nanoparticles to Brain Neuronal Cells and Astrocytes via Intracerebroventricular Administration. Mol Pharm 2018; 15:2060-2067. [DOI: 10.1021/acs.molpharmaceut.7b01084] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Hiroki Tanaka
- Graduate School of Pharmaceutical Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba City, Chiba 260-8675, Japan
| | - Taichi Nakatani
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Sapporo City, Hokkaido 060-0812, Japan
| | - Tomomi Furihata
- Department of Pharmacology, Graduate School of Medicine, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba City, Chiba 260-8670, Japan
| | - Kota Tange
- NOF CORPORATION, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki City, Kanagawa 210-0865, Japan
| | - Yuta Nakai
- NOF CORPORATION, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki City, Kanagawa 210-0865, Japan
| | - Hiroki Yoshioka
- NOF CORPORATION, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki City, Kanagawa 210-0865, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Sapporo City, Hokkaido 060-0812, Japan
| | - Hidetaka Akita
- Graduate School of Pharmaceutical Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba City, Chiba 260-8675, Japan
| |
Collapse
|
24
|
Identification and Evaluation of the Minimum Unit of a KALA Peptide Required for Gene Delivery and Immune Activation. J Pharm Sci 2017; 106:3113-3119. [DOI: 10.1016/j.xphs.2017.05.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 04/26/2017] [Accepted: 05/16/2017] [Indexed: 01/27/2023]
|
25
|
Yang Z, Sun Y, Xian L, Xun Z, Yu J, Yang T, Zhao X, Cai C, Wang D, Ding P. Disulfide‐bond‐containing agamatine‐cystaminebisacrylamide polymer demonstrates better transfection efficiency and lower cytotoxicity than polyethylenimine in NIH/3T3 cells. J Cell Biochem 2017; 119:1767-1779. [DOI: 10.1002/jcb.26338] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/08/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Zhen Yang
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
| | - Yanping Sun
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
| | - Lei Xian
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
| | - Zhe Xun
- Institute of Metabolic Disease Research and Drug DevelopmentChina Medical UniversityShenyangChina
| | - Jiankun Yu
- Department of Ion Channel PharmacologySchool of PharmacyChina Medical UniversityShenyangChina
| | - Tianzhi Yang
- Department of Basic Pharmaceutical SciencesSchool of PharmacyHusson UniversityBangorMaine
| | - Xiaoyun Zhao
- Department of Microbiology and Cell BiologySchool of Life Science and BiopharmaceuticsShenyang Pharmaceutical UniversityShenyangChina
| | - Cuifang Cai
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
| | - Dongkai Wang
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
| | - Pingtian Ding
- School of PharmacyShenyang Pharmaceutical UniversityShenyangChina
| |
Collapse
|
26
|
Zhou Z, Liu X, Zhu D, Wang Y, Zhang Z, Zhou X, Qiu N, Chen X, Shen Y. Nonviral cancer gene therapy: Delivery cascade and vector nanoproperty integration. Adv Drug Deliv Rev 2017; 115:115-154. [PMID: 28778715 DOI: 10.1016/j.addr.2017.07.021] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 02/07/2023]
Abstract
Gene therapy represents a promising cancer treatment featuring high efficacy and limited side effects, but it is stymied by a lack of safe and efficient gene-delivery vectors. Cationic polymers and lipid-based nonviral gene vectors have many advantages and have been extensively explored for cancer gene delivery, but their low gene-expression efficiencies relative to viral vectors limit their clinical translations. Great efforts have thus been devoted to developing new carrier materials and fabricating functional vectors aimed at improving gene expression, but the overall efficiencies are still more or less at the same level. This review analyzes the cancer gene-delivery cascade and the barriers, the needed nanoproperties and the current strategies for overcoming these barriers, and outlines PEGylation, surface-charge, size, and stability dilemmas in vector nanoproperties to efficiently accomplish the cancer gene-delivery cascade. Stability, surface, and size transitions (3S Transitions) are proposed to resolve those dilemmas and strategies to realize these transitions are comprehensively summarized. The review concludes with a discussion of the future research directions to design high-performance nonviral gene vectors.
Collapse
Affiliation(s)
- Zhuxian Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xiangrui Liu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Dingcheng Zhu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Yue Wang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Zhen Zhang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xuefei Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Nasha Qiu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xuesi Chen
- Changchun Institute of Applied Chemistry, Key Lab of Polymer Ecomaterials, Changchun, China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China.
| |
Collapse
|
27
|
Ni R, Zhou J, Hossain N, Chau Y. Virus-inspired nucleic acid delivery system: Linking virus and viral mimicry. Adv Drug Deliv Rev 2016; 106:3-26. [PMID: 27473931 DOI: 10.1016/j.addr.2016.07.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 07/02/2016] [Accepted: 07/20/2016] [Indexed: 12/21/2022]
Abstract
Targeted delivery of nucleic acids into disease sites of human body has been attempted for decades, but both viral and non-viral vectors are yet to meet our expectations. Safety concerns and low delivery efficiency are the main limitations of viral and non-viral vectors, respectively. The structure of viruses is both ordered and dynamic, and is believed to be the key for effective transfection. Detailed understanding of the physical properties of viruses, their interaction with cellular components, and responses towards cellular environments leading to transfection would inspire the development of safe and effective non-viral vectors. To this goal, this review systematically summarizes distinctive features of viruses that are implied for efficient nucleic acid delivery but not yet fully explored in current non-viral vectors. The assembly and disassembly of viral structures, presentation of viral ligands, and the subcellular targeting of viruses are emphasized. Moreover, we describe the current development of cationic material-based viral mimicry (CVM) and structural viral mimicry (SVM) in these aspects. In light of the discrepancy, we identify future opportunities for rational design of viral mimics for the efficient delivery of DNA and RNA.
Collapse
Affiliation(s)
- Rong Ni
- Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Institute for Advanced Study, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Junli Zhou
- Department of Chemical and Biomolecular Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Naushad Hossain
- Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ying Chau
- Department of Chemical and Biomolecular Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
| |
Collapse
|
28
|
Sato Y, Sakurai Y, Kajimoto K, Nakamura T, Yamada Y, Akita H, Harashima H. Innovative Technologies in Nanomedicines: From Passive Targeting to Active Targeting/From Controlled Pharmacokinetics to Controlled Intracellular Pharmacokinetics. Macromol Biosci 2016; 17. [DOI: 10.1002/mabi.201600179] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/19/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Yusuke Sato
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Yu Sakurai
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Kazuaki Kajimoto
- Health Research Institute; National Institute of Advanced Industrial Science and Technology (AIST); 2217-14 Hayashi-cho Takamatsu, Kagawa 761-0395 Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Hidetaka Akita
- Graduate School of Pharmaceutical Sciences; Chiba University; 1-8-1 Inohana Chuo-ku, Chiba-shi, Chiba 260-8675 Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| |
Collapse
|
29
|
Abstract
AIM Viral vectors are used commonly in gene therapy trials, but their potential toxic effects are a serious concern. Identification of highly efficient nonviral vectors may alleviate these effects. Results & methodology: We compared the abilities of TransfeX, TransIT-LT1 and adenovirus to deliver the firefly luciferase and green fluorescent protein genes into HeLa cervical carcinoma, and HSC-3 and H357 oral squamous cell carcinoma cells. TransfeX mediated fourfold higher gene expression in HeLa cells than adenovirus, even at the highest multiplicity of infection. Flow cytometry indicated that a population of transfected cells expresses higher levels of green fluorescent protein than transduced cells. CONCLUSION TransfeX may be useful for gene therapy applications, particularly where the use of adenovirus is contraindicated.
Collapse
|
30
|
Hasan M, Tarashima N, Fujikawa K, Ohgita T, Hama S, Tanaka T, Saito H, Minakawa N, Kogure K. The novel functional nucleic acid iRed effectively regulates target genes following cytoplasmic delivery by faint electric treatment. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2016; 17:554-562. [PMID: 27877903 PMCID: PMC5111564 DOI: 10.1080/14686996.2016.1221726] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 08/02/2016] [Accepted: 08/04/2016] [Indexed: 06/06/2023]
Abstract
An intelligent shRNA expression device (iRed) contains the minimum essential components needed for shRNA production in cells, and could be a novel tool to regulate target genes. However, general delivery carriers consisting of cationic polymers/lipids could impede function of a newly generated shRNA via electrostatic interaction in the cytoplasm. Recently, we found that faint electric treatment (fET) of cells enhanced delivery of siRNA and functional nucleic acids into the cytoplasm in the absence of delivery carriers. Here, we examined fET of cells stably expressing luciferase in the presence of iRed encoding anti-luciferase shRNA. Transfection of lipofectamine 2000 (LFN)/iRed lipoplexes showed an RNAi effect, but fET-mediated iRed transfection did not, likely because of the endosomal localization of iRed after delivery. However, fET in the presence of lysosomotropic agent chloroquine significantly improved the RNAi effect of iRed/fET to levels that were higher than those for the LFN/iRed lipoplexes. Furthermore, the amount of lipid droplets in adipocytes significantly decreased following fET with iRed against resistin in the presence of chloroquine. Thus, iRed could be a useful tool to regulate target genes following fET-mediated cytoplasmic delivery with endosomal escape devices.
Collapse
Affiliation(s)
- Mahadi Hasan
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Noriko Tarashima
- Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Koki Fujikawa
- Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takashi Ohgita
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Susumu Hama
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Tamotsu Tanaka
- Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hiroyuki Saito
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Noriaki Minakawa
- Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kentaro Kogure
- Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
31
|
Cai J, Yue Y, Wang Y, Jin Z, Jin F, Wu C. Quantitative study of effects of free cationic chains on gene transfection in different intracellular stages. J Control Release 2016; 238:71-79. [PMID: 27448443 DOI: 10.1016/j.jconrel.2016.07.031] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/08/2016] [Accepted: 07/19/2016] [Indexed: 12/12/2022]
Abstract
Previously, we revealed that in the application of using cationic polymer chains, polyethylenimine (PEI), to condense anionic plasmid DNA chains (pDNA) to form the DNA/polymer polyplexes, after all the pDNAs are complexed with PEI, further added PEIs exist individual chains and free in the solution mixture. It is those uncomplexed polycation chains that dramatically promote the gene transfection. In the current study, we studied how those free cationic chains with different lengths and topologies affect the intracellular trafficking of the polyplexes, the translocation of pDNA through the nuclear membrane, the transcription of pDNA to mRNA and the translocation of mRNA from nucleus to cytosol in HepG2 cells by using a combination of the three-dimensional confocal microscope and TaqMan real-time PCR. We found that free branched PEI chains with a molar mass of 25,000g/mol and a total concentration of 1.8×10(-6)g/mL promote the overall gene transfection efficiency by a factor of ~500 times. Our results quantitatively reveal that free chains help little in the cellular uptake, but clearly reduce the lysosomal entrapment of those internalized polyplexes (2-3 folds); assist the translocation of pDNA through nuclear membrane after it is released from the polyplexes in the cytosol (~5 folds); enhance the pDNA-to-mRNA transcription efficiency (~4 folds); and facilitate the nucleus-to-cytosol translocation of mRNA (7-8 folds). The total enhancement of those steps agrees well with the overall efficiency, demonstrating, for the first time, how free cationic polymer chains quantitatively promote the gene transfection in each step in the intracellular space.
Collapse
Affiliation(s)
- Jinge Cai
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong.
| | - Yanan Yue
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| | - Yanjing Wang
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| | - Zhenyu Jin
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Fan Jin
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Chi Wu
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong; Hefei National Laboratory of Physical Science at the Microscale, Department of Chemical Physics, University of Science and Technology of China, Hefei, Anhui 230026, China.
| |
Collapse
|
32
|
Pan J, Yuan Y, Wang H, Liu F, Xiong X, Chen H, Yuan L. Efficient Transfection by Using PDMAEMA-Modified SiNWAs as a Platform for Ca(2+)-Dependent Gene Delivery. ACS APPLIED MATERIALS & INTERFACES 2016; 8:15138-15144. [PMID: 27249181 DOI: 10.1021/acsami.6b04689] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The major bottleneck for gene delivery lies in the lack of safe and efficient gene vectors and delivery systems. In order to develop a much safer and efficient transfection system, a novel strategy of combining traditional Ca(2+)-dependent transfection with cationic polymer poly(N,N-dimethylamino)ethyl methacrylate (PDMAEMA) modified silicon nanowire arrays (SiNWAs) was proposed in this work. Detailed studies were carried out on the effects of the PDMAEMA polymerization time, the Ca(2+) concentration, and the incubation time of Ca(2+)@DNA complex with PDMAEMA-modified SiNWAs (SN-PDM) on the gene transfection in the cells. The results demonstrated that the transfection efficiency of SN-PDM assisted traditional Ca(2+)-dependent transfection was significantly enhanced compared to those without any surface assistance, and SN-PDM with polymerization time 24 h exhibited the highest efficiency. Moreover, the optimal transfection efficiency was found at the system of a complex containing Ca(2+) (100 mM) and plasmid DNA (pDNA) incubated on SN-PDM for 20 min. Compared with unmodified SiNWAs, SN-PDM has little cytotoxicity and can improve cell attachment. All of these results demonstrated that SN-PDM could significantly enhance Ca(2+)-dependent transfection; this process depends on the amino groups' density of PDMAEMA on the surface, the Ca(2+) concentration, and the available Ca(2+)@DNA complex. Our study provides a potential novel and excellent means of gene delivery for therapeutic applications.
Collapse
Affiliation(s)
- Jingjing Pan
- College of Chemistry, Chemical Engineering, and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Yuqi Yuan
- College of Chemistry, Chemical Engineering, and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Hongwei Wang
- College of Chemistry, Chemical Engineering, and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Feng Liu
- College of Chemistry, Chemical Engineering, and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Xinhong Xiong
- College of Chemistry, Chemical Engineering, and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Hong Chen
- College of Chemistry, Chemical Engineering, and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| | - Lin Yuan
- College of Chemistry, Chemical Engineering, and Materials Science, Soochow University , Suzhou 215123, People's Republic of China
| |
Collapse
|
33
|
Itakura S, Hama S, Matsui R, Kogure K. Effective cytoplasmic release of siRNA from liposomal carriers by controlling the electrostatic interaction of siRNA with a charge-invertible peptide, in response to cytoplasmic pH. NANOSCALE 2016; 8:10649-10658. [PMID: 27145993 DOI: 10.1039/c5nr08365f] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Condensing siRNA with cationic polymers is a major strategy used in the development of siRNA carriers that can avoid degradation by nucleases and achieve effective delivery of siRNA into the cytoplasm. However, ineffective release of siRNA from such condensed forms into the cytoplasm is a limiting step for induction of RNAi effects, and can be attributed to tight condensation of siRNA with the cationic polymers, due to potent electrostatic interactions. Here, we report that siRNA condensed with a slightly acidic pH-sensitive peptide (SAPSP), whose total charge is inverted from positive to negative in response to cytoplasmic pH, is effectively released via electrostatic repulsion of siRNA with negatively charged SAPSP at cytoplasmic pH (7.4). The condensed complex of siRNA and positively-charged SAPSP at acidic pH (siRNA/SAPSP) was found to result in almost complete release of siRNA upon charge inversion of SAPSP at pH 7.4, with the resultant negatively-charged SAPSP having no undesirable interactions with endogenous mRNA. Moreover, liposomes encapsulating siRNA/SAPSP demonstrated knockdown efficiencies comparable to those of commercially available siRNA carriers. Taken together, SAPSP may be very useful as a siRNA condenser, as it facilitates effective cytoplasmic release of siRNA, and subsequent induction of specific RNAi effects.
Collapse
Affiliation(s)
- Shoko Itakura
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan.
| | - Susumu Hama
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan.
| | - Ryo Matsui
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan.
| | - Kentaro Kogure
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan.
| |
Collapse
|
34
|
Tanaka H, Sato Y, Harashima H, Akita H. Cellular environment-responsive nanomaterials for use in gene and siRNA delivery: molecular design for biomembrane destabilization and intracellular collapse. Expert Opin Drug Deliv 2016; 13:1015-27. [DOI: 10.1517/17425247.2016.1154531] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Hiroki Tanaka
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | | | - Hidetaka Akita
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
35
|
Pyykkö I, Zou J, Schrott-Fischer A, Glueckert R, Kinnunen P. An Overview of Nanoparticle Based Delivery for Treatment of Inner Ear Disorders. Methods Mol Biol 2016; 1427:363-415. [PMID: 27259938 DOI: 10.1007/978-1-4939-3615-1_21] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nanoparticles offer new possibilities for inner ear treatment as they can carry a variety of drugs, protein, and nucleic acids to inner ear. Nanoparticles are equipped with several functions such as targetability, immuno-transparency, biochemical stability, and ability to be visualized in vivo and in vitro. A group of novel peptides can be attached to the surface of nanoparticles that will enhance the cell entry, endosomal escape, and nuclear targeting. Eight different types of nanoparticles with different payload carrying strategies are available now. The transtympanic delivery of nanoparticles indicates that, depending on the type of nanoparticle, different migration pathways into the inner ear can be employed, and that optimal carriers can be designed according to the intended cargo. The use of nanoparticles as drug/gene carriers is especially attractive in conjunction with cochlear implantation or even as an inclusion in the implant as a drug/gene reservoir.
Collapse
Affiliation(s)
- Ilmari Pyykkö
- Department of Otolaryngology, University of Tampere and University Hospital of Tampere, Tampere, 33014, Finland. .,Hearing and Balance Research Unit, Field of Otolaryngology, School of Medicine, University of Tampere, Medisiinarinkatu 3, Tampere, 33520, Finland.
| | - Jing Zou
- BECS, Department of Biomedical Engineering and Computational Science, Aalto University, Aalto, 02150, Espoo, Finland
| | - Annelies Schrott-Fischer
- Department of Otolaryngology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria
| | - Rudolf Glueckert
- Department of Otolaryngology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria
| | - Paavo Kinnunen
- BECS, Department of Biomedical Engineering and Computational Science, Aalto University, Aalto, Finland
| |
Collapse
|
36
|
Martin TM, Plautz SA, Pannier AK. Temporal endogenous gene expression profiles in response to lipid-mediated transfection. J Gene Med 2015; 17:14-32. [PMID: 25663588 DOI: 10.1002/jgm.2821] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/01/2015] [Accepted: 02/03/2015] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Design of efficient nonviral gene delivery systems is limited as a result of the rudimentary understanding of the specific molecules and processes that facilitate DNA transfer. METHODS Lipoplexes formed with Lipofectamine 2000 (LF2000) and plasmid-encoding green fluorescent protein (GFP) were delivered to the HEK 293T cell line. After treating cells with lipoplexes, HG-U133 Affymetrix microarrays were used to identify endogenous genes differentially expressed between treated and untreated cells (2 h exposure) or between flow-separated transfected cells (GFP+) and treated, untransfected cells (GFP-) at 8, 16 and 24 h after lipoplex treatment. Cell priming studies were conducted using pharmacologic agents to alter endogenous levels of the identified differentially expressed genes to determine effect on transfection levels. RESULTS Relative to untreated cells 2 h after lipoplex treatment, only downregulated genes were identified ≥ 30-fold: ALMS1, ITGB1, FCGR3A, DOCK10 and ZDDHC13. Subsequently, relative to GFP- cells, the GFP+ cell population showed at least a five-fold upregulation of RAP1A and PACSIN3 (8 h) or HSPA6 and RAP1A (16 and 24 h). Pharmacologic studies altering endogenous levels for ALMS1, FCGR3A, and DOCK10 (involved in filopodia protrusions), ITGB1 (integrin signaling), ZDDHC13 (membrane trafficking) and PACSIN3 (proteolytic shedding of membrane receptors) were able to increase or decrease transgene production. CONCLUSIONS RAP1A, PACSIN3 and HSPA6 may help lipoplex-treated cells overcome a transcriptional shutdown due to treatment with lipoplexes and provide new targets for investigating molecular mechanisms of transfection or for enhancing transfection through cell priming or engineering of the nonviral gene delivery system.
Collapse
Affiliation(s)
- Timothy M Martin
- Department of Pharmaceutical Sciences, Durham Research Center II, University of Nebraska-Medical Center, Omaha, NE, USA
| | | | | |
Collapse
|
37
|
Martin TM, Plautz SA, Pannier AK. Temporal endogenous gene expression profiles in response to polymer-mediated transfection and profile comparison to lipid-mediated transfection. J Gene Med 2015; 17:33-53. [PMID: 25663627 DOI: 10.1002/jgm.2822] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/01/2015] [Accepted: 02/03/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Design of efficient nonviral gene delivery systems is limited by the rudimentary understanding of specific molecules that facilitate transfection. METHODS Polyplexes using 25-kDa polyethylenimine (PEI) and plasmid-encoding green fluorescent protein (GFP) were delivered to HEK 293T cells. After treating cells with polyplexes, microarrays were used to identify endogenous genes differentially expressed between treated and untreated cells (2 h of exposure) or between flow-separated transfected cells (GFP+) and treated, untransfected cells (GFP-) at 8, 16 and 24 h after lipoplex treatment. Cell priming studies were conducted using pharmacologic agents to alter endogenous levels of the identified differentially expressed genes to determine effect on transfection levels. Differentially expressed genes in polyplex-mediated transfection were compared with those differentially expressed in lipoplex transfection to identify DNA carrier-dependent molecular factors. RESULTS Differentially expressed genes were RGS1, ARHGAP24, PDZD2, SNX24, GSN and IGF2BP1 after 2 h; RAP1A and ACTA1 after 8 h; RAP1A, WDR78 and ACTA1 after 16 h; and RAP1A, SCG5, ATF3, IREB2 and ACTA1 after 24 h. Pharmacologic studies altering endogenous levels for ARHGAP24, GSN, IGF2BP1, PDZD2 and RGS1 were able to increase or decrease transgene production. Comparing differentially expressed genes for polyplexes and lipoplexes, no common genes were identified at the 2-h time point, whereas, after the 8-h time point, RAP1A, ATF3 and HSPA6 were similarly expressed. SCG5 and PGAP1 were only upregulated in polyplex-transfected cells. CONCLUSIONS The identified genes and pharmacologic agents provide targets for improving transfection systems, although polyplex or lipoplex dependencies must be considered.
Collapse
Affiliation(s)
- Timothy M Martin
- Department of Pharmaceutical Sciences, Durham Research Center II, University of Nebraska-Medical Center, Omaha, NE, USA
| | | | | |
Collapse
|
38
|
Glucocorticoid Cell Priming Enhances Transfection Outcomes in Adult Human Mesenchymal Stem Cells. Mol Ther 2015; 24:331-341. [PMID: 26478250 DOI: 10.1038/mt.2015.195] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 10/12/2015] [Indexed: 12/11/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) are one of the most widely researched stem cell types with broad applications from basic research to therapeutics, the majority of which require introduction of exogenous DNA. However, safety and scalability issues hinder viral delivery, while poor efficiency hinders nonviral gene delivery, particularly to hMSCs. Here, we present the use of a pharmacologic agent (glucocorticoid) to overcome barriers to hMSC DNA transfer to enhance transfection using three common nonviral vectors. Glucocorticoid priming significantly enhances transfection in hMSCs, demonstrated by a 3-fold increase in efficiency, 4-15-fold increase in transgene expression, and prolonged transgene expression when compared to transfection without glucocorticoids. These effects are dependent on glucocorticoid receptor binding and caused in part by maintenance of normal metabolic function and increased cellular (5-fold) and nuclear (6-10-fold) DNA uptake over hMSCs transfected without glucocorticoids. Results were consistent across five human donors and in cells up to passage five. Glucocorticoid cell priming is a simple and effective technique to significantly enhance nonviral transfection of hMSCs that should enhance their clinical use, accelerate new research, and decrease reliance on early passage cells.
Collapse
|
39
|
Sato Y, Nakamura T, Yamada Y, Akita H, Harashima H. Multifunctional enveloped nanodevices (MENDs). ADVANCES IN GENETICS 2015; 88:139-204. [PMID: 25409606 DOI: 10.1016/b978-0-12-800148-6.00006-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
It is anticipated that nucleic acid medicines will be in widespread use in the future, since they have the potential to cure diseases based on molecular mechanisms at the level of gene expression. However, intelligent delivery systems are required to achieve nucleic acid therapy, since they can perform their function only when they reach the intracellular site of action. We have been developing a multifunctional envelope-type nanodevice abbreviated as MEND, which consists of functional nucleic acids as a core and lipid envelope, and can control not only biodistribution but also the intracellular trafficking of nucleic acids. In this chapter, we review the development and evolution of the MEND by providing several successful examples, including the R8-MEND, the KALA-MEND, the MITO-Porter, the YSK-MEND, and the PALM.
Collapse
Affiliation(s)
- Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo City, Hokkaido, Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo City, Hokkaido, Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo City, Hokkaido, Japan
| | - Hidetaka Akita
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo City, Hokkaido, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo City, Hokkaido, Japan
| |
Collapse
|
40
|
Rafiee A, Riazi-Rad F, Alimohammadian MH, Gazori T, Fatemi SMR, Havaskary M. Hydrogel nanoparticle encapsulated plasmid as a suitable gene delivery system. CYTOL GENET+ 2015. [DOI: 10.3103/s0095452715020097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
41
|
Martin TM, Wysocki BJ, Beyersdorf JP, Wysocki TA, Pannier AK. Integrating mitosis, toxicity, and transgene expression in a telecommunications packet-switched network model of lipoplex-mediated gene delivery. Biotechnol Bioeng 2015; 111:1659-71. [PMID: 25097912 DOI: 10.1002/bit.25207] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Gene delivery systems transport exogenous genetic information to cells or biological systems with the potential to directly alter endogenous gene expression and behavior with applications in functional genomics, tissue engineering, medical devices, and gene therapy. Nonviral systems offer advantages over viral systems because of their low immunogenicity, inexpensive synthesis, and easy modification but suffer from lower transfection levels. The representation of gene transfer using models offers perspective and interpretation of complex cellular mechanisms,including nonviral gene delivery where exact mechanisms are unknown. Here, we introduce a novel telecommunications model of the nonviral gene delivery process in which the delivery of the gene to a cell is synonymous with delivery of a packet of information to a destination computer within a packet-switched computer network. Such a model uses nodes and layers to simplify the complexity of modeling the transfection process and to overcome several challenges of existing models. These challenges include a limited scope and limited time frame, which often does not incorporate biological effects known to affect transfection. The telecommunication model was constructed in MATLAB to model lipoplex delivery of the gene encoding the green fluorescent protein to HeLa cells. Mitosis and toxicity events were included in the model resulting in simulation outputs of nuclear internalization and transfection efficiency that correlated with experimental data. A priori predictions based on model sensitivity analysis suggest that increasing endosomal escape and decreasing lysosomal degradation, protein degradation, and GFP-induced toxicity can improve transfection efficiency by three-fold. Application of the telecommunications model to nonviral gene delivery offers insight into the development of new gene delivery systems with therapeutically relevant transfection levels.
Collapse
|
42
|
Martin TM, Wysocki BJ, Wysocki TA, Pannier AK. Identifying Intracellular pDNA Losses From a Model of Nonviral Gene Delivery. IEEE Trans Nanobioscience 2015; 14:455-464. [PMID: 25622323 DOI: 10.1109/tnb.2015.2392777] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Nonviral gene delivery systems are a type of nanocommunication system that transmit plasmid packets (i.e., pDNA packets) that are programmed at the nanoscale to biological systems at the microscopic cellular level. This engineered nanocommunication system suffers large pDNA losses during transmission of the genetically encoded information, preventing its use in biotechnological and medical applications. The pDNA losses largely remain uncharacterized, and the ramifications of reducing pDNA loss from newly designed gene delivery systems remain difficult to predict. Here, the pDNA losses during primary and secondary transmission chains were identified utilizing a MATLAB model employing queuing theory simulating delivery of pEGFPLuc transgene to HeLa cells carried by Lipofectamine 2000 nonviral DNA carrier. Minimizing pDNA loss during endosomal escape of the primary transmission process results in increased number of pDNA in the nucleus with increased transfection, but with increased probability of cell death. The number of pDNA copies in the nucleus and the amount of time the pDNAs are in the nucleus directly correlates to improved transfection efficiency. During secondary transmission, pDNAs are degraded during distribution to daughter cells. Reducing pDNA losses improves transfection, but a balance in quantity of nuclear pDNA, mitosis, and toxicity must be considered in order to achieve therapeutically relevant transfection levels.
Collapse
|
43
|
Akita H, Ishiba R, Togashi R, Tange K, Nakai Y, Hatakeyama H, Harashima H. A neutral lipid envelope-type nanoparticle composed of a pH-activated and vitamin E-scaffold lipid-like material as a platform for a gene carrier targeting renal cell carcinoma. J Control Release 2014; 200:97-105. [PMID: 25543000 DOI: 10.1016/j.jconrel.2014.12.029] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 11/07/2014] [Accepted: 12/23/2014] [Indexed: 12/21/2022]
Abstract
A renal cell carcinoma (RCC) is one of the refractory tumors, since it readily acquires resistance against chemotherapy. Thus, alternative therapeutic approaches such as obstructing the neovasculature are needed. We previously reported on the development of a plasmid DNA (pDNA)-encapsulating liposomal nanoparticle (LNP) as a hepatic gene delivery system that is applicable to systemic administration. The key molecular component is a SS-cleavable and pH-activated lipid-like material (ssPalm) that mounts dual sensing motifs (ternary amines and disulfide bonding) that are responsive to the intracellular environment. The main purpose of the present study was to expand its application to a tumor-targeting gene delivery system in mice bearing tumors established from a RCC (OS-RC-2). When the modification of the surface of the particle is optimized for the polyethyleneglycol (PEG), stability in the blood circulation is improved, and consequently tumor-selective gene expression can be achieved. Furthermore, gene expression in the tumor was increased slightly when the hydrophobic scaffold of the ssPalm was replaced from the conventionally used myristic acid (ssPalmM) to α-tocopherol succinate (ssPalmE). Moreover, tumor growth was significantly suppressed when the completely CpG-free pDNA encoding the solute form of VEGFR (fms-like tyrosine kinase-1: sFlt-1) was used, especially when it was delivered by the LNP formed with ssPalmE (LNP(ssPalmE)). Thus, the PEG-modified LNP(ssPalmE) is a promising gene carrier for the cancer gene therapy of RCC.
Collapse
Affiliation(s)
- Hidetaka Akita
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, Japan.
| | - Ryohei Ishiba
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, Japan
| | - Ryohei Togashi
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, Japan
| | - Kota Tange
- NOF CORPORATION, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Yuta Nakai
- NOF CORPORATION, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Hiroto Hatakeyama
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, Japan
| | - Hideyoshi Harashima
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Science, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, Japan.
| |
Collapse
|
44
|
“Programmed packaging” for gene delivery. J Control Release 2014; 193:316-23. [DOI: 10.1016/j.jconrel.2014.04.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 03/26/2014] [Accepted: 04/10/2014] [Indexed: 11/21/2022]
|
45
|
Ukawa M, Akita H, Hayashi Y, Ishiba R, Tange K, Arai M, Kubo K, Higuchi Y, Shimizu K, Konishi S, Hashida M, Harashima H. Neutralized nanoparticle composed of SS-cleavable and pH-activated lipid-like material as a long-lasting and liver-specific gene delivery system. Adv Healthc Mater 2014; 3:1222-9. [PMID: 24668914 DOI: 10.1002/adhm.201300629] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 02/11/2014] [Indexed: 12/17/2022]
Abstract
Charge-neutralized lipid envelope-type nanoparticles formed with SS-cleavable and pH-activated lipid-like materials (ssPalm) accumulate rapidly in the liver without forming aggregates in the blood circulation, and result in a liver-specific gene expression for a long duration (>2 weeks) with neither immunological responses nor hepatotoxicity after intraveneous administration, when it carries pDNA free from CpG-motifs.
Collapse
Affiliation(s)
- Masami Ukawa
- Faculty of Pharmaceutical Sciences, Hokkaido University; Kita12 Nishi6, Kita-ku Sapporo City Hokkaido 060-0812 Japan
| | - Hidetaka Akita
- Faculty of Pharmaceutical Sciences, Hokkaido University; Kita12 Nishi6, Kita-ku Sapporo City Hokkaido 060-0812 Japan
| | - Yasuhiro Hayashi
- Faculty of Pharmaceutical Sciences, Hokkaido University; Kita12 Nishi6, Kita-ku Sapporo City Hokkaido 060-0812 Japan
| | - Ryohei Ishiba
- Faculty of Pharmaceutical Sciences, Hokkaido University; Kita12 Nishi6, Kita-ku Sapporo City Hokkaido 060-0812 Japan
| | - Kota Tange
- NOF Corporation; 3-3 Chidori-cho, Kawasaki-ku Kawasaki Kanagawa 210-0865 Japan
| | - Masaya Arai
- NOF Corporation; 3-3 Chidori-cho, Kawasaki-ku Kawasaki Kanagawa 210-0865 Japan
| | - Kazuhiro Kubo
- NOF Corporation; 3-3 Chidori-cho, Kawasaki-ku Kawasaki Kanagawa 210-0865 Japan
| | - Yuriko Higuchi
- Graduate School of Pharmaceutical Sciences, Kyoto University; 46-28 Yoshida-Shimo-Adachi-cho, Sakyo-ku Kyoto 606-8501 Japan
| | - Kazunori Shimizu
- Graduate School of Pharmaceutical Sciences, Kyoto University; 46-28 Yoshida-Shimo-Adachi-cho, Sakyo-ku Kyoto 606-8501 Japan
- Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University; 1-1-1 Nojihigashi Kusatsu Shiga 525-8577 Japan
| | - Satoshi Konishi
- Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University; 1-1-1 Nojihigashi Kusatsu Shiga 525-8577 Japan
- Department of Micro System Technology; Ritsumeikan University; 1-1-1 Nojihigashi Kusatsu Shiga 525-8577 Japan
| | - Mitsuru Hashida
- Graduate School of Pharmaceutical Sciences, Kyoto University; 46-28 Yoshida-Shimo-Adachi-cho, Sakyo-ku Kyoto 606-8501 Japan
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University; Yoshida-Ushinomiya-cho, Sakyo-ku Kyoto 606-8302 Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University; Kita12 Nishi6, Kita-ku Sapporo City Hokkaido 060-0812 Japan
| |
Collapse
|
46
|
A novel nonviral gene delivery system: multifunctional envelope-type nano device. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2014; 119:197-230. [PMID: 19343308 DOI: 10.1007/10_2008_40] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
In this review we introduce a new concept for developing a nonviral gene delivery system which we call "Programmed Packaging." Based on this concept, we succeeded in developing a multifunctional envelope-type nano device (MEND), which exerts high transfection activities equivalent to those of an adenovirus in a dividing cell. The use of MEND has been extended to in vivo applications. PEG/peptide/DOPE ternary conjugate (PPD)-MEND, a new in vivo gene delivery system for the targeting of tumor cells that dissociates surface-modified PEG in tumor tissue by matrix metalloproteinase (MMP) and exerts significant transfection activities, was developed. In parallel with the development of MEND, a quantitative gene delivery system, Confocal Image-assisted 3-dimensionally integrated quantification (CIDIQ), also was developed. This method identified the rate-limiting step of the nonviral gene delivery system by comparing it with adenoviral-mediated gene delivery. The results of this analysis provide a new direction for the development of rational nonviral gene delivery systems.
Collapse
|
47
|
Intracellular gene delivery is dependent on the type of non-viral carrier and defined by the cell surface glycosaminoglycans. J Control Release 2014; 187:59-65. [PMID: 24838099 DOI: 10.1016/j.jconrel.2014.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 04/28/2014] [Accepted: 05/03/2014] [Indexed: 11/23/2022]
Abstract
Intracellular limiting steps and molecules involved in internalization and intracellular routing of non-viral gene delivery systems are still poorly understood. In this study, the intracellular kinetics of three different gene delivery systems calcium phosphate precipitates (CaP), polyethyleneimine (PEI) and N-[1-(2,3-dioleyl)propyl]-N,N,N-trimethylammonium chloride (DOTAP)) were quantified at cellular, nuclear, transcriptional and translational levels by using qRT-PCR. Additionally, a role of cell surface glycosaminoglycans (GAGs) was evaluated by performing the aforementioned studies in cells devoid of GAGs (pgsB-618) and cells lacking heparan sulphate (HS). The obtained data showed that the intracellular kinetics was dependent on the type of gene carrier and the weakest intracellular step varied between the carriers; rapid elimination of cell-associated pDNA in CaP, nuclear uptake in DOTAP and transcriptional and translational events in PEI mediated transfections. Overall, neither the amount of cell- nor nuclear associated pDNA correlated with transgene expression but the mRNA expression of the transgene correlated well with the expression at protein level. The nuclear uptake of pDNA in all cases was rapid and efficient thus indicating that the post-nuclear processes including transcription and translation steps have a critical role in defining the efficiency of non-viral gene delivery systems. Our study demonstrated that cell-surface GAGs are not essential for cell surface binding and internalization of gene delivery complexes, but they are able to define the intracellular routing of the complexes by leading them to pathways with high pDNA elimination.
Collapse
|
48
|
Pozzi D, Marchini C, Cardarelli F, Salomone F, Coppola S, Montani M, Zabaleta ME, Digman MA, Gratton E, Colapicchioni V, Caracciolo G. Mechanistic evaluation of the transfection barriers involved in lipid-mediated gene delivery: interplay between nanostructure and composition. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:957-67. [PMID: 24296066 DOI: 10.1016/j.bbamem.2013.11.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 11/07/2013] [Accepted: 11/21/2013] [Indexed: 10/25/2022]
Abstract
Here we present a quantitative mechanism-based investigation aimed at comparing the cell uptake, intracellular trafficking, endosomal escape and final fate of lipoplexes and lipid-protamine/deoxyribonucleic acid (DNA) (LPD) nanoparticles (NPs) in living Chinese hamster ovary (CHO) cells. As a model, two lipid formulations were used for comparison. The first formulation is made of the cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and the zwitterionic lipid dioleoylphosphocholine (DOPC), while the second mixture is made of the cationic 3β-[N-(N,N-dimethylaminoethane)-carbamoyl] cholesterol (DC-Chol) and the zwitterionic helper lipid dioleoylphosphatidylethanolamine (DOPE). Our findings indicate that lipoplexes are efficiently taken up through fluid-phase macropinocytosis, while a less efficient uptake of LPD NPs occurs through a combination of both macropinocytosis and clathrin-dependent pathways. Inside the cell, both lipoplexes and LPD NPs are actively transported towards the cell nucleus, as quantitatively addressed by spatio-temporal image correlation spectroscopy (STICS). For each lipid formulation, LPD NPs escape from endosomes more efficiently than lipoplexes. When cells were treated with DOTAP-DOPC-containing systems the majority of the DNA was trapped in the lysosome compartment, suggesting that extensive lysosomal degradation was the rate-limiting factors in DOTAP-DOPC-mediated transfection. On the other side, escape from endosomes is large for DC-Chol-DOPE-containing systems most likely due to DOPE and cholesterol-like molecules, which are able to destabilize the endosomal membrane. The lipid-dependent and structure-dependent enhancement of transfection activity suggests that DNA is delivered to the nucleus synergistically: the process requires both the membrane-fusogenic activity of the nanocarrier envelope and the employment of lipid species with intrinsic endosomal rupture ability.
Collapse
Affiliation(s)
- D Pozzi
- Department of Molecular Medicine, "Sapienza" University of Rome, Viale Regina Elena, 324, 00161 Rome, Italy
| | - C Marchini
- Department of Bioscience and Biotechnology, University of Camerino, Via Gentile III da Varano, 62032 Camerino, MC, Italy
| | - F Cardarelli
- Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - F Salomone
- Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy; NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - S Coppola
- Department of Molecular Medicine, "Sapienza" University of Rome, Viale Regina Elena, 324, 00161 Rome, Italy; Department of Anatomy, Histology, Forensic Medicine and Orthopedics, "Sapienza" University of Rome, Via A. Borelli, 50, 00161 Rome, Italy
| | - M Montani
- Department of Bioscience and Biotechnology, University of Camerino, Via Gentile III da Varano, 62032 Camerino, MC, Italy
| | - M Elexpuru Zabaleta
- Department of Bioscience and Biotechnology, University of Camerino, Via Gentile III da Varano, 62032 Camerino, MC, Italy
| | - M A Digman
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, 3120 Natural Sciences 2, Irvine, CA 92697-2715, USA
| | - E Gratton
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, 3120 Natural Sciences 2, Irvine, CA 92697-2715, USA
| | - V Colapicchioni
- Department of Molecular Medicine, "Sapienza" University of Rome, Viale Regina Elena, 324, 00161 Rome, Italy
| | - G Caracciolo
- Department of Molecular Medicine, "Sapienza" University of Rome, Viale Regina Elena, 324, 00161 Rome, Italy.
| |
Collapse
|
49
|
Tanaka H, Akita H, Ishiba R, Tange K, Arai M, Kubo K, Harashima H. Neutral biodegradable lipid-envelope-type nanoparticle using vitamin A-Scaffold for nuclear targeting of plasmid DNA. Biomaterials 2013; 35:1755-61. [PMID: 24290811 DOI: 10.1016/j.biomaterials.2013.11.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 11/07/2013] [Indexed: 10/26/2022]
Abstract
Biomembranes and cytoplasm, a diffusion-limited region for nanoparticles are critical barriers to be overcome for the successful gene delivery. We herein report on a neutral, and intracellularly degradable lipid nanoparticle (LNP), containing encapsulated plasmid DNA (pDNA) that can be effectively delivered to the nucleus. A key material component in this particle is a vitamin A-scaffold SS-cleavable Proton-Activated Lipid-like Material ((SS)PalmA), which contains tertiary amine groups as proton sponge units that can respond to the acidic pH in endosomes, disulfide bonding for programmed collapse in the cytoplasm, and retinoic acid (RA) as a hydrophobic unit for assembly into LNP. LNP prepared using (SS)PalmA (LNP(PalmA)) exhibited a 15-fold higher gene expression activity compared to particles prepared with a simple acyl chain (myristoyl group)-scaffold one (LNPPalmM). Intracellular imaging studies revealed that LNP(PalmA) unexpectedly showed excessive endosome-disruptive characteristics. Furthermore, the decapsulation of pDNA slowly, but successively occurred in parallel with peri-nuclear accumulation. Nuclear targeting was blocked in the presence of native RA. Collectively, LNP(PalmA) is an intelligent particle that passes through the cytoplasm in particle form with the aid of the intrinsic nuclear transport system of RA, and thereafter releases its encapsulated pDNA for effective gene expression.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Sapporo City, Hokkaido 060-0812, Japan
| | - Hidetaka Akita
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Sapporo City, Hokkaido 060-0812, Japan.
| | - Ryohei Ishiba
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Sapporo City, Hokkaido 060-0812, Japan
| | - Kota Tange
- NOF Corporation, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Masaya Arai
- NOF Corporation, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Kazuhiro Kubo
- NOF Corporation, 3-3 Chidori-cho, Kawasaki-ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Sapporo City, Hokkaido 060-0812, Japan.
| |
Collapse
|
50
|
Investigation of siRNA Nanoparticle Formation Using Mono-Cationic Detergents and Its Use in Gene Silencing in Human HeLa Cells. Cancers (Basel) 2013; 5:1413-25. [PMID: 24202451 PMCID: PMC3875945 DOI: 10.3390/cancers5041413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/25/2013] [Accepted: 10/28/2013] [Indexed: 11/26/2022] Open
Abstract
The focus of recent research has been on the development of siRNA vectors to achieve an innovative gene therapy. Most of the conventional vectors are siRNA nanoparticles complexed with cationic polymers and liposomes, making it difficult to release siRNA. In this study, we report on the use of MCD, a quaternary ammonium salt detergent containing a long aliphatic chain (L-chain) as an siRNA complexation agent using human HeLa cells (a model cancer cell). We prepared siRNA nanoparticles using various MCDs, and measured the diameters and zeta-potentials of the particles. The use of an MCD with a long L-chain resulted in the formation of a positively charged nanoparticle. In contrast, a negatively charged nanoparticle was formed when a MCD with a short L-chain was used. We next evaluated the gene silencing efficiency of the nanoparticles using HeLa cells expressing the luciferase protein. The results showed that the siRNA/MCD nanoparticles showed a higher gene silencing efficiency than Lipofectamine 2000. We also found that the efficiency of gene silencing is a function of the length of the alkyl chain in MCD and zeta-potential of the siRNA/MCD nanoparticles. Such information provides another viewpoint for designing siRNA vectors.
Collapse
|