51
|
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
|
52
|
Fast therapeutic DNA internalization – A high potential transfection system based on a peptide mimicking cationic lipid. Eur J Pharm Biopharm 2017; 118:38-47. [DOI: 10.1016/j.ejpb.2016.12.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 11/21/2016] [Accepted: 12/14/2016] [Indexed: 02/08/2023]
|
53
|
Gu J, Chen X, Fang X, Sha X. Retro-inverso d-peptide-modified hyaluronic acid/bioreducible hyperbranched poly(amido amine)/pDNA core-shell ternary nanoparticles for the dual-targeted delivery of short hairpin RNA-encoding plasmids. Acta Biomater 2017; 57:156-169. [PMID: 28442415 DOI: 10.1016/j.actbio.2017.04.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 03/29/2017] [Accepted: 04/20/2017] [Indexed: 01/08/2023]
Abstract
The active targeting of gene carriers is a powerful strategy for improving tumour-specific delivery and therapy. Although numerous l-peptide ligands play significant roles in the active targeting of nanomedicine, retro-inverso d-peptides have been explored as targeting ligands due to their superior stability and bioactivity in vivo. In this study, retro-inverso d-peptide (RIF7)-modified hyaluronic acid (HA)/bioreducible hyperbranched poly(amido amine) (RHB)/plasmid DNA (pDNA) ternary nanoparticles were successfully developed using the layer-by-layer method for the CD44-positive tumour-specific delivery of short hairpin RNA (shRNA)-encoding pDNA through the combination of the Anxa1 (tumour vasculature) and CD44 (tumour cell-surface) receptors, which mediated the dual targeting. The potential of these newly designed nanoparticles was evaluated by examining the efficacy of their cellular uptake and transfection in cell monolayers, tumour spheroids, and malignant xenograft animal models. With negligible cytotoxicity, the spherical-shaped RIF7-HA/RHB/pDNA nanoparticles were the direct result of an electrostatic complex that had efficiently targeted CD44-positive tumour delivery, penetration, and cellular uptake in vitro. The nanoparticles showed excellent target-specific gene transfection even in the presence of serum. The in vivo therapeutic effect of RIF7-HA/RHB/pDNA-shRNA nanoparticle-mediated shRNA targeting of the Cyclin gene (shCyclin) was evaluated in tumour-bearing mice. The RIF7-HA/RHB/pDNA-shCyclin nanoparticles significantly increased the survival time of tumour-bearing mice and substantially reduced tumour growth due to their extremely specific tumour-targeting activity. These results suggested that the combination of HA and retro-inverso peptide RIF7 significantly increased the therapeutic effect of pDNA-shCyclin-loaded nanoparticles for CD44-positive tumours. Thus, RIF7-HA-mediated multi-target ternary gene vectors are an efficient and promising strategy for the delivery of pDNA-shRNA in the targeted treatment of malignant and metastatic cancers. STATEMENT OF SIGNIFICANCE Although l-peptide ligands play significant roles in the active targeting of nanomedicine, retro-inverso d-peptides have been explored as targeting ligands due to their superior stability and bioactivity in vivo. Retro-inverso peptide RIF7 was designed as a ligand of Anxa1 receptor. The resultant peptide, RIF7, displayed high binding efficiency within Anxa1 receptor, which is highly expressed tumour vasculature cells and some tumour cells such as B16F10 and U87MG cells. The most important feature of RIF7 is its high stability in the blood, which is suitable and promising for application in vivo. Multifunctional RIF7-HA was then synthesized by conjugating the RIF7 peptide to HA, which was used to modify the surface of RHB/pDNA nanoparticles to prepare RIF7-HA/RHB/pDNA core-shell ternary nanoparticles for the dual-targeted delivery of shRNA-encoding plasmids in vitro and in vivo.
Collapse
Affiliation(s)
- Jijin Gu
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China; Laboratory for Drug Delivery and Biomaterials, Faculty of Pharmacy, University of Manitoba, 750 McDermot Ave, Winnipeg, Manitoba R3E 0T5, Canada
| | - Xinyi Chen
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Xiaoling Fang
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Xianyi Sha
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China.
| |
Collapse
|
54
|
Nucleic acid combinations: A new frontier for cancer treatment. J Control Release 2017; 256:153-169. [DOI: 10.1016/j.jconrel.2017.04.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 12/19/2022]
|
55
|
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
|
56
|
Long-term regulation of gene expression in muscle cells by systemically delivered siRNA. J Control Release 2017; 256:101-113. [DOI: 10.1016/j.jconrel.2017.04.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/20/2017] [Accepted: 04/25/2017] [Indexed: 12/20/2022]
|
57
|
Pan L, Liu J, Shi J. Nuclear-Targeting Gold Nanorods for Extremely Low NIR Activated Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2017; 9:15952-15961. [PMID: 28447447 DOI: 10.1021/acsami.7b03017] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Photorelated nanomedicine is of particular interest as an emerging paradigm toward precise cancer therapy, as demonstrated by recent developments of photothermal therapy (PTT), an emerging technique employing light-converting agents to burn cancerous cells by overdosed optical energy-converted heat. However, most of the laser irradiations needed for effective PTT significantly exceed the maximal permissible power density in human skin, which is likely to damage surrounding normal tissues. Herein, we report a strategy of intranuclear PTT of cancer enabled by nuclear-targeted delivery of gold nanorods of ∼10.5 × 40.5 nm in size via conjugation with nuclear location signal peptides (GNRs-NLS) under an extremely low near-infrared irradiation of 0.2 W/cm2, much below the maximal permissible exposure of skin. Interestingly, we found that a mild but nuclear-focused temperature increase generated by GNRs-NLS is sufficient to cause damage to intranuclear DNA and the inhibition of DNA repair process, which, interestingly, led to the cancer cell apoptosis rather than to conventional cell necrosis by thermal ablation during PTT. Correspondingly, tumors treated with GNRs-NLS exhibited gradual but significant regressions rather than traditional harsh burning-up of tumors, in comparison with negligible antitumor effect by GNRs without nuclear targeting under the same ultralow NIR irradiation. This report demonstrates the successful intranuclear efficient photothermal therapy of cancer via cell apoptosis by photoadsorbing agents, e.g., GNRs-NLS in the present case, with largely mitigated side-effect on normal tissues and therefore substantially improved biosafety.
Collapse
Affiliation(s)
- Limin Pan
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences , 1295 Ding-Xi Road, Shanghai 200050, China
| | - Jianan Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences , 1295 Ding-Xi Road, Shanghai 200050, China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences , 1295 Ding-Xi Road, Shanghai 200050, China
| |
Collapse
|
58
|
Yang Z, Li Y, Gao J, Cao Z, Jiang Q, Liu J. pH and redox dual-responsive multifunctional gene delivery with enhanced capability of transporting DNA into the nucleus. Colloids Surf B Biointerfaces 2017; 153:111-122. [DOI: 10.1016/j.colsurfb.2017.02.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/25/2017] [Accepted: 02/13/2017] [Indexed: 12/16/2022]
|
59
|
Abstract
Physiological characteristics of diseases bring about both challenges and opportunities for targeted drug delivery. Various drug delivery platforms have been devised ranging from macro- to micro- and further into the nanoscopic scale in the past decades. Recently, the favorable physicochemical properties of nanomaterials, including long circulation, robust tissue and cell penetration attract broad interest, leading to extensive studies for therapeutic benefits. Accumulated knowledge about the physiological barriers that affect the in vivo fate of nanomedicine has led to more rational guidelines for tailoring the nanocarriers, such as size, shape, charge, and surface ligands. Meanwhile, progresses in material chemistry and molecular pharmaceutics generate a panel of physiological stimuli-responsive modules that are equipped into the formulations to prepare “smart” drug delivery systems. The capability of harnessing physiological traits of diseased tissues to control the accumulation of or drug release from nanomedicine has further improved the controlled drug release profiles with a precise manner. Successful clinical translation of a few nano-formulations has excited the collaborative efforts from the research community, pharmaceutical industry, and the public towards a promising future of smart drug delivery.
Collapse
Affiliation(s)
- Wujin Sun
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Quanyin Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Wenyan Ji
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Grace Wright
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
60
|
Yu J, Zhang J, Xing H, Sun Y, Yang Z, Yang T, Cai C, Zhao X, Yang L, Ding P. Novel guanidinylated bioresponsive poly(amidoamine)s designed for short hairpin RNA delivery. Int J Nanomedicine 2016; 11:6651-6666. [PMID: 27994462 PMCID: PMC5154728 DOI: 10.2147/ijn.s115773] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Two different disulfide (SS)-containing poly(amidoamine) (PAA) polymers were constructed using guanidino (Gua)-containing monomers (ie, arginine [Arg] and agmatine [Agm]) and N,N′-cystamine bisacrylamide (CBA) by Michael-addition polymerization. In order to characterize these two Gua-SS-PAA polymers and investigate their potentials as short hairpin RNA (shRNA)-delivery carriers, pSilencer 4.1-CMV FANCF shRNA was chosen as a model plasmid DNA to form complexes with these two polymers. The Gua-SS-PAAs and plasmid DNA complexes were determined with particle sizes less than 90 nm and positive ζ-potentials under 20 mV at nucleic acid:polymer weight ratios lower than 1:24. Bioresponsive release of plasmid DNA was observed from both newly constructed complexes. Significantly lower cytotoxicity was observed for both polymer complexes compared with polyethylenimine and Lipofectamine 2000, two widely used transfection reagents as reference carriers. Arg-CBA showed higher transfection efficiency and gene-silencing efficiency in MCF7 cells than Agm-CBA and the reference carriers. In addition, the cellular uptake of Arg-CBA in MCF7 cells was found to be higher and faster than Agm-CBA and the reference carriers. Similarly, plasmid DNA transport into the nucleus mediated by Arg-CBA was more than that by Agm-CBA and the reference carriers. The study suggested that guanidine and carboxyl introduced into Gua-SS-PAAs polymers resulted in a better nuclear localization effect, which played a key role in the observed enhancement of transfection efficiency and low cytotoxicity. Overall, two newly synthesized Gua-SS-PAAs polymers demonstrated great potential to be used as shRNA carriers for gene-therapy applications.
Collapse
Affiliation(s)
- Jiankun Yu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Jinmin Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Haonan Xing
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yanping Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhen Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME, USA
| | - Cuifang Cai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaoyun Zhao
- Department of Microbiology and Cell Biology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Li Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
61
|
Andersen H, Parhamifar L, Hunter AC, Shahin V, Moghimi SM. AFM visualization of sub-50 nm polyplex disposition to the nuclear pore complex without compromising the integrity of the nuclear envelope. J Control Release 2016; 244:24-29. [DOI: 10.1016/j.jconrel.2016.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 10/18/2016] [Accepted: 11/10/2016] [Indexed: 02/07/2023]
|
62
|
Patel BJ, Vignesh NK, Hortelano G. Chitosan DNA nanoparticles for oral gene delivery. World J Med Genet 2016; 6:22-33. [DOI: 10.5496/wjmg.v6.i3.22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 08/08/2016] [Indexed: 02/06/2023] Open
Abstract
Gene therapy is a promising technology with potential applications in the treatment of medical conditions, both congenital and acquired. Despite its label as breakthrough technology for the 21st century, the simple concept of gene therapy - the introduction of a functional copy of desired genes in affected individuals - is proving to be more challenging than expected. Oral gene delivery has shown intriguing results and warrants further exploration. In particular, oral administration of chitosan DNA nanoparticles, one the most commonly used formulations of therapeutic DNA, has repeatedly demonstrated successful in vitro and in vivo gene transfection. While oral gene therapy has shown immense promise as treatment options in a variety of diseases, there are still significant barriers to overcome before it can be considered for clinical applications. In this review we provide an overview of the physiologic challenges facing the use of chitosan DNA nanoparticles for oral gene delivery at both the extracellular and intracellular level. From administration at the oral cavity, chitosan nanoparticles must traverse the gastrointestinal tract and protect its DNA contents from significant jumps in pH levels, various intestinal digestive enzymes, thick mucus layers with high turnover, and a proteinaceous glycocalyx meshwork. Once these extracellular barriers are overcome, chitosan DNA nanoparticles must enter intestinal cells, escape endolysosomes, and disassociate from genetic material at the appropriate time allowing transport of genetic material into the nucleus to deliver a therapeutic effect. The properties of chitosan nanoparticles and modified nanoparticles are discussed in this review. An understanding of the barriers to oral gene delivery and how to overcome them would be invaluable for future gene therapy development.
Collapse
|
63
|
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
|
64
|
Kirchenbuechler I, Kirchenbuechler D, Elbaum M. Correlation between cationic lipid-based transfection and cell division. Exp Cell Res 2016; 345:1-5. [DOI: 10.1016/j.yexcr.2014.11.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 10/27/2014] [Accepted: 11/24/2014] [Indexed: 12/12/2022]
|
65
|
Graphene in therapeutics delivery: Problems, solutions and future opportunities. Eur J Pharm Biopharm 2016; 104:235-50. [DOI: 10.1016/j.ejpb.2016.04.015] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 04/12/2016] [Accepted: 04/18/2016] [Indexed: 01/05/2023]
|
66
|
Gu J, Chen X, Ren X, Zhang X, Fang X, Sha X. CD44-Targeted Hyaluronic Acid-Coated Redox-Responsive Hyperbranched Poly(amido amine)/Plasmid DNA Ternary Nanoassemblies for Efficient Gene Delivery. Bioconjug Chem 2016; 27:1723-36. [DOI: 10.1021/acs.bioconjchem.6b00240] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jijin Gu
- Key
Laboratory of Smart Drug Delivery (Fudan University), Ministry of
Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
- Laboratory
for Drug Delivery and Biomaterials, Faculty of Pharmacy, University of Manitoba, 750 McDermot Avenue, Winnipeg, Manitoba R3E 0T5, Canada
| | - Xinyi Chen
- Key
Laboratory of Smart Drug Delivery (Fudan University), Ministry of
Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Xiaoqing Ren
- Key
Laboratory of Smart Drug Delivery (Fudan University), Ministry of
Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Xiulei Zhang
- Key
Laboratory of Smart Drug Delivery (Fudan University), Ministry of
Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Xiaoling Fang
- Key
Laboratory of Smart Drug Delivery (Fudan University), Ministry of
Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Xianyi Sha
- Key
Laboratory of Smart Drug Delivery (Fudan University), Ministry of
Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| |
Collapse
|
67
|
Sun Y, Xian L, Xing H, Yu J, Yang Z, Yang T, Yang L, Ding P. Factors influencing the nuclear targeting ability of nuclear localization signals. J Drug Target 2016; 24:927-933. [DOI: 10.1080/1061186x.2016.1184273] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
68
|
Pérez-López AM, Soria-Gila ML, Marsden ER, Lilienkampf A, Bradley M. Fluorogenic Substrates for In Situ Monitoring of Caspase-3 Activity in Live Cells. PLoS One 2016; 11:e0153209. [PMID: 27168077 PMCID: PMC4864350 DOI: 10.1371/journal.pone.0153209] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/26/2016] [Indexed: 02/06/2023] Open
Abstract
The in situ detection of caspase-3 activity has applications in the imaging and monitoring of multiple pathologies, notably cancer. A series of cell penetrating FRET-based fluorogenic substrates were designed and synthesised for the detection of caspase-3 in live cells. A variety of modifications of the classical caspase-3 and caspase-7 substrate sequence Asp-Glu-Val-Asp were carried out in order to increase caspase-3 affinity and eliminate caspase-7 cross-reactivity. To allow cellular uptake and good solubility, the substrates were conjugated to a cationic peptoid. The most selective fluorogenic substrate 27, FAM-Ahx-Asp-Leu-Pro-Asp-Lys(MR)-Ahx, conjugated to the cell penetrating peptoid at the C-terminus, was able to detect and quantify caspase-3 activity in apoptotic cells without cross-reactivity by caspase-7.
Collapse
Affiliation(s)
- Ana M. Pérez-López
- School of Chemistry, EaStCHEM, University of Edinburgh, Joseph Black building, West Mains Road, Edinburgh EH9 3FJ, United Kingdom
| | - M. Lourdes Soria-Gila
- Department of Medicinal and Organic Chemistry, University of Granada, School of Pharmacy, Campus Cartuja s/n – 18071, Granada, Spain
| | - Emma R. Marsden
- School of Chemistry, EaStCHEM, University of Edinburgh, Joseph Black building, West Mains Road, Edinburgh EH9 3FJ, United Kingdom
| | - Annamaria Lilienkampf
- School of Chemistry, EaStCHEM, University of Edinburgh, Joseph Black building, West Mains Road, Edinburgh EH9 3FJ, United Kingdom
| | - Mark Bradley
- School of Chemistry, EaStCHEM, University of Edinburgh, Joseph Black building, West Mains Road, Edinburgh EH9 3FJ, United Kingdom
- * E-mail:
| |
Collapse
|
69
|
Chen W, Li H, Liu Z, Yuan W. Lipopolyplex for Therapeutic Gene Delivery and Its Application for the Treatment of Parkinson's Disease. Front Aging Neurosci 2016; 8:68. [PMID: 27092073 PMCID: PMC4820442 DOI: 10.3389/fnagi.2016.00068] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 03/21/2016] [Indexed: 01/10/2023] Open
Abstract
Lipopolyplex is a core-shell structure composed of nucleic acid, polycation and lipid. As a non-viral gene delivery vector, lipopolyplex combining the advantages of polyplex and lipoplex has shown superior colloidal stability, reduced cytotoxicity, extremely high gene transfection efficiency. Following intravenous administration, there are many strategies based on lipopolyplex to overcome the complex biological barriers in systemic gene delivery including condensation of nucleic acids into nanoparticles, long circulation, cell targeting, endosomal escape, release to cytoplasm and entry into cell nucleus. Parkinson's disease (PD) is the second most common neurodegenerative disorder and severely influences the patients' life quality. Current gene therapy clinical trials for PD employing viral vectors didn't achieve satisfactory efficacy. However, lipopolyplex may become a promising alternative approach owing to its stability in blood, ability to cross the blood-brain barrier (BBB) and specific targeting to diseased brain cells.
Collapse
Affiliation(s)
- Wei Chen
- Department of Neurology, Xinhua Hospital, Shanghai JiaoTong University School of Medicine Shanghai, China
| | - Hui Li
- School of Pharmacy, Shanghai JiaoTong University Shanghai, China
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital, Shanghai JiaoTong University School of Medicine Shanghai, China
| | - Weien Yuan
- School of Pharmacy, Shanghai JiaoTong University Shanghai, China
| |
Collapse
|
70
|
Yousefpour Marzbali M, Yari Khosroushahi A, Movassaghpour A, Yeganeh H. Polyurethane dispersion containing quaternized ammonium groups: An efficient nanosize gene delivery carrier for A549 cancer cell line transfection. Chem Biol Interact 2016; 244:27-36. [DOI: 10.1016/j.cbi.2015.11.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/29/2015] [Accepted: 11/25/2015] [Indexed: 12/13/2022]
|
71
|
Rombouts K, Braeckmans K, Remaut K. Fluorescent Labeling of Plasmid DNA and mRNA: Gains and Losses of Current Labeling Strategies. Bioconjug Chem 2015; 27:280-97. [PMID: 26670733 DOI: 10.1021/acs.bioconjchem.5b00579] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Live-cell imaging has provided the life sciences with insights into the cell biology and dynamics. Fluorescent labeling of target molecules proves to be indispensable in this regard. In this Review, we focus on the current fluorescent labeling strategies for nucleic acids, and in particular mRNA (mRNA) and plasmid DNA (pDNA), which are of interest to a broad range of scientific fields. By giving a background of the available techniques and an evaluation of the pros and cons, we try to supply scientists with all the information needed to come to an informed choice of nucleic acid labeling strategy aimed at their particular needs.
Collapse
Affiliation(s)
- K Rombouts
- Laboratory of general biochemistry and physical pharmacy, Faculty of pharmacy and ‡Centre for Nano- and Biophotonics, Ghent University , Ghent 9000, Belgium
| | - K Braeckmans
- Laboratory of general biochemistry and physical pharmacy, Faculty of pharmacy and ‡Centre for Nano- and Biophotonics, Ghent University , Ghent 9000, Belgium
| | - K Remaut
- Laboratory of general biochemistry and physical pharmacy, Faculty of pharmacy and ‡Centre for Nano- and Biophotonics, Ghent University , Ghent 9000, Belgium
| |
Collapse
|
72
|
Crowley ST, Rice KG. "Evolving nanoparticle gene delivery vectors for the liver: What has been learned in 30 years". J Control Release 2015; 219:457-470. [PMID: 26439664 DOI: 10.1016/j.jconrel.2015.10.008] [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: 06/03/2015] [Revised: 10/01/2015] [Accepted: 10/02/2015] [Indexed: 12/18/2022]
Abstract
Nonviral gene delivery to the liver has been under evolution for nearly 30years. Early demonstrations established relatively simple nonviral vectors could mediate gene expression in HepG2 cells which understandably led to speculation that these same vectors would be immediately successful at transfecting primary hepatocytes in vivo. However, it was soon recognized that the properties of a nonviral vector resulting in efficient transfection in vitro were uncorrelated with those needed to achieve efficient nonviral transfection in vivo. The discovery of major barriers to liver gene transfer has set the field on a course to design biocompatible vectors that demonstrate increased DNA stability in the circulation with correlating expression in liver. The improved understanding of what limits nonviral vector gene transfer efficiency in vivo has resulted in more sophisticated, low molecular weight vectors that allow systematic optimization of nanoparticle size, charge and ligand presentation. While the field has evolved DNA nanoparticles that are stable in the circulation, target hepatocytes, and deliver DNA to the cytosol, breaching the nucleus remains the last major barrier to a fully successful nonviral gene transfer system for the liver. The lessons learned along the way are fundamentally important to the design of all systemically delivered nanoparticle nonviral gene delivery systems.
Collapse
Affiliation(s)
- Samuel T Crowley
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, IA 52242,USA
| | - Kevin G Rice
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, IA 52242,USA.
| |
Collapse
|
73
|
Characterization of exogenous DNA mobility in live cells through fluctuation correlation spectroscopy. Sci Rep 2015; 5:13848. [PMID: 26354725 PMCID: PMC4564760 DOI: 10.1038/srep13848] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 08/04/2015] [Indexed: 12/28/2022] Open
Abstract
The spatial-temporal dynamics of delivered DNA is a critical aspect influencing successful gene delivery. A comprehensive model of DNA lipoplex trafficking through live cells has yet to be demonstrated. Here the bioimaging approaches Raster Image Correlation Spectroscopy (RICS) and image-Means Square Displacement (iMSD) were applied to quantify DNA mechanical dynamics in live cells. DNA lipoplexes formed from DNA with a range of 21 bp to 5.5 kbp exhibited a similar range of motion within the cytoplasm of myoblast cells regardless of size. However, the rate of motion was dictated by the intracellular location, and DNA cluster size. This analysis demonstrated that the different transport mechanisms either had a size dependent mobility, including random diffusion, whereas other mechanisms were not influenced by the DNA size such as active transport. The transport mechanisms identified followed a spatial dependence comparable to viral trafficking of non-active transport mechanism upon cellular entry, active transport within the cytoplasm and further inactive transportation along the peri-nuclear region. This study provides the first real-time insight into the trafficking of DNA delivered through lipofection using image-based fluctuation correlation spectroscopy approaches. Thereby, gaining information with single particle sensitivity to develop a deeper understanding of DNA lipoplex delivery through the cell.
Collapse
|
74
|
In vitro gene manipulation of spinal muscular atrophy fibroblast cell line using gene-targeting fragment for restoration of SMN protein expression. Gene Ther 2015; 23:10-7. [DOI: 10.1038/gt.2015.92] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 06/17/2015] [Accepted: 08/05/2015] [Indexed: 11/08/2022]
|
75
|
Vernon MM, Dean DA, Dobson J. DNA Targeting Sequence Improves Magnetic Nanoparticle-Based Plasmid DNA Transfection Efficiency in Model Neurons. Int J Mol Sci 2015; 16:19369-86. [PMID: 26287182 PMCID: PMC4581301 DOI: 10.3390/ijms160819369] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/28/2015] [Accepted: 08/04/2015] [Indexed: 11/19/2022] Open
Abstract
Efficient non-viral plasmid DNA transfection of most stem cells, progenitor cells and primary cell lines currently presents an obstacle for many applications within gene therapy research. From a standpoint of efficiency and cell viability, magnetic nanoparticle-based DNA transfection is a promising gene vectoring technique because it has demonstrated rapid and improved transfection outcomes when compared to alternative non-viral methods. Recently, our research group introduced oscillating magnet arrays that resulted in further improvements to this novel plasmid DNA (pDNA) vectoring technology. Continued improvements to nanomagnetic transfection techniques have focused primarily on magnetic nanoparticle (MNP) functionalization and transfection parameter optimization: cell confluence, growth media, serum starvation, magnet oscillation parameters, etc. Noting that none of these parameters can assist in the nuclear translocation of delivered pDNA following MNP-pDNA complex dissociation in the cell’s cytoplasm, inclusion of a cassette feature for pDNA nuclear translocation is theoretically justified. In this study incorporation of a DNA targeting sequence (DTS) feature in the transfecting plasmid improved transfection efficiency in model neurons, presumably from increased nuclear translocation. This observation became most apparent when comparing the response of the dividing SH-SY5Y precursor cell to the non-dividing and differentiated SH-SY5Y neuroblastoma cells.
Collapse
Affiliation(s)
- Matthew M Vernon
- Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| | - David A Dean
- School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Jon Dobson
- Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL 32603, USA.
- Institute for Cell Engineering & Regenerative Medicine (ICERM), University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
76
|
Truong DM, Hewitt FC, Hanson JH, Cui X, Lambowitz AM. Retrohoming of a Mobile Group II Intron in Human Cells Suggests How Eukaryotes Limit Group II Intron Proliferation. PLoS Genet 2015; 11:e1005422. [PMID: 26241656 PMCID: PMC4524724 DOI: 10.1371/journal.pgen.1005422] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 07/05/2015] [Indexed: 12/22/2022] Open
Abstract
Mobile bacterial group II introns are evolutionary ancestors of spliceosomal introns and retroelements in eukaryotes. They consist of an autocatalytic intron RNA (a “ribozyme”) and an intron-encoded reverse transcriptase, which function together to promote intron integration into new DNA sites by a mechanism termed “retrohoming”. Although mobile group II introns splice and retrohome efficiently in bacteria, all examined thus far function inefficiently in eukaryotes, where their ribozyme activity is limited by low Mg2+ concentrations, and intron-containing transcripts are subject to nonsense-mediated decay (NMD) and translational repression. Here, by using RNA polymerase II to express a humanized group II intron reverse transcriptase and T7 RNA polymerase to express intron transcripts resistant to NMD, we find that simply supplementing culture medium with Mg2+ induces the Lactococcus lactis Ll.LtrB intron to retrohome into plasmid and chromosomal sites, the latter at frequencies up to ~0.1%, in viable HEK-293 cells. Surprisingly, under these conditions, the Ll.LtrB intron reverse transcriptase is required for retrohoming but not for RNA splicing as in bacteria. By using a genetic assay for in vivo selections combined with deep sequencing, we identified intron RNA mutations that enhance retrohoming in human cells, but <4-fold and not without added Mg2+. Further, the selected mutations lie outside the ribozyme catalytic core, which appears not readily modified to function efficiently at low Mg2+ concentrations. Our results reveal differences between group II intron retrohoming in human cells and bacteria and suggest constraints on critical nucleotide residues of the ribozyme core that limit how much group II intron retrohoming in eukaryotes can be enhanced. These findings have implications for group II intron use for gene targeting in eukaryotes and suggest how differences in intracellular Mg2+ concentrations between bacteria and eukarya may have impacted the evolution of introns and gene expression mechanisms. Mobile group II introns are bacterial retrotransposons that are evolutionary ancestors of spliceosomal introns and retroelements in eukaryotes. They consist of an autocatalytic intron RNA (a ribozyme) and an intron-encoded reverse transcriptase, which together promote intron mobility to new DNA sites by a mechanism called retrohoming. Although found in bacteria, archaea and eukaryotic organelles, group II introns are absent from eukaryotic nuclear genomes, where host defenses impede their expression and lower intracellular Mg2+ concentrations limit their ribozyme activity. Here, we developed a mobile group II intron expression system that bypasses expression barriers and show that simply adding Mg2+ to culture medium enables group II intron retrohoming into plasmid and chromosomal target sites in human cells at appreciable frequencies. Genetic selections and deep sequencing identified intron RNA mutations that moderately enhance retrohoming in human cells, but not without added Mg2+. Thus, low Mg2+ concentrations in human cells are a natural barrier to efficient retrohoming that is not readily overcome by mutational variation and selection. Our results have implications for group II intron use for gene targeting in higher organisms and highlight the impact of different intracellular environments on intron evolution and gene expression mechanisms in bacteria and eukarya.
Collapse
Affiliation(s)
- David M. Truong
- Institute for Cellular and Molecular Biology, Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - F. Curtis Hewitt
- Institute for Cellular and Molecular Biology, Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Joseph H. Hanson
- Institute for Cellular and Molecular Biology, Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Xiaoxia Cui
- Institute for Cellular and Molecular Biology, Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Alan M. Lambowitz
- Institute for Cellular and Molecular Biology, Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
77
|
Maizels Y, Gerlitz G. Shaping of interphase chromosomes by the microtubule network. FEBS J 2015; 282:3500-24. [PMID: 26040675 DOI: 10.1111/febs.13334] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/11/2015] [Accepted: 06/01/2015] [Indexed: 12/31/2022]
Abstract
It is well established that microtubule dynamics play a major role in chromosome condensation and localization during mitosis. During interphase, however, it is assumed that the metazoan nuclear envelope presents a physical barrier, which inhibits interaction between the microtubules located in the cytoplasm and the chromatin fibers located in the nucleus. In recent years, it has become apparent that microtubule dynamics alter chromatin structure and function during interphase as well. Microtubule motor proteins transport several transcription factors and exogenous DNA (such as plasmid DNA) from the cytoplasm to the nucleus. Various soluble microtubule components are able to translocate into the nucleus, where they bind various chromatin elements leading to transcriptional alterations. In addition, microtubules may apply force on the nuclear envelope, which is transmitted into the nucleus, leading to changes in chromatin structure. Thus, microtubule dynamics during interphase may affect chromatin spatial organization, as well as transcription, replication and repair.
Collapse
Affiliation(s)
- Yael Maizels
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Israel
| | - Gabi Gerlitz
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Israel
| |
Collapse
|
78
|
Götz C, Warnke PH, Kolk A. Current and future options of regeneration methods and reconstructive surgery of the facial skeleton. Oral Surg Oral Med Oral Pathol Oral Radiol 2015; 120:315-23. [PMID: 26297391 DOI: 10.1016/j.oooo.2015.05.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 04/27/2015] [Accepted: 05/26/2015] [Indexed: 01/06/2023]
Abstract
Musculoskeletal defects attributable to trauma or infection or as a result of oncologic surgery present a common challenge in reconstructive maxillofacial surgery. The autologous vascularized bone graft still represents the gold standard for salvaging these situations. Preoperative virtual planning offers great potential and provides assistance in reconstructive surgery. Nevertheless, the applicability of autologous bone transfer might be limited within the medically compromised patient or because of the complexity of the defect and the required size of the graft to be harvested. The development of alternative methods are urgently needed in the field of regenerative medicine to enable the regeneration of the original tissue. Since the first demonstration of de novo bone formation by regenerative strategies and the application of bone growth factors some decades ago, further progress has been achieved by tissue engineering, gene transfer, and stem cell application concepts. This review summarizes recent approaches and current developments in regenerative medicine.
Collapse
Affiliation(s)
- Carolin Götz
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Patrick H Warnke
- Department of Oral and Maxillofacial Surgery, University of Kiel, Kiel, Germany; Belegärztliche Gemeinschaftspraxis für Oral-, Mund- und Kieferchirurgie und plastische Gesichtschirurgie Dres. Sprengel und Klebe, Flensburg, Germany
| | - Andreas Kolk
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany.
| |
Collapse
|
79
|
Abstract
Viruses are promising vehicles that result in high gene expression level, but issues of safety and virulent nature prevented its extensive use. Therefore, nonviral approach was investigated with the intervention of nanomedicine. The science of nanomedicine offered an excellent platform for therapeutic delivery as they provide options to include functionalities and engineer the system. As the term 'nano' refers to the generation of a very small dimension structure, their unique physicochemical characteristics with increased surface area/volume ratio made them potential vectors to perform gene therapy. Various forms of nanoparticles are continued to be synthesised, and this review discusses the immediate barriers that nanoparticles have to encounter both during systemic movement in the body and intracellular trafficking to deliver the genes at the site of action.
Collapse
Affiliation(s)
- Susan Muthe Alex
- Facility for Nano/Microparticles Based Biomaterials for Advanced Drug Delivery Systems (FADDS) Division of Biosurface Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Poojappura, Thiruvananthapuram, Kerala, 695012, India
| | | |
Collapse
|
80
|
Christensen MD, Elmer JJ, Eaton S, Gonzalez-Malerva L, LaBaer J, Rege K. Kinome-level screening identifies inhibition of polo-like kinase-1 (PLK1) as a target for enhancing non-viral transgene expression. J Control Release 2015; 204:20-9. [PMID: 25681050 PMCID: PMC8292636 DOI: 10.1016/j.jconrel.2015.01.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 01/13/2015] [Accepted: 01/28/2015] [Indexed: 01/01/2023]
Abstract
Human cells contain hundreds of kinase enzymes that regulate several cellular processes, which likely include transgene delivery and expression. We identified several kinases that influence gene delivery and/or expression by performing a kinome-level screen in which, we identified small-molecule kinase inhibitors that significantly enhanced non-viral (polymer-mediated) transgene (luciferase) expression in cancer cells. The strongest enhancement was observed with several small-molecule inhibitors of Polo-like Kinase 1 (PLK 1) (e.g., HMN-214 and BI 2536), which enhanced luciferase expression up to 30-fold by arresting cells in the G2/M phase of the cell cycle and influencing intracellular trafficking of plasmid DNA. Knockdown of PLK 1 using an shRNA-expressing lentivirus further confirmed the enhancement of polymer-mediated transgene expression. In addition, pairwise and three-way combinations of PLK1 inhibitors with the histone deacetylase-1 (HDAC-1) inhibitor Entinostat and the JAK/STAT inhibitor AG-490 enhanced luciferase expression to levels significantly higher than individual drug treatments acting alone. These findings indicate that inhibition of specific intracellular kinases (e.g., PLK1) can significantly enhance non-viral transgene expression for applications in biotechnology and medicine.
Collapse
Affiliation(s)
- Matthew D Christensen
- Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, USA
| | - Jacob J Elmer
- Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, USA
| | - Seron Eaton
- The Virginia G. Piper Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Laura Gonzalez-Malerva
- The Virginia G. Piper Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Joshua LaBaer
- The Virginia G. Piper Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Kaushal Rege
- Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
81
|
Pavlin M, Kandušer M. New insights into the mechanisms of gene electrotransfer--experimental and theoretical analysis. Sci Rep 2015; 5:9132. [PMID: 25778848 PMCID: PMC5390920 DOI: 10.1038/srep09132] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 02/11/2015] [Indexed: 01/14/2023] Open
Abstract
Gene electrotransfer is a promising non-viral method of gene delivery. In our in vitro study we addressed open questions about this multistep process: how electropermeabilization is related to electrotransfer efficiency; the role of DNA electrophoresis for contact and transfer across the membrane; visualization and theoretical analysis of DNA-membrane interaction and its relation to final transfection efficiency; and the differences between plated and suspended cells. Combinations of high-voltage and low-voltage pulses were used. We obtained that electrophoresis is required for the insertion of DNA into the permeabilized membrane. The inserted DNA is slowly transferred into the cytosol, and nuclear entry is a limiting factor for optimal transfection. The quantification and theoretical analysis of the crucial parameters reveals that DNA-membrane interaction (NDNA) increases with higher DNA concentration or with the addition of electrophoretic LV pulses while transfection efficiency reaches saturation. We explain the differences between the transfection of cell suspensions and plated cells due to the more homogeneous size, shape and movement of suspended cells. Our results suggest that DNA is either translocated through the stable electropores or enters by electo-stimulated endocytosis, possibly dependent on pulse parameters. Understanding of the mechanisms enables the selection of optimal electric protocols for specific applications.
Collapse
Affiliation(s)
- Mojca Pavlin
- Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, 1000 Ljubljana, Slovenia
| | - Maša Kandušer
- Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, 1000 Ljubljana, Slovenia
| |
Collapse
|
82
|
Navarro G, Pan J, Torchilin VP. Micelle-like nanoparticles as carriers for DNA and siRNA. Mol Pharm 2015; 12:301-13. [PMID: 25557580 DOI: 10.1021/mp5007213] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Gene therapy represents a potential efficient approach of disease prevention and therapy. However, due to their poor in vivo stability, gene molecules need to be associated with delivery systems to overcome extracellular and intracellular barriers and allow access to the site of action. Cationic polymeric nanoparticles are popular carriers for small interfering RNA (siRNA) and DNA-based therapeutics for which efficient and safe delivery are important factors that need to be optimized. Micelle-like nanoparticles (MNP) (half micelles, half polymeric nanoparticles) can overcome some of the disadvantages of such cationic carriers by unifying in one single carrier the best of both delivery systems. In this review, we will discuss how the unique properties of MNP including self-assembly, condensation and protection of nucleic acids, improved cell association and gene transfection, and low toxicity may contribute to the successful application of siRNA- and DNA-based therapeutics into the clinic. Recent developments of MNP involving the addition of stimulus-sensitive functions to respond specifically to pathological or externally applied "triggers" (e.g., temperature, pH or enzymatic catalysis, light, or magnetic fields) will be discussed. Finally, we will overview the use of MNP as two-in-one carriers for the simultaneous delivery of different agents (small molecules, imaging agents) and nucleic acid combinations.
Collapse
Affiliation(s)
- Gemma Navarro
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States
| | | | | |
Collapse
|
83
|
Nanoparticle-based technologies for retinal gene therapy. Eur J Pharm Biopharm 2015; 95:353-67. [PMID: 25592325 DOI: 10.1016/j.ejpb.2014.12.028] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/15/2014] [Accepted: 12/22/2014] [Indexed: 01/17/2023]
Abstract
For patients with hereditary retinal diseases, retinal gene therapy offers significant promise for the prevention of retinal degeneration. While adeno-associated virus (AAV)-based systems remain the most popular gene delivery method due to their high efficiency and successful clinical results, other delivery systems, such as non-viral nanoparticles (NPs) are being developed as additional therapeutic options. NP technologies come in several categories (e.g., polymer, liposomes, peptide compacted DNA), several of which have been tested in mouse models of retinal disease. Here, we discuss the key biochemical features of the different NPs that influence how they are internalized into cells, escape from endosomes, and are delivered into the nucleus. We review the primary mechanism of NP uptake by retinal cells and highlight various NPs that have been successfully used for in vivo gene delivery to the retina and RPE. Finally, we consider the various strategies that can be implemented in the plasmid DNA to generate persistent, high levels of gene expression.
Collapse
|
84
|
Andries O, Kitada T, Bodner K, Sanders NN, Weiss R. Synthetic biology devices and circuits for RNA-based ‘smart vaccines’: a propositional review. Expert Rev Vaccines 2015; 14:313-31. [DOI: 10.1586/14760584.2015.997714] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
85
|
Imani R, Emami SH, Faghihi S. Synthesis and characterization of an octaarginine functionalized graphene oxide nano-carrier for gene delivery applications. Phys Chem Chem Phys 2015; 17:6328-39. [DOI: 10.1039/c4cp04301d] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The success of gene therapy is largely dependent on the development of a gene carrier.
Collapse
Affiliation(s)
- Rana Imani
- Department of Biomedical Engineering
- Amirkabir University of Technology
- Tehran 15875/4413
- Iran
- Tissue Engineering and Biomaterials Division
| | - Shahriar Hojjati Emami
- Department of Biomedical Engineering
- Amirkabir University of Technology
- Tehran 15875/4413
- Iran
| | - Shahab Faghihi
- Tissue Engineering and Biomaterials Division
- National Institute of Genetic Engineering and Biotechnology
- Tehran 14965/161
- Iran
| |
Collapse
|
86
|
Yue J, Wu J, Liu D, Zhao X, Lu WW. BMP2 gene delivery to bone mesenchymal stem cell by chitosan-g-PEI nonviral vector. NANOSCALE RESEARCH LETTERS 2015; 10:203. [PMID: 25977673 PMCID: PMC4420764 DOI: 10.1186/s11671-015-0906-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/18/2015] [Indexed: 05/21/2023]
Abstract
Nanotechnology has made a significant impact on the development of nanomedicine. Nonviral vectors have been attracting more attention for the advantage of biosafety in gene delivery. Polyethylenimine (PEI)-conjugated chitosan (chitosan-g-PEI) emerged as a promising nonviral vector and has been demonstrated in many tumor cells. However, there is a lack of study focused on the behavior of this vector in stem cells which hold great potential in regenerative medicine. Therefore, in this study, in vitro gene delivering effect of chitosan-g-PEI was investigated in bone marrow stem cells. pIRES2-ZsGreen1-hBMP2 dual expression plasmid containing both the ZsGreen1 GFP reporter gene and the BMP2 functional gene was constructed for monitoring the transgene expression level. Chitosan-g-PEI-mediated gene transfer showed 17.2% of transfection efficiency and more than 80% of cell viability in stem cells. These values were higher than that of PEI. The expression of the delivered BMP2 gene in stem cells enhanced the osteogenic differentiation. These results demonstrated that chitosan-g-PEI is capable of applying in delivering gene to stem cells and providing potential applications in stem cell-based gene therapy.
Collapse
Affiliation(s)
- Jianhui Yue
- />Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Rd., Shenzhen, 518055 People’s Republic of China
- />Shenzhen Key Laboratory of Marine Biomedical Materials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Rd., Shenzhen, 518055 People’s Republic of China
| | - Jun Wu
- />Department of Orthopaedic and Traumatology, The University of Hong Kong, 21 Sassoon Rd., Pokfulam, Hong Kong, 999077 People’s Republic of China
| | - Di Liu
- />Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Rd., Shenzhen, 518055 People’s Republic of China
- />Department of Pharmacology, Harbin Medical University, 157 Baojian Rd., Harbin, 150081 People’s Republic of China
| | - Xiaoli Zhao
- />Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Rd., Shenzhen, 518055 People’s Republic of China
- />Shenzhen Key Laboratory of Marine Biomedical Materials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Rd., Shenzhen, 518055 People’s Republic of China
| | - William W Lu
- />Department of Orthopaedic and Traumatology, The University of Hong Kong, 21 Sassoon Rd., Pokfulam, Hong Kong, 999077 People’s Republic of China
| |
Collapse
|
87
|
Kapoor M, Burgess DJ. Targeted Delivery of Nucleic Acid Therapeutics via Nonviral Vectors. ADVANCES IN DELIVERY SCIENCE AND TECHNOLOGY 2015. [DOI: 10.1007/978-3-319-11355-5_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
88
|
Gaspar V, de Melo-Diogo D, Costa E, Moreira A, Queiroz J, Pichon C, Correia I, Sousa F. Minicircle DNA vectors for gene therapy: advances and applications. Expert Opin Biol Ther 2014; 15:353-79. [PMID: 25539147 DOI: 10.1517/14712598.2015.996544] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Nucleic-acid-based biopharmaceuticals enclose a remarkable potential for treating debilitating or life-threatening diseases that currently remain incurable. This promising area of research envisages the creation of state-of-the-art DNA vaccines, pluripotent cells or gene-based therapies, which can be used to overcome current issues. To achieve this goal, DNA minicircles are emerging as ideal nonviral vectors due to their safety and persistent transgene expression in either quiescent or actively dividing cells. AREAS COVERED This review focuses on the characteristics of minicircle DNA (mcDNA) technology and the current advances in their production. The possible modifications to further improve minicircle efficacy are also emphasized and discussed in light of recent advances. As a final point, the main therapeutic applications of mcDNA are summarized, with a special focus on pluripotent stem cells production and cancer therapy. EXPERT OPINION Achieving in-target and persistent transgene expression is a challenging issue that is of critical importance for a successful therapeutic outcome. The use of miniaturized mcDNA cassettes with additional modifications that increase and prolong expression may contribute to an improved generation of biopharmaceuticals. The unique features of mcDNA render it an attractive alternative to overcome current technical issues and to bridge the significant gap that exists between basic research and clinical applications.
Collapse
Affiliation(s)
- Vítor Gaspar
- University of Beira Interior, CICS-UBI - Health Sciences Research Center , Av. Infante D. Henrique, 6200-506, Covilhã , Portugal +351 275 329 002, +351 275 329 055 ; +351 275 329 099 ; ;
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Pan L, Liu J, He Q, Shi J. MSN-mediated sequential vascular-to-cell nuclear-targeted drug delivery for efficient tumor regression. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2014; 26:6742-6748. [PMID: 25159109 DOI: 10.1002/adma.201402752] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Indexed: 06/03/2023]
Abstract
Mesoporous silica nanoparticles functionalized with peptides are developed for sequential drug delivery. The RGD peptide is used for vasculature/cell membrane targeting and the TAT peptide for nuclear targeting. Using this delivery strategy, a tumor in a murine xenograft model is successfully regressed.
Collapse
Affiliation(s)
- Limin Pan
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Ding-xi Road, Shanghai, 200050, China
| | | | | | | |
Collapse
|
90
|
Zhang J, Bae S, Lee JS, Webb K. Efficacy and mechanism of poloxamine-assisted polyplex transfection. J Gene Med 2014; 15:271-81. [PMID: 23813893 DOI: 10.1002/jgm.2719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 06/10/2013] [Accepted: 06/24/2013] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Amphiphilic block copolymers acting as biological response modifiers provide an attractive approach for improving the transfection efficiency of polycationic polymer/DNA complexes (polyplexes) by altering cellular processes crucial for efficient transgene expression. METHODS The present study aimed to investigate the effect of the poloxamine Tetronic T904, a four-arm polyethylene oxide/polypropylene oxide block copolymer, on polyplex transfection and to determine its mechanism of action by analyzing the cellular uptake of polyplex, the nuclear localization of plasmid and RNA transcript production. RESULTS T904 significantly increased the transfection efficiency of polyplexes based on 25-kDa branched polyethylenimine in a dose-dependent manner in the presence of serum in C6 glioma cells, as well as human fibroblasts and mesenchymal stem cells. The activity of T904 was not promoter-dependent, increasing the expression of reporter genes under both cytomegalovirus and SV40 promoters. Although T904 did not affect the internalization or nuclear uptake of plasmid, mRNA expression levels from both promoters showed dose-dependent increases that closely paralleled increases in gene expression. CONCLUSIONS The present study demonstrates that T904 significantly increases polyplex transfection efficiency and suggests a mechanism of increased transcriptional activity. As a four-arm, hydroxyl-terminated polymer, T904 is amenable to a variety of end group functionalization and covalent cross-linking strategies that have been developed for preparing hydrogels from multi-arm polyethylene glycol, making it particularly attractive for scaffold-mediated gene delivery.
Collapse
Affiliation(s)
- Jeremy Zhang
- Microenvironmental Engineering Laboratory, Department of Bioengineering, Clemson University, Clemson, SC, USA
| | | | | | | |
Collapse
|
91
|
Lee YS, Kim SW. Bioreducible polymers for therapeutic gene delivery. J Control Release 2014; 190:424-39. [PMID: 24746626 DOI: 10.1016/j.jconrel.2014.04.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/09/2014] [Accepted: 04/09/2014] [Indexed: 01/18/2023]
Abstract
Most currently available cationic polymers have significant acute toxicity concerns such as cellular toxicity, aggregation of erythrocytes, and entrapment in the lung capillary bed, largely due to their poor biocompatibility and non-degradability under physiological conditions. To develop more intelligent polymers, disulfide bonds are introduced in the design of biodegradable polymers. Herein, the sustained innovations of biomimetic nano-sized constructs with bioreducible poly(disulfide amine)s demonstrate a viable clinical tool for the treatment of cardiovascular disease, anemia, diabetes, and cancer.
Collapse
Affiliation(s)
- Young Sook Lee
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, USA.
| | - Sung Wan Kim
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, USA; Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
92
|
Fioretti D, Iurescia S, Rinaldi M. Enhancement of plasmid-mediated transgene expression. Methods Mol Biol 2014; 1143:11-20. [PMID: 24715279 DOI: 10.1007/978-1-4939-0410-5_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
A large number of studies aimed at the treatment of cancer, autoimmune and metabolic diseases, neurodegenerative disorders, allergic diseases, as well as muscle disorders strengthen the fact that gene therapy could represent an alternative method to treat human diseases where conventional approaches are less effective. To improve transgene expression from plasmid vectors, DNA nuclear targeting sequences (DTSs) can be introduced in a vector backbone to increase in vivo expression up to 20-fold using electroporation (EP) delivery in muscle tissue. The purpose of this chapter is to represent a step-by-step strategy for the construction of a plasmid vector with enhanced efficiency of nuclear plasmid uptake and the methodic for the in vivo efficiency evaluation of the obtained expression vector.
Collapse
Affiliation(s)
- Daniela Fioretti
- Section of Medical Biotechnology, Institute of Translational Pharmacology (IFT), National Research Council (CNR), via Fosso del Cavaliere 100, 00133, Rome, Italy
| | | | | |
Collapse
|
93
|
Bao T, Wang H, Zhang W, Xia X, Zhou J, Weng W, Yu D. APPLICATION OF DENDRIMER/PLASMID hBMP-2 COMPLEXES LOADED INTO β-TCP/COLLAGEN SCAFFOLD IN THE TREATMENT OF FEMORAL DEFECTS IN RATS. BIOMEDICAL ENGINEERING-APPLICATIONS BASIS COMMUNICATIONS 2014. [DOI: 10.4015/s1016237214500057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Purpose: Plasmid loading into scaffolds to enhance sustained release of growth factors is an important focus of regenerative medicine. The aim of this study was to build gene-activated matrices (GAMs) and examine the bone augmentation properties. Methods: Generation 5 polyamidoamine dendrimers (G5 dPAMAM)/plasmid recombinant human bone morphogenetic protein-2 (rhBMP-2) complexes were immobilized into beta-tricalcium phosphate (β-TCP)/type I collagen porous scaffolds. After cultured with rat mesenchymal stem cells (rMSCs), transfection efficiencies were examined. The secretion of rhBMP-2 and alkaline phosphatase (ALP) were detected to evaluate the osteogenic properties. Scanning electron microscopy (SEM) was used to observe attachment and proliferation. Moreover, we applied these GAMs directly into freshly created segmental bone defects in rat femurs, and their osteogenic efficiencies were evaluated. Results: Released plasmid complexes were transfected into stem cells and were expressed, which caused osteogenic differentiations of rat mesenchymal stem cells (rMSCs). SEM analysis showed excellent cell attachment. Bioactivity of plasmid rhBMP-2 was maintained in vivo, and the X-ray observation, histological analysis and immunohistochemistry (IHC) of bone tissue demonstrated that the bone healing in segmental femoral defects was enhanced by implantation of GAMs. Conclusions: Such biomaterials offer therapeutic opportunities in critical-sized bone defects.
Collapse
Affiliation(s)
- Tingwei Bao
- Department of Stomatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Faculty of Dentistry, Zhejiang University, Hangzhou 310003, China
| | - Huiming Wang
- Department of Stomatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Faculty of Dentistry, Zhejiang University, Hangzhou 310003, China
| | - Wentao Zhang
- Department of Stomatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Faculty of Dentistry, Zhejiang University, Hangzhou 310003, China
| | - Xuefeng Xia
- Department of Hepatobiliary Surgery, Key Laboratory of Multi-Organ Transplantation of Ministry of Public Health, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jiabei Zhou
- State Key Laboratory of Silicon Materials and Department of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Wenjian Weng
- State Key Laboratory of Silicon Materials and Department of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Dan Yu
- Department of Stomatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Faculty of Dentistry, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
94
|
On the possible involvement of bovine serum albumin precursor in lipofection pathway. J Biosci 2014; 39:43-52. [PMID: 24499789 DOI: 10.1007/s12038-014-9415-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Protein factors involved in lipofection pathways remain elusive. Using avidin-biotin affinity chromatography and mass finger printing analysis technique, herein we report the identification of a 70 kDa size protein (bovine serum albumin precursor, BSAP) which binds strongly with lipoplexes and may play role in lipofection pathway. Using multiple cultured animal cells and three structurally different cationic transfection lipids, we show that the efficiencies of liposomal transfection vectors get significantly enhanced (by ~2.5- to 5.0-fold) in cells pre-transfected with lipoplexes of reporter plasmid construct encoding BSAP. Findings in the cellular uptake experiments in A549 cells cultured in DMEM supplemented with 10 percent (w/w) BODIPY-labelled BSAP are consistent with the supposition that BSAP enters cell cytoplasm from the cell culture medium (DMEM supplemented with 10 percent FBS) used in lipofection. Cellular uptake studies by confocal microscopy using BODIPY-labelled BSAP and FITC-labelled plasmid DNA revealed co-localization of plasmid DNA and BSAP within the cell cytoplasm and nucleus. In summary, the present findings hint at the possible involvement of BSAP in lipofection pathway.
Collapse
|
95
|
New views and insights into intracellular trafficking of drug-delivery systems by fluorescence fluctuation spectroscopy. Ther Deliv 2014; 5:173-88. [DOI: 10.4155/tde.13.148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Biomaterials in the nanometer size range can be engineered for site-specific delivery of drugs after injection into the blood circulation. However, translation of such nanomedicines from the bench to the bedside is still hindered by many extracellular and intracellular barriers. To realize the concept of targeted drug delivery with nanomedicines, research groups are studying intensively the extra- and intra-cellular mechanisms involved as a response to the physicochemical properties of the nanomedicines. In this review, we highlight the contributions of fluorescence fluctuations spectroscopy techniques to better understand, and in turn to bypass, the major hurdles to therapeutic delivery, focusing mostly on the intracellular dynamics of drug-delivery systems.
Collapse
|
96
|
Abstract
Although safety concerns have been overcome, lower immunogenicity profiles of DNA vaccines have hindered their progress in humans. DNA vaccines need to make up for this limitation by altering plasmid construction through vector design innovations intended for enhancement of transgene expression and immunogenicity. The next-generation vectors also address safety issues such as selection markers. This chapter discusses (a) plasmid backbone design, (b) enhancement of antigenic protein expression and immunogenicity, and (c) vector modification to increase innate immunity. Modifications of the basic design, when combined with improved delivery devices and/or prime/boost regimens, may enhance DNA vaccine performance and clinical outcomes.
Collapse
Affiliation(s)
- Sandra Iurescia
- Section of Medical Biotechnology, Institute of Translational Pharmacology (IFT), National Research Council (CNR), via Fosso del Cavaliere 100, 00133, Rome, Italy
| | | | | |
Collapse
|
97
|
Wegman F, van der Helm Y, Öner FC, Dhert WJ, Alblas J. Bone Morphogenetic Protein-2 Plasmid DNA as a Substitute for Bone Morphogenetic Protein-2 Protein in Bone Tissue Engineering. Tissue Eng Part A 2013; 19:2686-92. [DOI: 10.1089/ten.tea.2012.0569] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Fiona Wegman
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Yvonne van der Helm
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - F. Cumhur Öner
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wouter J.A. Dhert
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jacqueline Alblas
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
98
|
De La Vega J, Braak BT, Azzoni AR, Monteiro GA, Prazeres DMF. Impact of plasmid quality on lipoplex-mediated transfection. J Pharm Sci 2013; 102:3932-41. [PMID: 23996350 DOI: 10.1002/jps.23709] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 07/22/2013] [Accepted: 07/29/2013] [Indexed: 01/20/2023]
Abstract
This work investigates the impact of quality attributes (impurity content, plasmid charge, and compactness) of plasmid DNA isolated with different purification methodologies on the characteristics of lipoplexes prepared thereof (size, zeta potential, stability) and on their ability to transfect mammalian cells. A 3.7 kb plasmid with a green fluorescence protein (GFP) reporter gene, Lipofectamine®-based liposomes, and Chinese Hamster Ovary (CHO) cells were used as models. The plasmid was purified by hydrophobic interaction chromatography (HIC)/gel filtration, and with three commercial kits, which combine the use of chaotropic salts with silica membranes/glass fiber fleeces. The HIC-based protocol delivered a plasmid with the smallest hydrodynamic diameter (144 nm) and zeta potential (-46.5 mV), which is virtually free from impurities. When formulated with Lipofectamine®, this plasmid originated the smallest (146 nm), most charged (+13 mV), and most stable lipoplexes. In vitro transfection experiments further showed that these lipoplexes performed better in terms of plasmid uptake (∼500,000 vs. ∼100,000-200,000 copy number/cell), transfection efficiency (50% vs. 20%-40%), and GFP expression levels (twofold higher) when compared with lipoplexes prepared with plasmids isolated using commercial kits. Overall our observations highlight the potential impact that plasmid purification methodologies can have on the outcome of gene transfer experiments and trials.
Collapse
Affiliation(s)
- Jonathan De La Vega
- IBB-Institute for Biotechnology and Bioengineering, Centre for Biological and Chemical Engineering, Department of Bioengineering, Instituto Superior Técnico, Universidade Técnica de Lisboa, Lisboa, 1049-001, Portugal
| | | | | | | | | |
Collapse
|
99
|
Abstract
Development of antitumor preparations with low toxicity and high selectivity of action is one of the top priorities of cancer gene therapy. Mesenchymal stem cells possess natural tropism towards tumors, a property that makes possible their use as a vehicle for targeted delivery of therapeutic genes into tumors of various etiologies. At present, genes encoding enzymes (cytosine deaminase, thymidine kinase, carboxyl esterase), cytokines (IL-2, IL-4, IL-12, IFN-beta) and apoptosis inducing factors (TRAIL) are used as therapeutic genes. Mesenchymal stem cells, as demonstrated using experimental models of tumors of various etiologies as well as animals with metastases in brain and lungs, are able to successfully deliver therapeutic genes into tumors and produce significant antitumor effect. However, to effectively use this therapeutic strategy in clinic, one still has to solve a number of technical problems.
Collapse
|
100
|
Gaspar VM, Marques JG, Sousa F, Louro RO, Queiroz JA, Correia IJ. Biofunctionalized nanoparticles with pH-responsive and cell penetrating blocks for gene delivery. NANOTECHNOLOGY 2013; 24:275101. [PMID: 23759860 DOI: 10.1088/0957-4484/24/27/275101] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Bridging the gap between nanoparticulate delivery systems and translational gene therapy is a long sought after requirement in nanomedicine-based applications. However, recent developments regarding nanoparticle functionalization have brought forward the ability to synthesize materials with biofunctional moieties that mimic the evolved features of viral particles. Herein we report the versatile conjugation of both cell penetrating arginine and pH-responsive histidine moieties into the chitosan polymeric backbone, to improve the physicochemical characteristics of the native material. Amino acid coupling was confirmed by 2D TOCSY NMR and Fourier transform infrared spectroscopy. The synthesized chitosan-histidine-arginine (CH-H-R) polymer complexed plasmid DNA biopharmaceuticals, and spontaneously assembled into stable 105 nm nanoparticles with spherical morphology and positive surface charge. The functionalized delivery systems were efficiently internalized into the intracellular compartment, and exhibited remarkably higher transfection efficiency than unmodified chitosan without causing any cytotoxic effect. Additional findings regarding intracellular trafficking events reveal their preferential escape from degradative lysosomal pathways and nuclear localization. Overall, this assembly of nanocarriers with bioinspired moieties provides the foundations for the design of efficient and customizable materials for cancer gene therapy.
Collapse
Affiliation(s)
- V M Gaspar
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | | | | | | | | | | |
Collapse
|