1
|
Maze D, Pichon C, Midoux P. Reversible stabilization of DNA/PEI complexes by reducible click-linkage between DNA and polymer. A new polyplex concept for lowering polymer quantity. Gene Ther 2023; 30:783-791. [PMID: 36755129 DOI: 10.1038/s41434-023-00386-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 01/18/2023] [Accepted: 01/27/2023] [Indexed: 02/10/2023]
Abstract
Nonviral transfection of mammalian cells can be performed with electrostatic complexes (polyplexes) between a plasmid DNA (pDNA) encoding a foreign gene and a cationic polymer. However, an excess of the cationic polymer is required for pDNA condensation and polyplexes formation, which generate in vivo toxicity. Here, we present a new concept of polyplexes preparation aiming to reduce the polymer quantity. pDNA was functionalized with 3,6,9-trioxaundecan-1- {4 - [(2-chloroethyl) ethylamino)] - benzylamino}, 11-azide, and polyethyleneimine (lPEI) with reducible dibenzocyclooctyl (SS-DBCO) groups allowing azide-alkyne cycloaddition between pDNA and lPEI after condensation. The size of polyplexes with DBCO-SS-lPEI was smaller than with lPEI due to a stronger DNA condensation thanks to linkages between polymer and pDNA preventing dissociation until disulfide bridges reduction. In vitro transfection showed that the amount of DBCO-SS-lPEI leading to the most efficient polyplexes was three times lower than lPEI. As expected, toxicity in mice was significantly reduced upon intravenous injection of DBCO-SS-lPEI polyplexes at doses where the lPEI polyplexes killed mice. This is probably due to the high stability of the DBCO-SS-lPEI polyplexes which prevented their aggregation in the pulmonary capillaries. Overall, this new concept of polyplexes with DBCO-SS-lPEI offering the possibility of administering higher doses of polyplexes than lPEI and their ability to pass the pulmonary barrier could be favorably exploited for transfection of distant organs or tissues, such as tumors.
Collapse
Affiliation(s)
- Delphine Maze
- Centre de Biophysique Moléculaire, CNRS UPR4301, Inserm and University of Orléans, F-45071, Orléans cedex 02, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS UPR4301, Inserm and University of Orléans, F-45071, Orléans cedex 02, France
| | - Patrick Midoux
- Centre de Biophysique Moléculaire, CNRS UPR4301, Inserm and University of Orléans, F-45071, Orléans cedex 02, France.
| |
Collapse
|
2
|
Maze D, Girardin C, Benz N, Montier T, Pichon C, Midoux P. CFTR and dystrophin encoding plasmids carrying both luciferase reporter gene, nuclear import specific sequences and triple helix sites. Plasmid 2023; 127:102686. [PMID: 37207938 DOI: 10.1016/j.plasmid.2023.102686] [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: 11/07/2022] [Revised: 04/26/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023]
Abstract
Duchenne Muscular Dystrophy and Cystic Fibrosis are two major monogenetic diseases which could be treated by non-viral gene therapy. For this purpose, plasmid DNA (pDNA) coding for the functional genes requires its equipment with signal molecules favouring its intracellular trafficking and delivery in the nucleus of the target cells. Here, two novel constructions of large pDNAs encoding the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) and full-length dystrophin (DYS) genes are reported. The expression of CFTR and DYS genes are driven respectively by the hCEF1 airway epithelial cells and spc5-12 muscle cells specific promoter. Those pDNAs encode also the luciferase reporter gene driven by the CMV promoter to evaluate gene delivery in animals by bioluminescence. In addition, oligopurine • oligopyrimidine sequences are inserted to enable equipment of pDNAs with peptides conjugated with a triple helix forming oligonucleotide (TFO). Furthermore, specific κB sequences are also inserted to promote their NFκB-mediated nuclear import. pDNA constructions are reported; transfection efficiency, tissue specific expression of CFTR and dystrophin in target cells, and triple helix formation are demonstrated. These plasmids are tools of interest to develop non-viral gene therapy of Cystic Fibrosis and Duchenne Muscular Dystrophy.
Collapse
Affiliation(s)
- Delphine Maze
- Centre de Biophysique Moléculaire, CNRS UPR4301, Inserm and University of Orléans, 45071 Orléans cedex 02, France
| | - Caroline Girardin
- Centre de Biophysique Moléculaire, CNRS UPR4301, Inserm and University of Orléans, 45071 Orléans cedex 02, France
| | - Nathalie Benz
- Univ Brest, INSERM, EFS, UMR 1078, GGB - GTCA Team, Brest F-29200, France
| | - Tristan Montier
- Univ Brest, INSERM, EFS, UMR 1078, GGB - GTCA Team, Brest F-29200, France; Service de Génétique Médicale et Biologie de la Reproduction, Centre de référence des maladies rares 'Maladies neuromusculaires', CHRU de Brest, Brest F-29200, France
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS UPR4301, Inserm and University of Orléans, 45071 Orléans cedex 02, France
| | - Patrick Midoux
- Centre de Biophysique Moléculaire, CNRS UPR4301, Inserm and University of Orléans, 45071 Orléans cedex 02, France.
| |
Collapse
|
3
|
CNS Delivery of Nucleic Acid Therapeutics: Beyond the Blood-Brain Barrier and Towards Specific Cellular Targeting. Pharm Res 2023; 40:77-105. [PMID: 36380168 DOI: 10.1007/s11095-022-03433-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022]
Abstract
Nucleic acid-based therapeutic molecules including small interfering RNA (siRNA), microRNA(miRNA), antisense oligonucleotides (ASOs), messenger RNA (mRNA), and DNA-based gene therapy have tremendous potential for treating diseases in the central nervous system (CNS). However, achieving clinically meaningful delivery to the brain and particularly to target cells and sub-cellular compartments is typically very challenging. Mediating cell-specific delivery in the CNS would be a crucial advance that mitigates off-target effects and toxicities. In this review, we describe these challenges and provide contemporary evidence of advances in cellular and sub-cellular delivery using a variety of delivery mechanisms and alternative routes of administration, including the nose-to-brain approach. Strategies to achieve subcellular localization, endosomal escape, cytosolic bioavailability, and nuclear transfer are also discussed. Ultimately, there are still many challenges to translating these experimental strategies into effective and clinically viable approaches for treating patients.
Collapse
|
4
|
Shin S, Kim SH, Lee JS, Lee GM. Streamlined Human Cell-Based Recombinase-Mediated Cassette Exchange Platform Enables Multigene Expression for the Production of Therapeutic Proteins. ACS Synth Biol 2021; 10:1715-1727. [PMID: 34133132 DOI: 10.1021/acssynbio.1c00113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A platform, based on targeted integration of transgenes using recombinase-mediated cassette exchange (RMCE) coupled with CRISPR/Cas9, is increasingly being used for the development of mammalian cell lines that produce therapeutic proteins, because of reduced clonal variation and predictable transgene expression. However, low efficiency of the RMCE process has hampered its application in multicopy or multisite integration of transgenes. To improve RMCE efficiency, nuclear transport of RMCE components such as site-specific recombinase and donor plasmid was accelerated by incorporation of nuclear localization signal and DNA nuclear-targeting sequence, respectively. Consequently, the efficiency of RMCE in dual-landing pad human embryonic kidney 293 (HEK293) cell lines harboring identical or orthogonal pairs of recombination sites at two well-known human safe harbors (AAVS1 and ROSA26 loci), increased 6.7- and 8.1-fold, respectively. This platform with enhanced RMCE efficiency enabled simultaneous integration of transgenes at the two sites using a single transfection without performing selection and enrichment processes. The use of a homotypic dual-landing pad HEK293 cell line capable of incorporating the same transgenes at two sites resulted in a 2-fold increase in the transgene expression level compared to a single-landing pad HEK293 cell line. In addition, the use of a heterotypic dual-landing pad HEK293 cell line, which can incorporate transgenes for a recombinant protein at one site and an effector transgene for cell engineering at another site, increased recombinant protein production. Overall, a streamlined RMCE platform can be a versatile tool for mammalian cell line development by facilitating multigene expression at genomic safe harbors.
Collapse
Affiliation(s)
- Seunghyeon Shin
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Su Hyun Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Jae Seong Lee
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| |
Collapse
|
5
|
CEBA: A new heterobifunctional reagent for plasmid DNA functionalization by click chemistry. Int J Pharm 2021; 601:120566. [PMID: 33812974 DOI: 10.1016/j.ijpharm.2021.120566] [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: 12/19/2020] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 11/20/2022]
Abstract
Here, we report the synthesis of 3,6,9-trioxaundecan-1-{4-[(2-Chloroethyl)Ethylamino)]-Benzylamino},11-Azide (CEBA). CEBA alkylates the N7 of guanine of DNA thanks its chloroethyl group and can be coupled by a strain-promoted azide-alkyne cycloaddition to an alkynylated molecule. The optimization of the alkylation level of pDNA reveals that the expression of the encoded gene is preserved when it is randomly modified with at most 1 CEBA molecule per 150 bp. We show that the azido group of CEBA allows the linkage via click chemistry of CEBA-pDNA with a fluorophore or a peptide containing a dibenzocyclooctyne (DBCO) function. This new heterobifunctional reagent opens new ways to equip pDNA easily with signal molecules including peptides and nucleic acids without side products providing great interest for non-viral gene therapy.
Collapse
|
6
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 181] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
7
|
Lopez-Salas FE, Nadella R, Maldonado-Berny M, Escobedo-Sanchez ML, Fiorentino-Pérez R, Gatica-García B, Fernandez-Parrilla MA, Mario Gil M, Reyes-Corona D, García U, Orozco-Barrios CE, Gutierrez-Castillo ME, Martinez-Fong D. Synthetic Monopartite Peptide That Enables the Nuclear Import of Genes Delivered by the Neurotensin-Polyplex Vector. Mol Pharm 2020; 17:4572-4588. [PMID: 33125243 DOI: 10.1021/acs.molpharmaceut.0c00755] [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] [Indexed: 12/12/2022]
Abstract
Neurotensin (NTS)-polyplex is a multicomponent nonviral vector that enables gene delivery via internalization of the neurotensin type 1 receptor (NTSR1) to dopaminergic neurons and cancer cells. An approach to improving its therapeutic safety is replacing the viral karyophilic component (peptide KPSV40; MAPTKRKGSCPGAAPNKPK), which performs the nuclear import activity, by a shorter synthetic peptide (KPRa; KMAPKKRK). We explored this issue and the mechanism of plasmid DNA translocation through the expression of the green fluorescent protein or red fluorescent protein fused with KPRa and internalization assays and whole-cell patch-clamp configuration experiments in a single cell together with importin α/β pathway blockers. We showed that KPRa electrostatically bound to plasmid DNA increased the transgene expression compared with KPSV40 and enabled nuclear translocation of KPRa-fused red fluorescent proteins and plasmid DNA. Such translocation was blocked with ivermectin or mifepristone, suggesting importin α/β pathway mediation. KPRa also enabled NTS-polyplex-mediated expression of reporter or physiological genes such as human mesencephalic-derived neurotrophic factor (hMANF) in dopaminergic neurons in vivo. KPRa is a synthetic monopartite peptide that showed nuclear import activity in NTS-polyplex vector-mediated gene delivery. KPRa could also improve the transfection of other nonviral vectors used in gene therapy.
Collapse
Affiliation(s)
- Francisco E Lopez-Salas
- Programa de Doctorado en Nanociencias y Nanotecnología, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional, No. 2508, San Pedro Zacatenco, 07360 Ciudad de México, Mexico
| | - Rasajna Nadella
- Biosciences, IIIT Srikakulam-RGUKT, Etcherla 532402, Srikakulam District, Andhra Pradesh, India
| | - Minerva Maldonado-Berny
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional, No. 2508, San Pedro Zacatenco, 07360 Ciudad de México, Mexico
| | - Maria L Escobedo-Sanchez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional, No. 2508, San Pedro Zacatenco, 07360 Ciudad de México, Mexico
| | - Rosana Fiorentino-Pérez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional, No. 2508, San Pedro Zacatenco, 07360 Ciudad de México, Mexico
| | - Bismark Gatica-García
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional, No. 2508, San Pedro Zacatenco, 07360 Ciudad de México, Mexico
| | - Manuel A Fernandez-Parrilla
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional, No. 2508, San Pedro Zacatenco, 07360 Ciudad de México, Mexico
| | - Moreno Mario Gil
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional, No. 2508, San Pedro Zacatenco, 07360 Ciudad de México, Mexico
| | - David Reyes-Corona
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional, No. 2508, San Pedro Zacatenco, 07360 Ciudad de México, Mexico
| | - Ubaldo García
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional, No. 2508, San Pedro Zacatenco, 07360 Ciudad de México, Mexico
| | - Carlos E Orozco-Barrios
- Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Unidad de Investigaciones Médicas en Enfermedades Neurológicas, CONACyT, Av. Cuauhtémoc 330, Doctores, 06720 Ciudad de México, Mexico
| | - Maria E Gutierrez-Castillo
- Departamento de Biociencias e Ingeniería, Centro Interdisciplinario de Investigaciones y Estudios sobre Medio Ambiente y Desarrollo, Instituto Politécnico Nacional, 30 de junio de 1520 s/n, La Laguna Ticoman, 07340 Ciudad de Mexico, Mexico
| | - Daniel Martinez-Fong
- Programa de Doctorado en Nanociencias y Nanotecnología, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional, No. 2508, San Pedro Zacatenco, 07360 Ciudad de México, Mexico.,Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Av. Instituto Politécnico Nacional, No. 2508, San Pedro Zacatenco, 07360 Ciudad de México, Mexico
| |
Collapse
|
8
|
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: 6.0] [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
|
9
|
Asayama S. Molecular Design of Polymer-based Carriers for Plasmid DNA Delivery In Vitro and In Vivo. CHEM LETT 2020. [DOI: 10.1246/cl.190696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Shoichiro Asayama
- Department of Applied Chemistry, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo 192-0397, Japan
| |
Collapse
|
10
|
Zhang YM, Zhang J, Liu YH, Guo Y, Yu XQ, Huang Z. Zinc( ii)-cyclen coordinative amphiphiles for enhanced gene delivery. RSC Adv 2020; 10:39842-39853. [PMID: 35515358 PMCID: PMC9057412 DOI: 10.1039/d0ra08027f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 10/23/2020] [Indexed: 12/18/2022] Open
Abstract
In this study, we developed coordinative amphiphiles for use as novel non-viral DNA vectors. As a modification of a conventional cationic lipid structure, we replaced the cationic head with a zinc(ii)-1,4,7,10-tetraazacyclododecane (Zn-cyclen) complex as a phosphate-directing group, and used biocompatible skeletons (α-tocopherol or cholesterol) as hydrophobic tails. The structure-activity relationship (SAR) was systematically investigated to study the effect of Zn-coordination on the gene transfection between cyclen-based traditional head-tail lipids and Zn(ii)-cyclen coordinative amphiphiles. The results reveal that both Zn-free lipids and Zn-containing amphiphiles could condense DNA into nano-sized particles with appropriate size and zeta-potentials. Agarose gel retardation assay and MTS-based cell viability assays demonstrated that the Zn(ii)-cyclen complex exhibited slightly lower DNA binding ability and much lower cytotoxicity compared to liposome analogues, respectively. Most importantly, in vitro transfection studies showed that the coordination of zinc(ii) to cyclen may dramatically increase the transfection efficiency of the conventional cationic lipid, and α-tocopherol-containing coordinative amphiphile Zn-Cyc-Toc gives the best transfection efficiency, which was enhanced 24.4 times after coordination and was 6.1 times higher than commercial transfection reagent lipofectamine 2000. Mechanism studies confirmed that the DNA complex formed from Zn-Cyc-Toc might induce higher cellular uptake and better endosomal escape ability than the lipoplexes formed from Zn-free lipid Cyc-Toc. This study not only demonstrates that these coordinative amphiphiles might be promising non-viral gene vectors, but also presents a novel strategy to enhance the gene transfection efficiency and biocompatibility of cyclen-based cationic lipids. Zn-coordination could dramatically enhance the gene transfection efficiency and reduce the cytotoxicity of conventional cyclen-based cationic liposomes.![]()
Collapse
Affiliation(s)
- Yi-Mei Zhang
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics
- Chongqing Key Laboratory of Kinase Modulators As Innovative Medicine
- College of Pharmacy & International Academy of Targeted Therapeutics and Innovation
- Chongqing University of Arts and Sciences
- Chongqing 402160
| | - Ji Zhang
- Key Laboratory of Green Chemistry & Technology (Ministry of Education)
- College of Chemistry
- Sichuan University
- Chengdu 610064
- China
| | - Yan-Hong Liu
- Key Laboratory of Green Chemistry & Technology (Ministry of Education)
- College of Chemistry
- Sichuan University
- Chengdu 610064
- China
| | - Yu Guo
- Key Laboratory of Green Chemistry & Technology (Ministry of Education)
- College of Chemistry
- Sichuan University
- Chengdu 610064
- China
| | - Xiao-Qi Yu
- Key Laboratory of Green Chemistry & Technology (Ministry of Education)
- College of Chemistry
- Sichuan University
- Chengdu 610064
- China
| | - Zheng Huang
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics
- Chongqing Key Laboratory of Kinase Modulators As Innovative Medicine
- College of Pharmacy & International Academy of Targeted Therapeutics and Innovation
- Chongqing University of Arts and Sciences
- Chongqing 402160
| |
Collapse
|
11
|
Dendrimers in gene delivery. PHARMACEUTICAL APPLICATIONS OF DENDRIMERS 2020. [DOI: 10.1016/b978-0-12-814527-2.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
12
|
You HS, Ok YJ, Lee EJ, Kang SS, Hyun SH. Development of a novel DsRed-NLS vector with a monopartite classical nuclear localization signal. 3 Biotech 2019; 9:232. [PMID: 31139547 DOI: 10.1007/s13205-019-1770-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/18/2019] [Indexed: 12/01/2022] Open
Abstract
The nuclear localization signal (NLS) marks proteins for transport to the nucleus and is used in various applications in many fields. NLSs are used to achieve efficient and stable transport of biomolecules. Previously, commercial vectors used in NLS studies contained three iterations of the NLS sequence, but these sequences can affect experimental results and alter protein function. Here, we investigated a new vector using a single classical NLS sequence with a mutation in pDsRed2-C1-wt to reduce experimental artifacts. In the newly constructed pDsRed2-C1-1NLS vector, the NLS sequence is placed near the multiple cloning sites of pDsRed2-C1-wt, and the multiple cloning site region was designed to facilitate insertion of the desired gene by site-directed mutagenesis. Fluorescent protein expression in the nucleus can be visually confirmed. The results show that the fluorescent protein was bound to the transport protein. The constructed vector had a cell survival rate of 89-95% and a transfection efficiency of 39-56% when introduced into animal cells, which are similar to those of other NLS vectors. Additionally, the constructed NLS vector can be used to demonstrate complementary binding between target proteins, and that the target protein is transported by the NLS transport system. Especially, we show that the vector can be useful for experiments involving the S100A10 gene. In addition, the constructed vector is useful for studies of genes and proteins that show potential for gene therapy or drug delivery applications.
Collapse
Affiliation(s)
- Hee Sang You
- 1Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, 77, Gyeryong-ro, 771 beon-gil, Jung-gu, Daejeon, 34824 Republic of Korea
- 2Department of Biomedical Laboratory Science, School of Medicine, Eulji University, 77, Gyeryong-ro, 771 beon-gil, Jung-gu, Daejeon, 34824 Republic of Korea
| | - Yeon Jeong Ok
- 2Department of Biomedical Laboratory Science, School of Medicine, Eulji University, 77, Gyeryong-ro, 771 beon-gil, Jung-gu, Daejeon, 34824 Republic of Korea
| | - Eun Jeong Lee
- 3Department of Biology Education, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644 Republic of Korea
| | - Sang Sun Kang
- 3Department of Biology Education, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644 Republic of Korea
| | - Sung Hee Hyun
- 1Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, 77, Gyeryong-ro, 771 beon-gil, Jung-gu, Daejeon, 34824 Republic of Korea
- 2Department of Biomedical Laboratory Science, School of Medicine, Eulji University, 77, Gyeryong-ro, 771 beon-gil, Jung-gu, Daejeon, 34824 Republic of Korea
| |
Collapse
|
13
|
Durymanov M, Reineke J. Non-viral Delivery of Nucleic Acids: Insight Into Mechanisms of Overcoming Intracellular Barriers. Front Pharmacol 2018; 9:971. [PMID: 30186185 PMCID: PMC6111240 DOI: 10.3389/fphar.2018.00971] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/06/2018] [Indexed: 12/27/2022] Open
Abstract
Delivery of genes, including plasmid DNAs, short interfering RNAs (siRNAs), and messenger RNAs (mRNAs), using artificial non-viral nanotherapeutics is a promising approach in cancer gene therapy. However, multiple physiological barriers upon systemic administration remain a key challenge in clinical translation of anti-cancer gene therapeutics. Besides extracellular barriers including sequestration of gene delivery nanoparticles from the bloodstream by resident organ-specific macrophages, and their poor extravasation and tissue penetration in tumors, overcoming intracellular barriers is also necessary for successful delivery of nucleic acids. Whereas for RNA delivery the endosomal barrier holds a key importance, transfer of DNA cargo additionally requires translocation into the nucleus. Better understanding of crossing membrane barriers by nucleic acid nanoformulations is essential to the improvement of current non-viral carriers. This review aims to summarize relevant literature on intracellular trafficking of non-viral nanoparticles and determine key factors toward surmounting intracellular barriers. Moreover, recent data allowed us to propose new interpretations of current hypotheses of endosomal escape mechanisms of nucleic acid nanoformulations.
Collapse
Affiliation(s)
- Mikhail Durymanov
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, South Dakota State University, Brookings, SD, United States
| | - Joshua Reineke
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, South Dakota State University, Brookings, SD, United States
| |
Collapse
|
14
|
Li X, Kang P, Chen Z, Lal S, Zhang L, Gassensmith JJ, Qin Z. Rock the nucleus: significantly enhanced nuclear membrane permeability and gene transfection by plasmonic nanobubble induced nanomechanical transduction. Chem Commun (Camb) 2018; 54:2479-2482. [DOI: 10.1039/c7cc09613e] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanomechanical transduction increases permeability of the nuclear membrane and facilitates nuclear uptake of macromolecules that would otherwise not enter the nucleus.
Collapse
Affiliation(s)
- Xiuying Li
- Department of Mechanical Engineering
- University of Texas at Dallas
- Richardson
- USA
| | - Peiyuan Kang
- Department of Mechanical Engineering
- University of Texas at Dallas
- Richardson
- USA
| | - Zhuo Chen
- Department of Chemistry and Biochemistry
- University of Texas at Dallas
- Richardson
- USA
| | - Sneha Lal
- Department of Biological Sciences
- University of Texas at Dallas
- Richardson
- USA
| | - Li Zhang
- Department of Biological Sciences
- University of Texas at Dallas
- Richardson
- USA
| | - Jeremiah J. Gassensmith
- Department of Chemistry and Biochemistry
- University of Texas at Dallas
- Richardson
- USA
- Department of Bioengineering, University of Texas at Dallas
| | - Zhenpeng Qin
- Department of Mechanical Engineering
- University of Texas at Dallas
- Richardson
- USA
- Department of Bioengineering, University of Texas at Dallas
| |
Collapse
|
15
|
Wang YJ, Zhou Q, Cao S, Hu B, Deng Q, Jiang N, Cui J. Efficient gene therapy with a combination of ultrasound‑targeted microbubble destruction and PEI/DNA/NLS complexes. Mol Med Rep 2017; 16:7685-7691. [PMID: 28944824 DOI: 10.3892/mmr.2017.7510] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 01/27/2017] [Indexed: 11/05/2022] Open
Abstract
Current strategies of gene transfection are not efficient at achieving a notable therapeutic effect. The aim of the present study was to combine ultrasound‑targeted microbubble destruction (UTMD) with a polyethylenimine/pEGFP‑N3 plasmid/nuclear localization sequence (PEI/DNA/NLS) complex gene delivery system, and evaluate the transfection efficiency of enhanced green fluorescent protein (EGFP) gene delivery to 293T cells using this system. The formation of PEI/DNA/NLS complexes and the protective effects of PEI/NLS were verified by gel electrophoresis. Solutions consisting of the plasmid alone, PEI/DNA complexes, PEI/DNA/NLS complexes, UTMD+DNA, UTMD+PEI/DNA complexes, and UTMD+PEI/DNA/NLS complexes were transduced into 293T cells via ultrasound irradiation. The expression of GFP was observed using an inverted microscope and transfection efficiency was detected by flow cytometry following 24 h incubation in vitro. Cell activity was detected using a Cell Counting kit (CCK)‑8 assay. Gel electrophoresis confirmed the formation of PEI/DNA/NLS complexes and demonstrated that PEI/NLS exhibited protective effects on plasmid integrity for a limited time. Inverted microscope observations revealed that a greater GFP signal was observed with the combined action of PEI/DNA/NLS complexes with UTMD, and flow cytometry analysis demonstrated the highest level of transfection efficiency in this group. In addition, the viability of the cells detected by CCK‑8 and treated with PEI/DNA/NLS complexes with UTMD was >80%. In conclusion, the combination of UTMD and PEI/DNA/NLS complexes was highly effective for the efficient transfection of 293T cells without causing excessive cell damage. This method may provide a novel and effective gene transduction system to be applied in clinical treatments.
Collapse
Affiliation(s)
- Yi-Jia Wang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing Zhou
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Sheng Cao
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bo Hu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing Deng
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Nan Jiang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jingjing Cui
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
16
|
He C, Wang S, Liu M, Zhao C, Xiang S, Zeng Y. Design, synthesis and in vitro evaluation of d-glucose-based cationic glycolipids for gene delivery. Org Biomol Chem 2016; 14:1611-22. [PMID: 26670704 DOI: 10.1039/c5ob02107c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A cationic lipid consists of a hydrophilic headgroup, backbone and hydrophobic tails which have an immense influence on the transfection efficiency of the lipid. In this paper, two novel series of cationic cyclic glycolipids with a quaternary ammonium headgroup and different-length hydrophobic tails (dodecyl, tetradecyl, hexadecyl) have been designed and synthesized for gene delivery. One contains lipids 1-3 with two hydrophobic alkyl chains linked to the glucose ring directly via an ether link. The other contains lipids 4-6 with two hydrophobic chains on the positively charged nitrogen atoms. All of the lipids were characterized for their ability to bind to DNA, size, ζ-potential, and toxicity. Atomic force microscopy showed that the lipids and DNA-lipid complexes were sphere-like forms. The lipids were used to transfer enhanced green fluorescent protein (EGFP-C3) to HEK293 cells without a helper lipid, the results indicated that lipids 4-6 have better transfection efficiency, in particular lipids 5-6 have similar or better efficiency, compared with the commercial transfection reagent lipofectamine 2000.
Collapse
Affiliation(s)
- Chengxi He
- National & Local Joint Engineering Laboratory for New Petro-chemical Materials and Fine Utilization of Resources, Hunan Normal University, Changsha, Hunan 410081, P. R. China.
| | - Shang Wang
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P. R. China
| | - Meiyan Liu
- National & Local Joint Engineering Laboratory for New Petro-chemical Materials and Fine Utilization of Resources, Hunan Normal University, Changsha, Hunan 410081, P. R. China.
| | - Chunyan Zhao
- National & Local Joint Engineering Laboratory for New Petro-chemical Materials and Fine Utilization of Resources, Hunan Normal University, Changsha, Hunan 410081, P. R. China.
| | - Shuanglin Xiang
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P. R. China
| | - Youlin Zeng
- National & Local Joint Engineering Laboratory for New Petro-chemical Materials and Fine Utilization of Resources, Hunan Normal University, Changsha, Hunan 410081, P. R. China.
| |
Collapse
|
17
|
Pasquet L, Bellard E, Rols MP, Golzio M, Teissie J. Post-pulse addition of trans-cyclohexane-1,2-diol improves electrotransfer mediated gene expression in mammalian cells. Biochem Biophys Rep 2016; 7:287-294. [PMID: 28955917 PMCID: PMC5613639 DOI: 10.1016/j.bbrep.2016.07.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/08/2016] [Accepted: 07/14/2016] [Indexed: 12/17/2022] Open
Abstract
Electric field mediated gene transfer is facing a problem in expression yield due to the poor transfer across the nuclear envelope. Trans-cyclohexane-1,2-diol (TCHD) was shown to significantly increase chemically mediated transfection by collapsing the permeability barrier of the nuclear pore complex. We indeed observed a significant increase in expression by electrotransfer when cells are treated post pulse by a low non toxic concentration of TCHD. This was obtained for different pulsing conditions, cell strains and plasmid constructs. An interesting improvement in cell viability can be obtained. This can significantly enhance the non-viral gene electrical delivery. Trans-cyclohexane-1,2-diol (TCHD) collapses the permeability barrier of the nuclear pore complex. TCHD improves expression in gene electrotransfer. Post pulse TCHD addition is the most effective protocol. TCHD does not affect the cell viability when coupled to electrotransfer.
Collapse
Affiliation(s)
- L Pasquet
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, BP64182, 205 route de Narbonne, F-31077 Toulouse, France.,Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, UPS, IPBS, F-31077 Toulouse, France
| | - E Bellard
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, BP64182, 205 route de Narbonne, F-31077 Toulouse, France.,Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, UPS, IPBS, F-31077 Toulouse, France
| | - M P Rols
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, BP64182, 205 route de Narbonne, F-31077 Toulouse, France.,Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, UPS, IPBS, F-31077 Toulouse, France
| | - M Golzio
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, BP64182, 205 route de Narbonne, F-31077 Toulouse, France.,Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, UPS, IPBS, F-31077 Toulouse, France
| | - J Teissie
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, BP64182, 205 route de Narbonne, F-31077 Toulouse, France.,Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, UPS, IPBS, F-31077 Toulouse, France
| |
Collapse
|
18
|
Kim JB, Lee YM, Ryu J, Lee E, Kim WJ, Keum G, Bang EK. Coordinative Amphiphiles as Tunable siRNA Transporters. Bioconjug Chem 2016; 27:1850-6. [DOI: 10.1021/acs.bioconjchem.6b00260] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jin Bum Kim
- Center
for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Yeong Mi Lee
- Center
for Self-Assembly and Complexity, Institute for Basic Science (IBS),
and Department of Chemistry, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang 37673, Republic of Korea
| | - Jooyeon Ryu
- Graduate
School of Analytical Science and Technology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Eunji Lee
- Graduate
School of Analytical Science and Technology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Won Jong Kim
- Center
for Self-Assembly and Complexity, Institute for Basic Science (IBS),
and Department of Chemistry, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang 37673, Republic of Korea
| | - Gyochang Keum
- Center
for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Eun-Kyoung Bang
- Center
for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
19
|
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: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
20
|
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: 0.8] [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
|
21
|
Swami R, Singh I, Khan W, Ramakrishna S. Diseases originate and terminate by genes: unraveling nonviral gene delivery. Drug Deliv Transl Res 2015; 3:593-610. [PMID: 25786377 DOI: 10.1007/s13346-013-0159-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The world is driving in to the era of transformation of chemical therapeutic molecules to biological genetic material therapeutics, and that is where the biological drugs especially "genes" come into existence. These genes worked as "magical bullets" to specifically silence faulty genes responsible for progression of diseases. Viral gene delivery research is far ahead of nonviral gene delivery technique. However, with more advancement in polymer science, new ways are opening for better and efficient nonviral gene delivery. But efficient delivery method is always considered as a bottleneck for gene delivery as success of which will decide the fate of gene in cells. During the past decade, it became evident that extracellular as well as intracellular barriers compromise the transfection efficiency of nonviral vectors. The challenge for gene therapy research is to pinpoint the rate-limiting steps in this complex process and implement strategies to overcome the biological physiochemical and metabolic barriers encountered during targeting. The synergy between studies that investigate the mechanism of breaking in and breaking out of nonviral gene delivery carrier through various extracellular and intracellular barriers with desired characteristics will enable the rational design of vehicles and revolutionize the treatment of various diseases.
Collapse
Affiliation(s)
- Rajan Swami
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
| | | | | | | |
Collapse
|
22
|
He D, Wagner E. Defined Polymeric Materials for Gene Delivery. Macromol Biosci 2015; 15:600-12. [PMID: 25655078 DOI: 10.1002/mabi.201400524] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 01/12/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Dongsheng He
- Pharmaceutical Biotechnology; Center for System-based Drug Research and Center for NanoScience (CeNS); Ludwig-Maximilians-University; 81377 Munich Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology; Center for System-based Drug Research and Center for NanoScience (CeNS); Ludwig-Maximilians-University; 81377 Munich Germany
| |
Collapse
|
23
|
Gahan PB. The Biology of CNAPS. ADVANCES IN PREDICTIVE, PREVENTIVE AND PERSONALISED MEDICINE 2015. [DOI: 10.1007/978-94-017-9168-7_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
24
|
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]
|
25
|
Kwilasz AJ, Grace PM, Serbedzija P, Maier SF, Watkins LR. The therapeutic potential of interleukin-10 in neuroimmune diseases. Neuropharmacology 2014; 96:55-69. [PMID: 25446571 DOI: 10.1016/j.neuropharm.2014.10.020] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 10/19/2014] [Accepted: 10/21/2014] [Indexed: 02/07/2023]
Abstract
Neuroimmune diseases have diverse symptoms and etiologies but all involve pathological inflammation that affects normal central nervous system signaling. Critically, many neuroimmune diseases also involve insufficient signaling/bioavailability of interleukin-10 (IL-10). IL-10 is a potent anti-inflammatory cytokine released by immune cells and glia, which drives the regulation of a variety of anti-inflammatory processes. This review will focus on the signaling pathways and function of IL-10, the current evidence for insufficiencies in IL-10 signaling/bioavailability in neuroimmune diseases, as well as the implications for IL-10-based therapies to treating such problems. We will review in detail four pathologies as examples of the common etiologies of such disease states, namely neuropathic pain (nerve trauma), osteoarthritis (peripheral inflammation), Parkinson's disease (neurodegeneration), and multiple sclerosis (autoimmune). A number of methods to increase IL-10 have been developed (e.g. protein administration, viral vectors, naked plasmid DNA, plasmid DNA packaged in polymers to enhance their uptake into target cells, and adenosine 2A agonists), which will also be discussed. In general, IL-10-based therapies have been effective at treating both the symptoms and pathology associated with various neuroimmune diseases, with more sophisticated gene therapy-based methods producing sustained therapeutic effects lasting for several months following a single injection. These exciting results have resulted in IL-10-targeted therapeutics being positioned for upcoming clinical trials for treating neuroimmune diseases, including neuropathic pain. Although further research is necessary to determine the full range of effects associated with IL-10-based therapy, evidence suggests IL-10 may be an invaluable target for the treatment of neuroimmune disease. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- A J Kwilasz
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado-Boulder, Boulder, CO 80309-0345, USA.
| | - P M Grace
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado-Boulder, Boulder, CO 80309-0345, USA
| | - P Serbedzija
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado-Boulder, Boulder, CO 80309-0345, USA
| | - S F Maier
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado-Boulder, Boulder, CO 80309-0345, USA
| | - L R Watkins
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado-Boulder, Boulder, CO 80309-0345, USA
| |
Collapse
|
26
|
Wong PT, Tang K, Coulter A, Tang S, Baker JR, Choi SK. Multivalent Dendrimer Vectors with DNA Intercalation Motifs for Gene Delivery. Biomacromolecules 2014; 15:4134-45. [PMID: 25285357 DOI: 10.1021/bm501169s] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Pamela T. Wong
- Michigan Nanotechnology Institute for Medicine
and Biological Sciences and ‡Department of
Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Kenny Tang
- Michigan Nanotechnology Institute for Medicine
and Biological Sciences and ‡Department of
Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Alexa Coulter
- Michigan Nanotechnology Institute for Medicine
and Biological Sciences and ‡Department of
Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Shengzhuang Tang
- Michigan Nanotechnology Institute for Medicine
and Biological Sciences and ‡Department of
Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - James R. Baker
- Michigan Nanotechnology Institute for Medicine
and Biological Sciences and ‡Department of
Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Seok Ki Choi
- Michigan Nanotechnology Institute for Medicine
and Biological Sciences and ‡Department of
Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
27
|
Pan J, Lyu Z, Jiang W, Wang H, Liu Q, Tan M, Yuan L, Chen H. Stimulation of gene transfection by silicon nanowire arrays modified with polyethylenimine. ACS APPLIED MATERIALS & INTERFACES 2014; 6:14391-14398. [PMID: 25032791 DOI: 10.1021/am5036626] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
In this work, a novel gene delivery strategy was proposed based on silicon nanowire arrays modified with high-molecular-weight 25 kDa branched polyethylenimine (SN-PEI). Both the plasmid DNA (pDNA) binding capacity and the in vitro gene transfection efficiency of silicon nanowire arrays (SiNWAs) were significantly enhanced after modification with high-molecular-weight bPEI. Moreover, the transfection efficiency was substantially further increased by the introduction of free pDNA/PEI complexes formed by low-molecular-weight branched PEI (bPEI, 2 kDa). Additionally, factors affecting the in vitro transfection efficiency of the novel gene delivery system were investigated in detail, and the transfection efficiency was optimized on SN-PEI with a bPEI grafting time of 3 h, an incubation time of 10 min for tethered pDNA/PEI complexes consisting of high-molecular-weight bPEI grafted onto SiNWAs, and with an N/P ratio of 80 for free pDNA/PEI complexes made of low-molecular-weight bPEI. Together, our results indicate that high-molecular-weight bPEI modified SiNWAs can serve as an efficient platform for gene delivery.
Collapse
Affiliation(s)
- Jingjing Pan
- The Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou 215123, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Malmevik J, Rogers ML, Nilsson M, Nakanishi Y, Rush RA, Sims NR, Muyderman H. Selective transfection of microglia in the brain using an antibody-based non-viral vector. Brain Res 2014; 1586:12-22. [PMID: 25150592 DOI: 10.1016/j.brainres.2014.08.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 08/07/2014] [Indexed: 01/07/2023]
Abstract
There are currently few approaches to transiently manipulate the expression of specific proteins in microglia of the brain. An antibody directed against an extracellular epitope of scavenger receptor class B, type I (SR-BI) was found to be selectively taken up by these cells in the brain. Other antibodies tested were not internalised by microglia. A vector was produced by linking the SR-BI antibody to polyethyleneimine and binding a DNA plasmid encoding green fluorescent protein. Infusions of this vector into the hippocampus produced a widespread transfection of cells, more than 80% of which were immunoreactive for microglial/macrophage markers. Transfection was not detected in cells expressing markers for astrocytes or neurons. Reporter gene expression was most prominent near the infusion site but was seen in tissue up to 4mm away. DNA bound to polyethyleneimine alone or to a vector containing a different antibody did not produce transfection in the brain. Single injections of the vector containing the SR-BI antibody into the brain also resulted in transfection of microglia, albeit with lower efficiency. Vector modifications to promote lysis of endosomes or entry of DNA into the nucleus did not increase efficiency. The findings clearly demonstrate the capacity of the SR-BI antibody to selectively target brain microglia. This approach offers considerable potential to deliver DNA and other molecules capable of modifying the function of these cells in vivo.
Collapse
Affiliation(s)
- J Malmevik
- Centre for Neuroscience, Flinders Medical Science and Technology, Flinders University, GPO Box 2100, Adelaide SA 5001, Australia
| | - M-L Rogers
- Centre for Neuroscience, Flinders Medical Science and Technology, Flinders University, GPO Box 2100, Adelaide SA 5001, Australia
| | - M Nilsson
- Hunter Medical Research Institute (HMRI), University of Newcastle, Australia; Centre for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Sweden
| | - Y Nakanishi
- Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - R A Rush
- Centre for Neuroscience, Flinders Medical Science and Technology, Flinders University, GPO Box 2100, Adelaide SA 5001, Australia
| | - N R Sims
- Centre for Neuroscience, Flinders Medical Science and Technology, Flinders University, GPO Box 2100, Adelaide SA 5001, Australia
| | - H Muyderman
- Centre for Neuroscience, Flinders Medical Science and Technology, Flinders University, GPO Box 2100, Adelaide SA 5001, Australia.
| |
Collapse
|
29
|
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
|
30
|
Mishra D, Kang HC, Cho H, Bae YH. Dexamethasone-loaded reconstitutable charged polymeric (PLGA)n -b-bPEI micelles for enhanced nuclear delivery of gene therapeutics. Macromol Biosci 2014; 14:831-41. [PMID: 24550091 DOI: 10.1002/mabi.201300432] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 01/09/2014] [Indexed: 11/06/2022]
Abstract
This study investigates the potential of dexamethasone (Dex) to enhance the nuclear accumulation and subsequent gene expression of plasmid DNA (pDNA) delivered using a charged polymeric micelle-based gene delivery system. (PLGA)n -b-bPEI25kDa block copolymers are synthesized and used to prepare Dex-loaded cationic micelles (DexCM). After preparing DexCM/pDNA complexes, bPEI1.8kDa is coated on the complexes using a Layer-by-Layer (LbL) technique to construct DexCM/pDNA/bPEI1.8kDa complexes (i.e., LbL-DexCM polyplexes) that are 100-180 nm in diameter and have a zeta potential of 30-40 mV. In MCF7 cells, LbL-DexCM polyplexes cause 3-13-fold higher transfection efficiencies compared to LbL-CM polyplexes and show negligible cytotoxicity. LbL-DexCM3 polyplexes induce much higher nuclear delivery of pDNA compared to LbL-CM3 polyplexes. These results suggest that Dex-loaded polyplexes could be used in gene and drug delivery applications to increase nuclear accumulation of therapeutic payloads, further leading to a decrease in the dose of the drug and gene necessary to achieve equivalent therapeutic effects.
Collapse
Affiliation(s)
- Deepa Mishra
- Department of Bioengineering, The University of Utah, 20 S. 2030 E., Rm. 108, Salt Lake City, Utah, 84112, USA
| | | | | | | |
Collapse
|
31
|
Affiliation(s)
- Peter Gahan
- Anatomy & Human Sciences; King's College London; London Bridge London SE1 1UL UK
| |
Collapse
|
32
|
Kawazu T, Kanzaki H, Uno A, Azuma H, Nagasaki T. HVJ-E/importin-β hybrid vector for overcoming cytoplasmic and nuclear membranes as double barrier for non-viral gene delivery. Biomed Pharmacother 2012; 66:519-24. [DOI: 10.1016/j.biopha.2012.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 02/29/2012] [Indexed: 10/28/2022] Open
|
33
|
Abstract
This article focuses on drug targeting to specific cellular organelles for therapeutic purposes. Drugs can be delivered to all major organelles of the cell (cytosol, endosome/lysosome, nucleus, nucleolus, mitochondria, endoplasmic reticulum, Golgi apparatus, peroxisomes and proteasomes) where they exert specific effects in those particular subcellular compartments. Delivery can be achieved by chemical (e.g., polymeric) or biological (e.g., signal sequences) means. Unidirectional targeting to individual organelles has proven to be immensely successful for drug therapy. Newer technologies that accommodate multiple signals (e.g., protein switch and virus-like delivery systems) mimic nature and allow for a more sophisticated approach to drug delivery. Harnessing different methods of targeting multiple organelles in a cell will lead to better drug delivery and improvements in disease therapy.
Collapse
|
34
|
Matschke J, Bohla A, Maucksch C, Mittal R, Rudolph C, Rosenecker J. Characterization of Ku70(2)-NLS as bipartite nuclear localization sequence for non-viral gene delivery. PLoS One 2012; 7:e24615. [PMID: 22347357 PMCID: PMC3275586 DOI: 10.1371/journal.pone.0024615] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Accepted: 08/16/2011] [Indexed: 11/24/2022] Open
Abstract
Several barriers have to be overcome in order to achieve gene expression in target cells, e.g. cellular uptake, endosomal release and translocation to the nucleus. Nuclear localization sequences (NLS) enhance gene delivery by increasing the uptake of plasmid DNA (pDNA) to the nucleus. So far, only monopartite NLS were analysed for non-viral gene delivery. In this study, we examined the characteristics of a novel bipartite NLS like construct, namely NLS Ku70. We synthesized a dimeric structure of a modified NLS from the Ku70 protein (Ku702-NLS), a nuclear transport active mutant of Ku702-NLS (s1Ku702-NLS) and a nuclear transport deficient mutant of Ku702-NLS (s2Ku702). We examined the transfection efficiency of binary Ku702-NLS/DNA and ternary Ku702-NLS/PEI/DNA gene vector complexes in vitro by using standard transfection protocols as well as the magnetofection method. The application of Ku702-NLS and s1Ku702-NLS increased gene transfer efficiency in vitro and in vivo. This study shows for the first time that the use of bipartite NLS compounds alone or in combination with cationic polymers is a promising strategy to enhance the efficiency of non-viral gene transfer.
Collapse
Affiliation(s)
- Johannes Matschke
- Department of Pediatrics, Ludwig-Maximilians University, Munich, Germany
| | - Alexander Bohla
- Department of Pediatrics, Ludwig-Maximilians University, Munich, Germany
- Institute of Animal Physiology, University of Muenster, Muenster, Germany
| | - Christof Maucksch
- Department of Pediatrics, Ludwig-Maximilians University, Munich, Germany
- Department of Pharmacy, Free University of Berlin, Berlin, Germany
| | - Rashmi Mittal
- Department of Neonatology, Ludwig-Maximilians University, Munich, Germany
| | - Carsten Rudolph
- Department of Pediatrics, Ludwig-Maximilians University, Munich, Germany
- Department of Pharmacy, Free University of Berlin, Berlin, Germany
| | - Joseph Rosenecker
- Department of Pediatrics, Ludwig-Maximilians University, Munich, Germany
- * E-mail:
| |
Collapse
|
35
|
Yamada Y, Akita H, Harashima H. Multifunctional Envelope-Type Nano Device (MEND) for Organelle Targeting Via a Stepwise Membrane Fusion Process. Methods Enzymol 2012; 509:301-26. [DOI: 10.1016/b978-0-12-391858-1.00015-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
36
|
Symens N, Soenen SJ, Rejman J, Braeckmans K, De Smedt SC, Remaut K. Intracellular partitioning of cell organelles and extraneous nanoparticles during mitosis. Adv Drug Deliv Rev 2012; 64:78-94. [PMID: 22210278 DOI: 10.1016/j.addr.2011.11.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 11/21/2011] [Accepted: 11/23/2011] [Indexed: 02/06/2023]
Abstract
The nucleocytoplasmic partitioning of nanoparticles as a result of cell division is highly relevant to the field of nonviral gene delivery. We reviewed the literature on the intracellular distribution of cell organelles (the endosomal vesicles, Golgi apparatus, endoplasmic reticulum and nucleus), foreign macromolecules (dextrans and plasmid DNA) and inorganic nanoparticles (gold, quantum dot and iron oxide) during mitosis. For nonviral gene delivery particles (lipid- or polymer-based), indirect proof of nuclear entry during mitosis is provided. We also describe how retroviruses and latent DNA viruses take advantage of mitosis to transfer their viral genome and segregate their episomes into the host daughter nuclei. Based on this knowledge, we propose strategies to improve nonviral gene delivery in dividing cells with the ultimate goal of designing nonviral gene delivery systems that are as efficient as their viral counterparts but non-immunogenic, non-oncogenic and easy and inexpensive to prepare.
Collapse
Affiliation(s)
- Nathalie Symens
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium.
| | | | | | | | | | | |
Collapse
|
37
|
Kievit FM, Zhang M. Cancer nanotheranostics: improving imaging and therapy by targeted delivery across biological barriers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2011; 23:H217-47. [PMID: 21842473 PMCID: PMC3397249 DOI: 10.1002/adma.201102313] [Citation(s) in RCA: 350] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 07/12/2011] [Indexed: 05/03/2023]
Abstract
Cancer nanotheranostics aims to combine imaging and therapy of cancer through use of nanotechnology. The ability to engineer nanomaterials to interact with cancer cells at the molecular level can significantly improve the effectiveness and specificity of therapy to cancers that are currently difficult to treat. In particular, metastatic cancers, drug-resistant cancers, and cancer stem cells impose the greatest therapeutic challenge for targeted therapy. Targeted therapy can be achieved with appropriately designed drug delivery vehicles such as nanoparticles, adult stem cells, or T cells in immunotherapy. In this article, we first review the different types of nanotheranostic particles and their use in imaging, followed by the biological barriers they must bypass to reach the target cancer cells, including the blood, liver, kidneys, spleen, and particularly the blood-brain barrier. We then review how nanotheranostics can be used to improve targeted delivery and treatment of cancer cells. Finally, we discuss development of nanoparticles to overcome current limitations in cancer therapy.
Collapse
Affiliation(s)
- Forrest M Kievit
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
38
|
Symens N, Walczak R, Demeester J, Mattaj I, De Smedt SC, Remaut K. Nuclear inclusion of nontargeted and chromatin-targeted polystyrene beads and plasmid DNA containing nanoparticles. Mol Pharm 2011; 8:1757-66. [PMID: 21859089 DOI: 10.1021/mp200120v] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The nuclear membrane is one of the major cellular barriers in the delivery of plasmid DNA (pDNA). Cell division has a positive influence on the expression efficiency since, at the end of mitosis, pDNA or pDNA containing complexes near the chromatin are probably included by a random process in the nuclei of the daughter cells. However, very little is known about the nuclear inclusion of nanoparticles during cell division. Using the Xenopus nuclear envelope reassembly (XNER) assay, we found that the nuclear enclosure of nanoparticles was dependent on size (with 100 and 200 nm particles being better included than the 500 nm ones) and charge (with positively charged particles being better included than negatively charged or polyethyleneglycolated (PEGylated) ones) of the beads. Also, coupling chromatin-targeting peptides to the polystyrene beads or pDNA complexes improved their inclusion by 2- to 3-fold. Upon microinjection in living HeLa cells, however, nanoparticles were never observed in the nuclei of cells postdivision but accumulated in a specific perinuclear region, which was identified as the lysosomal compartment. This indicates that nanoparticles can end up in the lysosomes even when they were not delivered through endocytosis. To elucidate if the chromatin binding peptides also have potential in living cells, this additional barrier first has to be tackled, since it prevents free particles from being present near the chromatin at the moment of cell division.
Collapse
Affiliation(s)
- Nathalie Symens
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicines, Faculty of Pharmaceutical Sciences, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium
| | | | | | | | | | | |
Collapse
|
39
|
Akita H, Masuda T, Nishio T, Niikura K, Ijiro K, Harashima H. Improving in Vivo Hepatic Transfection Activity by Controlling Intracellular Trafficking: The Function of GALA and Maltotriose. Mol Pharm 2011; 8:1436-42. [PMID: 21598999 DOI: 10.1021/mp200189s] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Hidetaka Akita
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Kita-Ku, Sapporo, Hokkaido, 060-0812 Japan
| | - Tomoya Masuda
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Kita-Ku, Sapporo, Hokkaido, 060-0812 Japan
| | - Takashi Nishio
- Research Institute for Electronic Science, Hokkaido University, Kita 21 Nishi 10, Kita-Ku, Sapporo, Hokkaido, 001-0021, Japan
| | - Kenichi Niikura
- Research Institute for Electronic Science, Hokkaido University, Kita 21 Nishi 10, Kita-Ku, Sapporo, Hokkaido, 001-0021, Japan
| | - Kuniharu Ijiro
- Research Institute for Electronic Science, Hokkaido University, Kita 21 Nishi 10, Kita-Ku, Sapporo, Hokkaido, 001-0021, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Kita-Ku, Sapporo, Hokkaido, 060-0812 Japan
| |
Collapse
|
40
|
van Gaal EVB, Oosting RS, van Eijk R, Bakowska M, Feyen D, Kok RJ, Hennink WE, Crommelin DJA, Mastrobattista E. DNA nuclear targeting sequences for non-viral gene delivery. Pharm Res 2011; 28:1707-22. [PMID: 21424159 PMCID: PMC3109246 DOI: 10.1007/s11095-011-0407-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 02/21/2011] [Indexed: 12/02/2022]
Abstract
Purpose To evaluate if introduction of DNA nuclear Targeting Sequences (DTS; i.e. recognition sequences for endogenous DNA-binding proteins) in plasmid DNA (pDNA) leads to increased transfection efficiency of non-viral gene delivery by virtue of enhanced nuclear import of the pDNA. Methods A set of DTS was identified and cloned into EGFP-reporter plasmids controlled by the CMV-promoter. These pDNA constructs were delivered into A431 and HeLa cells using standard electroporation, pEI-based polyfection or lipofection methods. The amount of pDNA delivered into the nucleus was determined by qPCR; transfection efficiency was determined by flow cytometry. Results Neither of these DTS increased transgene expression. We varied several parameters (mitotic activity, applied dose and delivery strategy), but without effect. Although upregulated transgene expression was observed after stimulation with TNF-α, this effect could be ascribed to non-specific upregulation of transcription rather than enhanced nuclear import. Nuclear copy numbers of plasmids containing or lacking a DTS did not differ significantly after lipofectamine-based transfection in dividing and non-dividing cells. Conclusion No beneficial effects of DTS on gene expression or nuclear uptake were observed in this study.
Collapse
Affiliation(s)
- Ethlinn V B van Gaal
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, 3584 CA, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Tavernier G, Andries O, Demeester J, Sanders NN, De Smedt SC, Rejman J. mRNA as gene therapeutic: How to control protein expression. J Control Release 2011; 150:238-47. [DOI: 10.1016/j.jconrel.2010.10.020] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 10/13/2010] [Indexed: 10/18/2022]
|
42
|
Won YW, Lim KS, Kim YH. Intracellular organelle-targeted non-viral gene delivery systems. J Control Release 2011; 152:99-109. [PMID: 21255626 DOI: 10.1016/j.jconrel.2011.01.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 12/30/2010] [Accepted: 01/07/2011] [Indexed: 10/18/2022]
Abstract
Gene therapy is a rapidly growing approach for the treatment of various diseases. To achieve successful gene therapy, a gene delivery system is necessary to overcome several barriers in the extracellular and intracellular spaces. Polymers, peptides, liposomes and nanoparticles developed as gene carriers have achieved efficient cellular uptake of genes. Among these carriers, cationic polymers and peptides have been further developed as intracellular organelle-targeted delivery systems. The cytoplasm, nucleus and mitochondria have been considered primary targets for gene delivery using targeting moieties or environment-responsive materials. In this review, we explore recently developed non-viral gene carriers based on reducible systems specialized to target the cytoplasm, nucleus and mitochondria.
Collapse
Affiliation(s)
- Young-Wook Won
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, and Institute of Aging Society, Hanyang University, 17 Haengdang-dong, Seongdong-gu, Seoul 133-791, Republic of Korea
| | | | | |
Collapse
|
43
|
Gene therapy: a pharmacokinetic/pharmacodynamic modelling overview. Pharm Res 2010; 27:1487-97. [PMID: 20387096 DOI: 10.1007/s11095-010-0136-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 03/24/2010] [Indexed: 12/20/2022]
Abstract
Since gene therapy started over 20 years ago, more than one-thousand clinical trials have been carried out. Nonviral vectors present interesting properties for their clinical application, but their efficiency in vivo is relatively low, and further improvements in these vectors are needed. Elucidating how nonviral vectors behave at the intracellular level is enlightening for vector improvement and optimization. Model-based approach is a powerful tool to understand and describe the different processes that gene transfer systems should overcome inside the body. Model-based approach allows for proposing and predicting the effect of parameter changes on the overall gene therapy response, as well as the known application of the pharmacokinetic/pharmacodynamic modelling in conventional therapies. The objective of this paper is to critically review the works in which the time-course of naked or formulated DNA have been quantitatively studied or modelled.
Collapse
|
44
|
Yamada Y, Nomura T, Harashima H, Yamashita A, Katoono R, Yui N. Intranuclear DNA Release Is a Determinant of Transfection Activity for a Non-viral Vector: Biocleavable Polyrotaxane as a Supramolecularly Dissociative Condenser for Efficient Intranuclear DNA Release. Biol Pharm Bull 2010; 33:1218-22. [DOI: 10.1248/bpb.33.1218] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yuma Yamada
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Taku Nomura
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Hideyoshi Harashima
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Atsushi Yamashita
- School of Materials Science, Japan Advanced Institute of Science and Technology
| | - Ryo Katoono
- School of Materials Science, Japan Advanced Institute of Science and Technology
| | - Nobuhiko Yui
- School of Materials Science, Japan Advanced Institute of Science and Technology
| |
Collapse
|
45
|
SHI Y, LIU XH, LIANG DS, FENG M, WU LQ, YANG JL, LI Z, ZHAO K, PAN Q, LONG ZG, XIA JH. The Transfection Efficiency Improvement of hrDNA Targeting Vectors With NLS Peptide*. PROG BIOCHEM BIOPHYS 2009. [DOI: 10.3724/sp.j.1206.2009.00174] [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]
|
46
|
Kim TI, Lee M, Kim SW. A guanidinylated bioreducible polymer with high nuclear localization ability for gene delivery systems. Biomaterials 2009; 31:1798-804. [PMID: 19854504 DOI: 10.1016/j.biomaterials.2009.10.034] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Accepted: 10/12/2009] [Indexed: 11/19/2022]
Abstract
Guanidinylated bioreducible polymer (GBP) was developed for gene delivery systems utilizing cellular penetrating ability of guanidine groups. GBP could retard pDNA from a weight ratio of 5 completely in agarose gel electrophoresis but pDNA was released from GBP polyplexes even at a weight ratio of 20 in reducing condition (2.5mm DTT) due to their biodegradation. GBP also could construct 200 nm-sized and positively charged (approximately 30 mV) polyplex nanoparticles with pDNA. The cytotoxicity of GBP was found to be minimal and GBP showed about 8 folds improved transfection efficiency than a scaffold polymer, poly(cystaminebisacrylamide-diaminohexane) (poly(CBA-DAH)) and even higher transfection efficiency than PEI25k in mammalian cell lines. Its high cellular uptake efficiency (96.1%) and strong nuclear localization ability for pDNA delivery due to the structural advantage of bioreducible polymer and guanidine groups were also identified, suggesting GBP is a promising candidate for efficient gene delivery systems.
Collapse
Affiliation(s)
- Tae-il Kim
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA.
| | | | | |
Collapse
|
47
|
Babiuk S, Babiuk LA, van Drunen Littel-van den Hurk S. Editorial: DNA Vaccination: A Simple Concept with Challenges Regarding Implementation. Int Rev Immunol 2009; 25:51-81. [PMID: 16818365 DOI: 10.1080/08830180600743008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
48
|
|
49
|
Miller AM, Dean DA. Tissue-specific and transcription factor-mediated nuclear entry of DNA. Adv Drug Deliv Rev 2009; 61:603-13. [PMID: 19393704 DOI: 10.1016/j.addr.2009.02.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2008] [Accepted: 02/05/2009] [Indexed: 12/22/2022]
Abstract
Low levels of gene transfer and a lack of tissue-specific targeting of vectors have limited the therapeutic potential of non-viral gene therapy. This is due to the numerous cellular barriers that hinder nuclear delivery of vectors and the paucity of methods that restrict expression to specific cells types. In non-dividing cells, the nuclear envelope is an especially problematic hurdle to gene transfer. Given that the majority of target tissues are non-dividing in vivo, the nuclear membrane is a major obstacle to therapeutic gene transfer. In this review, the various barriers to gene transfer are discussed. In particular, the role of the nuclear pore complex (NPC) in regulating passage of plasmid vectors during interphase is reviewed. Several methods of modifying plasmid (pDNA) vectors to enhance nuclear import through the NPC are also discussed, including the use of tissue-specific transcription factors to mediate nuclear entry of pDNA in a cell-specific manner.
Collapse
Affiliation(s)
- Aaron M Miller
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | |
Collapse
|
50
|
|