51
|
Li J, Crowley ST, Duskey J, Khargharia S, Wu M, Rice KG. Miniaturization of gene transfection assays in 384- and 1536-well microplates. Anal Biochem 2015; 470:14-21. [PMID: 25448623 PMCID: PMC4601643 DOI: 10.1016/j.ab.2014.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 10/01/2014] [Accepted: 10/03/2014] [Indexed: 11/20/2022]
Abstract
The miniaturization of gene transfer assays to either 384- or 1536-well plates greatly economizes the expense and allows much higher throughput when transfecting immortalized and primary cells compared with more conventional 96-well assays. To validate the approach, luciferase and green fluorescent protein (GFP) reporter gene transfer assays were developed to determine the influence of cell seeding number, transfection reagent to DNA ratios, transfection time, DNA dose, and luciferin dose on linearity and sensitivity. HepG2, CHO, and NIH 3T3 cells were transfected with polyethylenimine (PEI)-DNA in both 384- and 1536-well plates. The results established optimal transfection parameters in 384-well plates in a total assay volume of 35μl and in 1536-well plates in a total assay volume of 8μl. A luciferase assay performed in 384-well plates produced a Z' score of 0.53, making it acceptable for high-throughput screening. Primary hepatocytes were harvested from mouse liver and transfected with PEI DNA and calcium phosphate DNA nanoparticles in 384-well plates. Optimal transfection of primary hepatocytes was achieved on as few as 250cellsperwell in 384-well plates, with CaPO4 proving to be 10-fold more potent than PEI.
Collapse
Affiliation(s)
- Jing Li
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Samuel T Crowley
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Jason Duskey
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Sanjib Khargharia
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Meng Wu
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA; Department of Biochemistry, University of Iowa, Iowa City, IA 52242, USA; University of Iowa High Throughput Screening Facility, 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
|
52
|
Chen S, Rong L, Jia HZ, Qin SY, Zeng X, Zhuo RX, Zhang XZ. Co-delivery of proapoptotic peptide and p53 DNA by reduction-sensitive polypeptides for cancer therapy. Biomater Sci 2015. [DOI: 10.1039/c5bm00046g] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The xPolyR8–KLA(TPP)/p53 complex releases the p53 gene and C-KLA(TPP) in the cytoplasm, and initiates a more efficient cell apoptosis due to the regulation of both apoptotic pathways through p53 and C-KLA(TPP).
Collapse
Affiliation(s)
- Si Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Lei Rong
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Hui-Zhen Jia
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Si-Yong Qin
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Xuan Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Ren-Xi Zhuo
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- PR China
| |
Collapse
|
53
|
Zhou J, Li Y, Dong H, Yuan H, Ren T, Li Y. Effect of monomer sequence of poly(histidine/lysine) catiomers on gene packing capacity and delivery efficiency. RSC Adv 2015. [DOI: 10.1039/c4ra13785j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
This work presents a novel method to synthesize reducible polycations with specific monomer sequence, and provides new insight on how a monomer sequence of the polymeric catiomer will affect its gene packing capacity and delivery efficiency.
Collapse
Affiliation(s)
- Jiashan Zhou
- School of Materials and Engineering
- Tongji University
- Shanghai 200092
- China
| | - Yan Li
- Shanghai East Hospital
- The Institute for Biomedical Engineering and Nano Science
- Tongji University School of Medicine
- Shanghai 200120
- China
| | - Haiqing Dong
- Shanghai East Hospital
- The Institute for Biomedical Engineering and Nano Science
- Tongji University School of Medicine
- Shanghai 200120
- China
| | - Hua Yuan
- School of Materials and Engineering
- Tongji University
- Shanghai 200092
- China
| | - Tianbin Ren
- School of Materials and Engineering
- Tongji University
- Shanghai 200092
- China
| | - Yongyong Li
- Shanghai East Hospital
- The Institute for Biomedical Engineering and Nano Science
- Tongji University School of Medicine
- Shanghai 200120
- China
| |
Collapse
|
54
|
Klein PM, Wagner E. Bioreducible polycations as shuttles for therapeutic nucleic acid and protein transfection. Antioxid Redox Signal 2014; 21:804-17. [PMID: 24219092 PMCID: PMC4098974 DOI: 10.1089/ars.2013.5714] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 11/12/2013] [Indexed: 01/05/2023]
Abstract
SIGNIFICANCE Nucleic acids such as gene-encoding DNAs, gene-silencing small interfering RNAs, or recombinant proteins addressing intracellular molecular targets present a major new therapeutic modality, provided efficient solutions for intracellular delivery can be found. The different physiological redox environments inside and outside the cell can be utilized for optimizing the involved transport processes. RECENT ADVANCES Intracellular delivery of nucleic acids or proteins requires dynamic carriers that discriminate between different cellular locations. Bioreducible cationic polymers can package their therapeutic cargo stably in the extracellular environment, but sense the reducing intracellular cytosolic environment. Based on disulfide cleavage, carriers are degraded into biocompatible fragments and release the cargo in functional form. Disulfide linkages between oligocations, between the carrier and the cargo, or spatial caging of complexed cargo by disulfides have been pursued, with polymers or precise sequence-defined peptides and oligomers. CRITICAL ISSUES A quantitative knowledge of the bioreductive capacities within different biological compartments and the involved cellular reduction processes would be greatly helpful for improved carriers with disulfides cleaved within the right compartment at the right time. FUTURE DIRECTIONS Novel designs of multifunctional nanocarriers will incorporate macromolecular disulfide entry mechanisms previously optimized by natural evolution of toxins and viruses. In addition to extracellular stabilization and intracellular disassembly, tuned disulfides will contribute to deshielding at the cell surface, or translocation from intracellular compartments to the cytosol.
Collapse
Affiliation(s)
- Philipp M. Klein
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-University, Munich, Germany
- Center for Nanoscience (CeNS), Ludwig-Maximilians-University, Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-University, Munich, Germany
- Center for Nanoscience (CeNS), Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
55
|
Oupický D, Li J. Bioreducible polycations in nucleic acid delivery: past, present, and future trends. Macromol Biosci 2014; 14:908-22. [PMID: 24678057 PMCID: PMC4410047 DOI: 10.1002/mabi.201400061] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 02/19/2014] [Indexed: 12/16/2022]
Abstract
Polycations that are degradable by reduction of disulfide bonds are developed for applications in delivery of nucleic acids. This Feature Article surveys methods of synthesis of bioreducible polycations and discusses current understanding of the mechanism of action of bioreducible polyplexes. Emphasis is placed on the relationship between the biological redox environment and toxicity, trafficking, transfection activity, and in vivo behavior of bioreducible polycations and polyplexes.
Collapse
Affiliation(s)
- David Oupický
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Durham Research Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA.
| | | |
Collapse
|
56
|
Raad MD, Teunissen EA, Mastrobattista E. Peptide vectors for gene delivery: from single peptides to multifunctional peptide nanocarriers. Nanomedicine (Lond) 2014; 9:2217-32. [DOI: 10.2217/nnm.14.90] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The therapeutic use of nucleic acids relies on the availability of sophisticated delivery systems for targeted and intracellular delivery of these molecules. Such a gene delivery should possess essential characteristics to overcome several extracellular and intracellular barriers. Peptides offer an attractive platform for nonviral gene delivery, as several functional peptide classes exist capable of overcoming these barriers. However, none of these functional peptide classes contain all the essential characteristics required to overcome all of the barriers associated with successful gene delivery. Combining functional peptides into multifunctional peptide vectors will be pivotal for improving peptide-based gene delivery systems. By using combinatorial strategies and high-throughput screening, the identification of multifunctional peptide vectors will accelerate the optimization of peptide-based gene delivery systems.
Collapse
Affiliation(s)
- Markus de Raad
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, University of Utrecht, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Erik A Teunissen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, University of Utrecht, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, University of Utrecht, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
57
|
Sharma R, Nisakar D, Shivpuri S, Ganguli M. Contrasting effects of cysteine modification on the transfection efficiency of amphipathic peptides. Biomaterials 2014; 35:6563-75. [PMID: 24816284 DOI: 10.1016/j.biomaterials.2014.04.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 04/13/2014] [Indexed: 12/20/2022]
Abstract
Delivery of DNA to cells remains a key challenge towards development of gene therapy. A better understanding of the properties involved in stability and transfection efficiency of the vector could critically contribute to the improvement of delivery vehicles. In the present work we have chosen two peptides differing only in amphipathicity and explored how presence of cysteine affects DNA uptake and transfection efficiency. We report an unusual observation that addition of cysteine selectively increases transfection efficiency of secondary amphipathic peptide (Mgpe-9) and causes a drop in the primary amphipathic peptide (Mgpe-10). Our results point the effect of cysteine is dictated by the importance of physicochemical properties of the carrier peptide. We also report a DNA delivery agent Mgpe-9 exhibiting high transfection efficiency in multiple cell lines (including hard-to-transfect cell lines) with minimal cytotoxicity which can be further explored for in vivo applications.
Collapse
Affiliation(s)
- Rajpal Sharma
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110 020, India
| | - Daniel Nisakar
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110 020, India
| | - Shivangi Shivpuri
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110 020, India
| | - Munia Ganguli
- CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110 020, India.
| |
Collapse
|
58
|
Battogtokh G, Ko YT. Self-assembling micelle-like nanoparticles with detachable envelopes for enhanced delivery of nucleic acid therapeutics. Mol Pharm 2014; 11:904-12. [PMID: 24446871 DOI: 10.1021/mp400579h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In spite of the great potential of nucleic acids as therapeutic agents, the clinical application of nucleic acid therapeutics requires the development of effective systemic delivery strategies. In an effort to develop effective nucleic acid delivery systems suitable for clinical application, we previously reported a self-assembling micelle-like nanoparticle that was based on phospholipid-polyethylenimine conjugates, i.e., "micelle-like nanoparticles" (MNPs). In this study, we aimed to improve the system by enhancing the efficiency of intracellular delivery of the payload via pH-responsive detachment of the monolayer envelope and release of the nucleic acid therapeutics upon reaching the target tissues with an acidic pH, e.g., tumors. The acid-cleavable phospholipid-polyethylenimine conjugate was synthesized via hydrazone bond, and acid-cleavable MNPs were then prepared and characterized as before. We evaluated the acid-cleavable MNP construct for in vitro and in vivo nucleic acid delivery efficiency using cultured tumor cells and tumor-bearing mice. The acid-cleavable nanocarrier showed an enhanced cellular delivery at pH 6.5 as compared to pH 7.4, whereas the noncleavable nanocarrier did not show any differences. Tail vein injections also led to enhanced intracellular uptake of the acid-cleavable nanocarrier compared to the noncleavable nanocarrier into tumor cells of tumor-bearing mice although no significant difference was observed in total tumor accumulation.
Collapse
|
59
|
Abstract
For the last five decades cationic polymers have been used for nucleic acids transfection. Our understanding of polymer-nucleic acid interactions and their rational use in delivery has continuously increased. The great improvements in macromolecular chemistry and the recognition of distinct biological extra- and intracellular delivery hurdles triggered several breakthrough developments, including the discovery of natural and synthetic polycations for compaction of nucleic acids into stable nanoparticles termed polyplexes; the incorporation of targeting ligands and surface-shielding of polyplexes to enable receptor-mediated gene delivery into defined target tissues; and strongly improved intracellular transfer efficacy by better endosomal escape of vesicle-trapped polyplexes into the cytosol. These experiences triggered the development of second-generation polymers with more dynamic properties, such as endosomal pH-responsive release mechanisms, or biodegradable units for improved biocompatibility and intracellular release of the nucleic acid pay load. Despite a better biological understanding, significant challenges such as efficient nuclear delivery and persistence of gene expression persist. The therapeutic perspectives widened from pDNA-based gene therapy to application of novel therapeutic nucleic acids including mRNA, siRNA, and microRNA. The finding that different therapeutic pay loads require different tailor-made carriers complicates preclinical developments. Convincing evidence of medical efficacy still remains to be demonstrated. Bioinspired multifunctional polyplexes resembling "synthetic viruses" appear as attractive opportunity, but provide additional challenges: how to identify optimum combinations of functional delivery units, and how to prepare such polyplexes reproducibly in precise form? Design of sequence-defined polymers, screening of combinatorial polymer and polyplex libraries are tools for further chemical evolution of polyplexes.
Collapse
Affiliation(s)
- Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-University Munich, and Nanosystems Initiative Munich (NIM), Munich, Germany
| |
Collapse
|
60
|
Parhiz H, Shier WT, Ramezani M. From rationally designed polymeric and peptidic systems to sophisticated gene delivery nano-vectors. Int J Pharm 2013; 457:237-59. [PMID: 24060371 DOI: 10.1016/j.ijpharm.2013.09.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Revised: 08/21/2013] [Accepted: 09/17/2013] [Indexed: 12/12/2022]
Abstract
Lack of safe, efficient and controllable methods for delivering therapeutic genes appears to be the most important factor preventing human gene therapy. Safety issues encountered with viral vectors have prompted substantial attention to in vivo investigations with non-viral vectors throughout the past decade. However, developing non-viral vectors with effectiveness comparable to viral ones has been a challenge. The strategy of designing multifunctional synthetic carriers targeting several extracellular and intracellular barriers in the gene transfer pathway has emerged as a promising approach to improving the efficacy of gene delivery systems. This review will explain how sophisticated synthetic vectors can be created by combining conventional polycationic vectors such as polyethylenimine and basic amino acid peptides with additional polymers and peptides that are designed to overcome potential barriers to the gene delivery process.
Collapse
Affiliation(s)
- Hamideh Parhiz
- Pharmaceutical Research Center, Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, P.O. Box 91775-1365, Mashhad, Iran
| | | | | |
Collapse
|
61
|
Abstract
The convergent syntheses of homogeneous disulfide cross-linked polypeptides are reported. Reducible polypeptides were synthesized containing four and eight dodecapeptides in two and three linear conjugation steps. Critical for the convergent methodology was the use of orthogonally protected cysteines as either acetamidomethyl (Acm) or Fmoc-thiazolidine (Thz). Both groups could be selectively deprotected with silver trifluoromethanesulfonate in the presence of internal disulfide bonds using TFA and aqueous conditions, respectively. This approach allows for large, reducible polypeptides to be synthesized in efficient yields and minimizes the number of conjugation steps, allowing the development and optimization of gene delivery polypeptides containing multiple peptide components necessary to overcome the numerous in vivo barriers for efficacious gene delivery.
Collapse
Affiliation(s)
- Mark D Ericson
- Division of Medicinal & Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City IA 52242
| | - Kevin G Rice
- Division of Medicinal & Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City IA 52242
| |
Collapse
|
62
|
Ericson MD, Rice KG. Iterative Reducible Ligation to form Homogeneous Penicillamine Cross-linked Polypeptides. Tetrahedron Lett 2013; 54. [PMID: 24347694 DOI: 10.1016/j.tetlet.2013.04.086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The syntheses of homogeneous penicillamine disulfide cross-linked polypeptides are reported. Dodecapeptides containing N-terminal, C-terminal, or N- and C-terminal Pen were serially ligated into 36 amino acid polypeptides linked through Cys-Pen, Pen-Cys or Pen-Pen disulfide bonds. Critical to the syntheses was the incorporation of thiazolidine masked Cys and Pen as the N-terminal residues and selective hydrolysis with silver trifluoromethanesulfonate in acidic aqueous conditions to generate a free thiol for subsequent ligation. This approach allows the synthesis of homogeneous disulfide cross-linked polypeptides that have different reductive stabilities and have application in gene delivery by undergoing a tempered reductive triggered release of DNA.
Collapse
Affiliation(s)
- Mark D Ericson
- Division of Medicinal & Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City IA 52242
| | - Kevin G Rice
- Division of Medicinal & Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City IA 52242
| |
Collapse
|
63
|
Alhakamy NA, Nigatu AS, Berkland CJ, Ramsey JD. Noncovalently associated cell-penetrating peptides for gene delivery applications. Ther Deliv 2013; 4:741-57. [PMID: 23738670 PMCID: PMC4207642 DOI: 10.4155/tde.13.44] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The use of various cell-penetrating peptides (CPPs) to deliver genetic material for gene therapy applications has been a topic of interest for more than 20 years. The delivery of genetic material by using CPPs can be divided into two categories: covalently bound and electrostatically bound. Complexity of the synthesis procedure can be a significant barrier to translation when using a strategy requiring covalent binding of CPPs. In contrast, electrostatically complexing CPPs with genetic material or with a viral vector is relatively simple and has been demonstrated to improve gene delivery in both in vitro and in vivo studies. This review highlights gene therapy applications of complexes formed noncovalently between CPPs and genetic material or viruses.
Collapse
Affiliation(s)
- Nabil A Alhakamy
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA 66047
| | - Adane S Nigatu
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK, USA 74078
| | - Cory J Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA 66047
- Department of Chemical & Petroleum Engineering, University of Kansas, Lawrence, KS, USA 66047
| | - Joshua D Ramsey
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK, USA 74078
| |
Collapse
|
64
|
Bioreducible polypeptide containing cell-penetrating sequence for efficient gene delivery. Pharm Res 2013; 30:1968-78. [PMID: 23604924 DOI: 10.1007/s11095-013-1040-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 03/27/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE To design excellent polypeptide-based gene vectors and determine the gene delivery efficiency. METHODS Polypeptides (designated as xPolyK6, xPolyK6-R81 and xPolyK6-R82), comprising the DNA condensing and buffering peptide HK6H as well as cell penetrating peptide (CPP) R8 were obtained by the oxidative polymerization of CHK6HC and CR8C at different molar ratios in 4 mL phosphate-buffered saline (PBS) containing 30% (v/v) DMSO at room temperature for 96 h. The cytotoxicity of vectors was studied by MTT assay. Moreover, particle size, zeta potential and morphology along with the in vitro transfection efficiency and cellular uptake of vector/plasmid DNA (pDNA) complexes were characterized at various w/w ratios to determine their potential in gene therapy. RESULTS All the vectors presented excellent ability of binding and condensing pDNA, additionally with low cytotoxicity. Simultaneously, transfection efficiency of the vectors appeared apparent dependence on the vector composition. The distinct correlation between the content of CR8C with the transfection efficiency demonstrated the effective improvement in transfection efficacy by the oxidative polymerization. Particularly, xPolyK6-R82 possessed the highest transfection efficiency at a w/w ratio of 50. Furthermore, xPolyK6-R82 also presented the best cellular uptake capability demonstrated by confocal microscopy and flow cytometry. CONCLUSIONS Bioreducible polypeptides incorporating with proper amount of CPP are promising as effective non-viral gene vectors in gene therapy.
Collapse
|
65
|
Ericson MD, Rice KG. Synthesis of homogenous disulfide cross-linked polypeptides by iterative reducible ligation. Biopolymers 2013. [PMID: 23203756 DOI: 10.1002/bip.22117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A new method of directed solution phase synthesis of polypeptides linked through iterative formation of disulfide bonds is reported. Four dodecapeptides were successfully ligated into a single 48 amino acid polypeptide using an N-terminal Fmoc-thiazolidine and a novel acidic silver trifluoromethanesulfonate thiazolidine hydrolysis to achieve efficient ligation in the presence of internal disulfide bonds. The approach allows the synthesis of homogeneous disulfide cross-linked polypeptides that have application in gene delivery by undergoing a reductively triggered release of DNA.
Collapse
Affiliation(s)
- Mark D Ericson
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, IA 52242, USA
| | | |
Collapse
|
66
|
Alexander C, Fernandez Trillo F. Bioresponsive Polyplexes and Micelleplexes. SMART MATERIALS FOR DRUG DELIVERY 2013. [DOI: 10.1039/9781849736800-00256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The delivery of nucleic acids (NAs) is hindered by several factors, such as the size of the biomolecule (micron size for plasmid DNA), the presence of different biological barriers or the degradation of NAs. Most of these limitations are avoided by complexation with polycationic species, which collapse NAs into nanometer-sized polyplexes that can be efficiently internalized into the target cells. Because there are subtle changes in physiological conditions, such as the drop in pH at the endosome, or the increase in temperature in tumor tissue, stimuli responsive synthetic polymers are ideal candidates for the synthesis of efficient gene delivery vehicles. In this chapter, representative examples of “smart” polypexes that exploit these changes in physiological environment for the delivery of NAs are described, and the transfection efficiency of pH-, redox-, temperature- and light-responsive polyplexes is analyzed.
Collapse
|
67
|
|
68
|
Shao Y, Huang W, Shi C, Atkinson ST, Luo J. Reversibly crosslinked nanocarriers for on-demand drug delivery in cancer treatment. Ther Deliv 2012; 3:1409-27. [PMID: 23323559 PMCID: PMC3575096 DOI: 10.4155/tde.12.106] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Polymer micelles have proven to be one of the most versatile nanocarriers for anticancer drug delivery. However, the in vitro and in vivo stability of micelles remains a challenge due to the dynamic nature of these self-assembled systems, which leads to premature drug release and nonspecific biodistribution in vivo. Recently, reversibly crosslinked micelles have been developed to provide solutions to stabilize nanocarriers in blood circulation. Increased stability allows nanoparticles to accumulate at tumor sites efficiently via passive and/or active tumor targeting, while cleavage of the micelle crosslinkages, through internal or external stimuli, facilitates on-demand drug release. In this review, various crosslinking chemistries as well as the choices for reversible linkages in these nanocarriers will be introduced. Then, the development of reversibly crosslinked micelles for on-demand drug release in response to single or dual stimuli in the tumor microenvironment is discussed, for example, acidic pH, reducing microenvironment, enzymatic microenvironment, photoirradiation and the administration of competitive reagents postmicelle delivery.
Collapse
Affiliation(s)
- Yu Shao
- Department of Pharmacology, SUNY Upstate Cancer Research Institute, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Wenzhe Huang
- Department of Pharmacology, SUNY Upstate Cancer Research Institute, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Changying Shi
- Department of Pharmacology, SUNY Upstate Cancer Research Institute, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Sean T Atkinson
- Department of Pharmacology, SUNY Upstate Cancer Research Institute, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Juntao Luo
- Department of Pharmacology, SUNY Upstate Cancer Research Institute, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
69
|
Shim MS, Kwon YJ. Stimuli-responsive polymers and nanomaterials for gene delivery and imaging applications. Adv Drug Deliv Rev 2012; 64:1046-59. [PMID: 22329941 DOI: 10.1016/j.addr.2012.01.018] [Citation(s) in RCA: 283] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 01/30/2012] [Accepted: 01/30/2012] [Indexed: 12/11/2022]
Abstract
Multiple extra- and intracellular obstacles, including low stability in blood, poor cellular uptake, and inefficient endosomal escape and disassembly in the cytoplasm, have to be overcome in order to deliver nucleic acids for gene therapy. This review introduces the recent advances in tackling the key challenges in achieving efficient, targeted, and safe nonviral gene delivery using various nucleic acid-containing nanomaterials that are designed to respond to various extra- and intracellular biological stimuli (e.g., pH, redox potential, and enzyme) as well as external artificial triggers (e.g., light and ultrasound). Gene delivery in combination with molecular imaging and targeting enables diagnostic assessment, treatment monitoring and quantification of efficiency, and confirmation of cure, thus fulfilling the great promise of efficient and personalized medicine. Nanomaterials platform for combined imaging and gene therapy, nanotheragnostics, using stimuli-responsive materials is also highlighted in this review. It is clear that developing novel multifunctional nonviral vectors, which transform their physico-chemical properties in response to various stimuli in a timely and spatially controlled manner, is highly desired to translate the promise of gene therapy for the clinical success.
Collapse
|
70
|
Kim BK, Kang H, Doh KO, Lee SH, Park JW, Lee SJ, Lee TJ. Homodimeric SV40 NLS peptide formed by disulfide bond as enhancer for gene delivery. Bioorg Med Chem Lett 2012; 22:5415-8. [PMID: 22871581 DOI: 10.1016/j.bmcl.2012.07.051] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 07/10/2012] [Accepted: 07/12/2012] [Indexed: 10/28/2022]
Abstract
Recently, cysteine residue incorporation increased liposome-mediated transfection compared to unmodified peptide. Therefore, we designed novel modified SV40 NLS peptides, homodimeric (NLS-CTHD, NLS-NTHD) and closed structure (cyclic NLS), simply using disulfide bond between cysteines to develop more efficient and safe non-viral gene delivery system. The simple mix of NLS-CTHD among these novel transfection enhancing peptides with DNA increased the gene transfer potency of cationic liposomes more efficiently with no additional cytotoxicity.
Collapse
Affiliation(s)
- Bieong-Kil Kim
- Department of Physiology, College of Medicine, Yeungnam University, Daegu 705-717, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
71
|
Son S, Namgung R, Kim J, Singha K, Kim WJ. Bioreducible polymers for gene silencing and delivery. Acc Chem Res 2012; 45:1100-12. [PMID: 22129162 DOI: 10.1021/ar200248u] [Citation(s) in RCA: 211] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Polymeric gene delivery vectors show great potential for the construction of the ideal gene delivery system. These systems harness their ability to incorporate versatile functional traits to overcome most impediments encountered in gene delivery: from the initial complexation to their target-specific release of the therapeutic nucleic acids at the cytosol. Among the numerous multifunctional polymers that have been designed and evaluated as gene delivery vectors, polymers with redox-sensitive (or bioreducible) functional domains have gained great attention in terms of their structural and functional traits. The redox environment plays a pivotal role in sustaining cellular homeostasis and natural redox potential gradients exist between extra- and intracellular space and between the exterior and interior of subcellular organelles. In some cases, researchers have designed the polymeric delivery vectors to exploit these gradients. For example, researchers have taken advantage of the high redox potential gradient between oxidizing extracellular space and the reducing environment of cytosolic compartments by integrating disulfide bonds into the polymer structure. Such polymers retain their cargo in the extracellular space but selectively release the therapeutic nucleic acids in the reducing space within the cytosol. Furthermore, bioreducible polymers form stable complex with nucleic acids, and researchers can fabricate these structures to impart several important features such as site-, timing-, and duration period-specific gene expression. Additionally, the introduction of disulfide bonds within these polymers promotes their biodegradability and limits their cytotoxicity. Many approaches have demonstrated the versatility of bioreducible gene delivery, but the underlying biological rationale of these systems remains poorly understood. The process of disulfide reduction depends on multiple variables in the cellular redox environment. Therefore, the quest to unravel various issues such as the site and time of disulfide bond reduction during the cellular uptake and trafficking have stimulated a number of interesting studies which have employed disulfide compounds with a variety of reducible linkers. Such studies help researchers understand not only how modifications made to disulfides can alter their thiol-disulfide exchange characteristics but also to decipher the effect of the induced changes on the dynamics of the redox environment. This Account discusses current research trends and recent progress in the disulfide chemistry enabling novel and versatile designs of reducible polymeric gene delivery systems. We present strategies for the introduction of disulfide bonds into polymers. These representative examples and their respective outcomes elaborate the benefit and efficiency of disulfides at the individual stages of gene delivery.
Collapse
Affiliation(s)
- Sejin Son
- Department of Chemistry, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Ran Namgung
- Department of Chemistry, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Jihoon Kim
- Department of Chemistry, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Kaushik Singha
- Department of Chemistry, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Won Jong Kim
- Department of Chemistry, Pohang University of Science and Technology, Pohang 790-784, Korea
| |
Collapse
|
72
|
Zhang Q, Noh SM, Nam JH, Jung HW, Park JM, Oh JK. Dual Temperature and Thiol-Responsive POEOMA-Multisegmented Polydisulfides: Synthesis and Thermoresponsive Properties. Macromol Rapid Commun 2012; 33:1528-34. [DOI: 10.1002/marc.201200289] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 05/10/2012] [Indexed: 12/28/2022]
|
73
|
Rosada RS, Silva CL, Santana MHA, Nakaie CR, de la Torre LG. Effectiveness, against tuberculosis, of pseudo-ternary complexes: Peptide-DNA-cationic liposome. J Colloid Interface Sci 2012; 373:102-9. [DOI: 10.1016/j.jcis.2011.09.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 09/16/2011] [Accepted: 09/19/2011] [Indexed: 12/31/2022]
|
74
|
Åmand HL, Nordén B, Fant K. Functionalization with C-terminal cysteine enhances transfection efficiency of cell-penetrating peptides through dimer formation. Biochem Biophys Res Commun 2012; 418:469-74. [DOI: 10.1016/j.bbrc.2012.01.041] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 01/08/2012] [Indexed: 10/14/2022]
|
75
|
Beloor J, Choi CS, Nam HY, Park M, Kim SH, Jackson A, Lee KY, Kim SW, Kumar P, Lee SK. Arginine-engrafted biodegradable polymer for the systemic delivery of therapeutic siRNA. Biomaterials 2012; 33:1640-50. [DOI: 10.1016/j.biomaterials.2011.11.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 11/07/2011] [Indexed: 01/31/2023]
|
76
|
Liu J, Zhou J, Luo Y. SiRNA Delivery Systems Based on Neutral Cross-Linked Dendrimers. Bioconjug Chem 2012; 23:174-83. [DOI: 10.1021/bc200433s] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Jie Liu
- Department of Biomedical Engineering, College of Engineering, Peking University, Room 206, Fangzheng Building, 298 Chengfu Road, Haidian District,
Beijing, China 100871
| | - Jihan Zhou
- Beijing
National Laboratory
for Molecular Sciences and the Key Laboratory of Polymer Chemistry
and Physics of Ministry of Education, College of Chemistry
and Molecular Engineering, Peking University, Beijing, China, 100871
| | - Ying Luo
- Department of Biomedical Engineering, College of Engineering, Peking University, Room 206, Fangzheng Building, 298 Chengfu Road, Haidian District,
Beijing, China 100871
| |
Collapse
|
77
|
Fernandes MM, Cavaco-Paulo A. Protein disulphide isomerase-assisted functionalization of proteinaceous substrates. BIOCATAL BIOTRANSFOR 2012. [DOI: 10.3109/10242422.2012.646657] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
78
|
González-Aramundiz JV, Lozano MV, Sousa-Herves A, Fernandez-Megia E, Csaba N. Polypeptides and polyaminoacids in drug delivery. Expert Opin Drug Deliv 2012; 9:183-201. [DOI: 10.1517/17425247.2012.647906] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
79
|
Miyata K, Nishiyama N, Kataoka K. Rational design of smart supramolecular assemblies for gene delivery: chemical challenges in the creation of artificial viruses. Chem Soc Rev 2012; 41:2562-74. [DOI: 10.1039/c1cs15258k] [Citation(s) in RCA: 378] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
80
|
Nanoparticles for Gene Delivery into Stem Cells and Embryos. MULTIFACETED DEVELOPMENT AND APPLICATION OF BIOPOLYMERS FOR BIOLOGY, BIOMEDICINE AND NANOTECHNOLOGY 2012. [DOI: 10.1007/12_2012_194] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
81
|
Yang J, Lei Q, Han K, Gong YH, Chen S, Cheng H, Cheng SX, Zhuo RX, Zhang XZ. Reduction-sensitive polypeptides incorporated with nuclear localization signal sequences for enhanced gene delivery. ACTA ACUST UNITED AC 2012. [DOI: 10.1039/c2jm32223d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
82
|
Soliman M, Nasanit R, Abulateefeh SR, Allen S, Davies MC, Briggs SS, Seymour LW, Preece JA, Grabowska AM, Watson SA, Alexander C. Multicomponent Synthetic Polymers with Viral-Mimetic Chemistry for Nucleic Acid Delivery. Mol Pharm 2011; 9:1-13. [DOI: 10.1021/mp200108q] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Mahmoud Soliman
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
- Department
of Pharmaceutics,
Faculty of Pharmacy, Ain Shams University, Monazamet El Wehda El Afrikia St., El Abbassia, Cairo, Egypt
| | - Rujikan Nasanit
- School of Chemistry, University of Birmingham, Birmingham B15 2TT, U.K
- Department of Biotechnology, Faculty
of Engineering and Industrial Technology, Silpakorn University, Nakorn Pathom 73000, Thailand
| | | | - Stephanie Allen
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Martyn C. Davies
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Simon S. Briggs
- Department of Clinical Pharmacology, University of Oxford, Oxford OX2 6HE, U.K
| | - Leonard W. Seymour
- Department of Clinical Pharmacology, University of Oxford, Oxford OX2 6HE, U.K
| | - Jon A. Preece
- School of Chemistry, University of Birmingham, Birmingham B15 2TT, U.K
| | - Anna M. Grabowska
- Division of Pre-Clinical Oncology, University of Nottingham, Queens Medical Centre, Nottingham
NG7 2UH, U.K
| | - Susan A. Watson
- Division of Pre-Clinical Oncology, University of Nottingham, Queens Medical Centre, Nottingham
NG7 2UH, U.K
| | - Cameron Alexander
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
| |
Collapse
|
83
|
Bolhassani A. Potential efficacy of cell-penetrating peptides for nucleic acid and drug delivery in cancer. Biochim Biophys Acta Rev Cancer 2011; 1816:232-46. [DOI: 10.1016/j.bbcan.2011.07.006] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 07/29/2011] [Accepted: 07/30/2011] [Indexed: 10/17/2022]
|
84
|
Zhao Y, Zhang S, Cui S, Wang B, Zhang S. Peptide-based cationic liposome-mediated gene delivery. Expert Opin Drug Deliv 2011; 9:127-39. [DOI: 10.1517/17425247.2011.630387] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
85
|
de Raad M, Teunissen EA, Lelieveld D, Egan DA, Mastrobattista E. High-content screening of peptide-based non-viral gene delivery systems. J Control Release 2011; 158:433-42. [PMID: 21983020 DOI: 10.1016/j.jconrel.2011.09.078] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 09/15/2011] [Accepted: 09/18/2011] [Indexed: 01/13/2023]
Abstract
High-content screening (HCS) uses high-capacity automated fluorescence imaging for the quantitative analysis of single cells and cell populations. Here, we developed an HCS assay for rapid screening of non-viral gene delivery systems as exemplified by the screening of a small library of peptide-based transfectants. These peptides were simultaneously screened for transfection efficiency, cytotoxicity, induction of cell permeability and the capacity to transfect non-dividing cells. We demonstrated that HCS is a valuable extension to the already existing screening methods for the in vitro evaluation of non-viral gene delivery systems with the added value that multiple parameters can be screened in parallel thereby obtaining more information from a single screening event, which will accelerate the development of novel gene delivery systems.
Collapse
Affiliation(s)
- Markus de Raad
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of science, University of Utrecht, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
86
|
Shi J, Johnson RN, Schellinger JG, Carlson PM, Pun SH. Reducible HPMA-co-oligolysine copolymers for nucleic acid delivery. Int J Pharm 2011; 427:113-22. [PMID: 21893178 DOI: 10.1016/j.ijpharm.2011.08.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 07/18/2011] [Accepted: 08/08/2011] [Indexed: 11/20/2022]
Abstract
Biodegradability can be incorporated into cationic polymers via use of disulfide linkages that are degraded in the reducing environment of the cell cytosol. In this work, N-(2-hydroxypropyl)methacrylamide (HPMA) and methacrylamido-functionalized oligo-l-lysine peptide monomers with either a non-reducible 6-aminohexanoic acid (AHX) linker or a reducible 3-[(2-aminoethyl)dithiol] propionic acid (AEDP) linker were copolymerized via reversible addition-fragmentation chain transfer (RAFT) polymerization. Both of the copolymers and a 1:1 (w/w) mixture of copolymers with reducible and non-reducible peptides were complexed with DNA to form polyplexes. The polyplexes were tested for salt stability, transfection efficiency, and cytotoxicity. The HPMA-oligolysine copolymer containing the reducible AEDP linkers was less efficient at transfection than the non-reducible polymer and was prone to flocculation in saline and serum-containing conditions, but was also not cytotoxic at charge ratios tested. Optimal transfection efficiency and toxicity were attained with mixed formulation of copolymers. Flow cytometry uptake studies indicated that blocking extracellular thiols did not restore transfection efficiency and that the decreased transfection of the reducible polyplex is therefore not primarily caused by extracellular polymer reduction by free thiols. The decrease in transfection efficiency of the reducible polymers could be partially mitigated by the addition of low concentrations of EDTA to prevent metal-catalyzed oxidation of reduced polymers.
Collapse
Affiliation(s)
- Julie Shi
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | | | | | | | | |
Collapse
|
87
|
CHEN J, WANG H, XU X, CHEN W, ZHANG X. PEPTIDES AND POLYPEPTIDES FOR GENE AND DRUG DELIVERY. ACTA POLYM SIN 2011. [DOI: 10.3724/sp.j.1105.2011.11100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
88
|
Kudsiova L, Fridrich B, Ho J, Mustapa MFM, Campbell F, Welser K, Keppler M, Ng T, Barlow DJ, Tabor AB, Hailes HC, Lawrence MJ. Lipopolyplex Ternary Delivery Systems Incorporating C14 Glycerol-Based Lipids. Mol Pharm 2011; 8:1831-47. [DOI: 10.1021/mp2001796] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Laila Kudsiova
- Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, Waterloo Campus, London SE1 9NH, U.K
| | - Barbara Fridrich
- Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, Waterloo Campus, London SE1 9NH, U.K
| | - Jimmy Ho
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K
| | - M. Firouz Mohd Mustapa
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K
| | - Frederick Campbell
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K
| | - Katharina Welser
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K
| | - Melanie Keppler
- Randall Division of Cell and Molecular Biophysics, King’s College London, Henriette Raphael Building, Guy's Campus, London SE1 1UL, U.K
| | - Tony Ng
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K
- Randall Division of Cell and Molecular Biophysics, King’s College London, Henriette Raphael Building, Guy's Campus, London SE1 1UL, U.K
| | - David J. Barlow
- Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, Waterloo Campus, London SE1 9NH, U.K
| | - Alethea B. Tabor
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K
| | - Helen C. Hailes
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K
| | - M. Jayne Lawrence
- Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, Waterloo Campus, London SE1 9NH, U.K
| |
Collapse
|
89
|
Troiber C, Wagner E. Nucleic Acid Carriers Based on Precise Polymer Conjugates. Bioconjug Chem 2011; 22:1737-52. [DOI: 10.1021/bc200251r] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Christina Troiber
- Pharmaceutical Biotechnology, Center for System-based Drug Research and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-based Drug Research and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Butenandtstrasse 5-13, 81377 Munich, Germany
| |
Collapse
|
90
|
Olton DYE, Close JM, Sfeir CS, Kumta PN. Intracellular trafficking pathways involved in the gene transfer of nano-structured calcium phosphate-DNA particles. Biomaterials 2011; 32:7662-70. [PMID: 21774979 DOI: 10.1016/j.biomaterials.2011.01.043] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Accepted: 01/14/2011] [Indexed: 12/29/2022]
Abstract
Nano-structured calcium phosphate (NanoCaP) particles have been proven to be a powerful means of non-viral gene delivery. In order to better understand the mechanisms through which NanoCaPs-mediated mammalian cell transfection is achieved, we have sought to define the intracellular trafficking pathways involved in the cellular uptake and intracellular processing of these particles. Previous work has indicated that NanoCaP-DNA complexes are most likely internalized via endocytosis, however the subsequent pathways involved have not been determined. Through the use of specific inhibitors, we show that endocytosis of NanoCaP particles is both clathrin- and caveolae-dependent, and suggest that the caveolaer mechanism is the major contributor. We demonstrate colocalization of NanoCaP-pDNA complexes with known markers of both clathrin-coated and caveolar vesicles. Furthermore, through the use of quantitative flow cytometry, we present the first work in which the percent internalization of CaP-DNA complexes into cells is quantified. The overall goal of this research is to foster the continued improvement of NanoCaP-based gene delivery strategies.
Collapse
Affiliation(s)
- Dana Y E Olton
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
91
|
Protein disulphide isomerase-assisted functionalization of keratin-based matrices. Appl Microbiol Biotechnol 2011; 90:1311-21. [DOI: 10.1007/s00253-011-3194-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 02/14/2011] [Accepted: 02/20/2011] [Indexed: 10/18/2022]
|
92
|
Abstract
Bioreducible polymers, which possess mainly disulfide linkages in the polymer structures, have appeared as ideal gene delivery carriers due to the high stability in extracellular physiological condition and bioreduction-triggered release of genetic materials, as well as decreased cytotoxicity because intracellular cytosol is a reducing environment containing high level of reducing molecules such as glutathione. This review will describe the initiation and recent advances in the development of bioreducible polymers for gene delivery, which includes reducibly cross-linked PEIs, polypeptides, polyion complex micelles, and poly(amido amine)s. There have been extensive researches performed to exhibit great gene delivery efficacy but still several important issues about pharmacokinetics or safety should be answered thoroughly for further rational design of bioreducible polymers having potentials in human gene delivery systems.
Collapse
Affiliation(s)
- Tae-il Kim
- Department of Biosystems and Biomaterials Science and Engineering, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sung Wan Kim
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
93
|
Lamarre B, Ryadnov MG. Self-assembling viral mimetics: one long journey with short steps. Macromol Biosci 2010; 11:503-13. [PMID: 21165940 DOI: 10.1002/mabi.201000330] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Indexed: 12/14/2022]
Abstract
Recently, the Foresight Institute has pronounced six economic challenges that can be addressed through the progress of nanotechnology. One of these is the health and longevity of human life. Amongst applications anticipated to provide a solution to this challenge, gene therapy appears to be particularly promising. In theory, many diseases that result from genetic disorders can be cured by correcting defective genes. In practice, finding efficient and safe delivery vectors remains the stumbling point on the path of genetic therapies to the clinic. Viruses, otherwise the most efficient transfectors, pose safety concerns over immune reactions, whereas synthetic gene packages greatly lack the structural integrity of viruses. An ideal vector is therefore seen as a compromise between the two: a nanoscale device, which would mimic a virus and act as a virus, but would do this at the designer's whim. A strategy to achieve this is offered by the virus architecture itself, the principles of which are translated into the function via exquisitely reproducible self-assembly mechanisms. Thus, to mimic a virus is to mimic the way it is built, i.e., self-assembly. With just a few attempts made so far, the journey to an artificial virus has had a short lifetime, but the promise it holds is not expected to reduce any time soon.
Collapse
Affiliation(s)
- Baptiste Lamarre
- National Physical Laboratory, Teddington, Middlesex, TW110LW, UK
| | | |
Collapse
|
94
|
Drake CR, Aissaoui A, Argyros O, Serginson JM, Monnery BD, Thanou M, Steinke JHG, Miller AD. Bioresponsive small molecule polyamines as noncytotoxic alternative to polyethylenimine. Mol Pharm 2010; 7:2040-55. [PMID: 20929266 DOI: 10.1021/mp9002249] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Nonviral gene therapy continues to require novel synthetic vectors to deliver therapeutic nucleic acids effectively and safely. The majority of synthetic nonviral vectors employed in clinical trials to date have been cationic liposomes; however, cationic polymers are attracting increasing attention. One of the few cationic polymers to enter clinical trials has been polyethylenimine (PEI); however, doubts remain over its cytotoxicity, and in addition it displays lower levels of transfection than viral systems. Herein, we report on the development of a series of small molecule analogues of PEI that are bioresponsive to the presence of pDNA, forming poly(disulfide)s that are capable of efficacious transfection with no associated toxicity. The most effective small molecule developed, a cyclic disulfide based upon a spermine backbone, is shown to form very well-defined polyplexes (100-200 nm in diameter) that mediate murine lung transfection in vivo to within an order of magnitude of in vivo jetPEI, and at the same time display a much improved cytotoxicity profile.
Collapse
Affiliation(s)
- Christopher R Drake
- Department of Chemistry, Imperial College London, Imperial College Genetic Therapies Centre, Flowers Building, Armstrong Road, London SW7 2AZ, UK
| | | | | | | | | | | | | | | |
Collapse
|
95
|
Fernandez CA, Baumhover NJ, Anderson K, Rice KG. Discovery of metabolically stabilized electronegative polyacridine-PEG peptide DNA open polyplexes. Bioconjug Chem 2010; 21:723-30. [PMID: 20218669 DOI: 10.1021/bc900514s] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cationic condensing peptides and polymers bind electrostatically to DNA to form cationic polyplexes. While many cationic polyplexes are able to achieve in vitro transfection mediated through electrostatic interactions, few have been able to mediate gene transfer in vivo. The present study describes the development and testing of polyacridine PEG-peptides that bind to plasmid DNA by intercalation resulting in electronegative open polyplex DNA. Polyacridine PEG-peptides were prepared by chemically conjugating 6-(9-acridinylamino) hexanoic acid onto side chains of Lys in PEG-Cys-Trp-(Lys)(3, 4, or 5). The resulting PEG-Cys-Trp-(Lys-(Acr))(3, 4, or 5) peptides bound tightly to DNA by polyintercalation, rather than electrostatic binding. Unlike polycationic polyplexes, polyacridine PEG-peptide polyplexes were anionic and open coiled, as revealed by zeta potential and atomic force microscopy. PEG-Cys-Trp-(Lys-(Acr))(5) showed the highest DNA binding affinity and the greatest ability to protect DNA from metabolism by DNase. Polyacridine PEG-peptide DNA open polyplexes were dosed intramuscularly and electroporated in mice to demonstrate their functional activity in gene transfer. These results establish polyacridine PEG-peptide DNA open polyplexes as a novel gene delivery method for in vivo use.
Collapse
Affiliation(s)
- Christian A Fernandez
- Divisions of Pharmaceutics and Medicinal & Natural Products Chemistry, College of Pharmacy, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | |
Collapse
|
96
|
Baumhover NJ, Anderson K, Fernandez CA, Rice KG. Synthesis and in vitro testing of new potent polyacridine-melittin gene delivery peptides. Bioconjug Chem 2010; 21:74-83. [PMID: 19968269 DOI: 10.1021/bc9003124] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The combination of a polyacridine peptide modified with a melittin fusogenic peptide results in a potent gene transfer agent. Polyacridine peptides of the general formula (Acr-X)(n)-Cys were prepared by solid-phase peptide synthesis, where Acr is Lys modified on its epsilon-amine with acridine, X is Arg, Leu, or Lys and n is 2, 3, or 4 repeats. The Cys residue was modified by either a maleimide-melittin or a thiolpyridine-Cys-melittin fusogenic peptide resulting in reducible or non-reducible polyacridine-melittin peptides. Hemolysis assays established that polyacridine-melittin peptides retained their membrane lytic potency relative to melittin at pH 7.4 and 5. When combined with plasmid DNA, the membrane lytic potency of polyacridine-melittin peptides was neutralized. Gene transfer experiments in multiple cell lines established that polyacridine-melittin peptides mediate expression as efficiently as PEI. The expression was very dependent upon a disulfide bond linking polyacridine to melittin. The gene transfer was most efficient when X is Arg and n is 3 or 4 repeats. These studies establish polyacridine peptides as a novel DNA binding anchor peptide.
Collapse
|
97
|
Ulrich-Vinther M. Gene therapy methods in bone and joint disorders. ACTA ORTHOPAEDICA. SUPPLEMENTUM 2010. [DOI: 10.1080/17453690610046512] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
98
|
Sun W, Davis PB. Reducible DNA nanoparticles enhance in vitro gene transfer via an extracellular mechanism. J Control Release 2010; 146:118-27. [PMID: 20438780 DOI: 10.1016/j.jconrel.2010.04.031] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 04/23/2010] [Accepted: 04/27/2010] [Indexed: 11/18/2022]
Abstract
We developed polylysine based DNA nanoparticles (DNA NPs) that contain disulfide linkage in the carrier and demonstrated that this reducible DNA NP enhances in vitro gene transfer via an extracellular mechanism. Polylysine was conjugated through an N-terminal cysteine to a polyethylene glycol chain (PEG) by either a disulfide bond (SS) or a thioether bond (CS), and the resulting PEG-peptide conjugates were used to compact plasmid DNA into reducible SS-DNA NPs or non-reducible CS-DNA NPs with identical physical properties. SS-DNA NPs mediated more than 10-fold higher in vitro gene transfer. Others have suggested that disulfide bonds in synthetic gene carriers undergo cleavage in the reducing environment inside the cell, allowing increased intracellular DNA release. In this study, however, both higher cellular uptake of SS-DNA NPs and inhibition of SS-DNA NP mediated in vitro gene transfer by blocking extracellular free thiols suggested an extracellular mechanism. DePEGylation of SS-DNA NPs by extracellular thiols caused aggregation which might lead to higher cellular uptake and higher transgene expression. A series of SS-DNA NPs prepared with stabilized disulfide bonds survived the extracellular environment without aggregation but lost the superior gene transfer ability, indicating that, in our system, intracellular mechanisms are not involved. These results provided further insight into the mechanisms of in vitro gene transfer enhancement by introducing reducible linkages, contributing to the rational design of more efficient non-viral gene delivery systems.
Collapse
Affiliation(s)
- Wenchao Sun
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | | |
Collapse
|
99
|
Blacklock J, Sievers TK, Handa H, You YZ, Oupický D, Mao G, Möhwald H. Cross-linked bioreducible layer-by-layer films for increased cell adhesion and transgene expression. J Phys Chem B 2010; 114:5283-91. [PMID: 20369813 PMCID: PMC2861295 DOI: 10.1021/jp100486h] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The effect of cross-linking layer-by-layer (LbL) films consisting of bioreducible poly(2-dimethylaminoethyl methacrylate) (rPDMAEMA) and DNA is examined with regard to rigidity, biodegradability, cell adhesion, and transfection activity using 1,5-diiodopentane (DIP) cross-linker. DIP chemically reacts with the tertiary amines of rPDMAEMA, altering the chemical composition of these LbL films. The result is a change in surface morphology, film swelling behavior, and film rigidity, measured with AFM and ellipsometry. It is found that the apparent Young's modulus is increased more than 4 times its original value upon cross-linking. Cross-linking mass is additionally confirmed with a quartz crystal microbalance with dissipation (QCM-D). Comprehensive analyses of these experimental values were investigated to calculate the degree of cross-linking using the rubber elasticity theory and the Flory-Rehner theory. Additionally, the Flory-Huggins parameter, chi, was calculated. Good agreement in the two methods yields a cross-linking density of approximately 0.82 mmol/cm(3). The Flory-Huggins parameter increased upon cross-linking from 1.07 to 1.2, indicating increased hydrophobicity of the network and formation of bulk water droplets within the films. In addition, the effects of cross-linking on film disassembly by 1,4-dithiothreitol (DTT) are found to be insignificant despite the alteration in film rigidity. Mouse fibroblast cells and smooth muscle cells are used to study the effect of cross-linking on cell adhesion and cell transfection activity. In vitro transfection activity up to seven days is quantified using secreted alkaline phosphatase (SEAP) DNA. Film cross-linking is found to enhance cell adhesion and prolong the duration of cellular transfection. These results contribute to the development of bioreducible polymer coatings for localized gene delivery.
Collapse
Affiliation(s)
- Jenifer Blacklock
- Department of Biomedical Engineering, Wayne State University, Detroit, Michigan 48202, USA.
| | | | | | | | | | | | | |
Collapse
|
100
|
Xue YN, Liu M, Peng L, Huang SW, Zhuo RX. Improving Gene Delivery Efficiency of Bioreducible Poly(amidoamine)s via Grafting with Dendritic Poly(amidoamine)s. Macromol Biosci 2010; 10:404-14. [DOI: 10.1002/mabi.200900300] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|