1
|
Hu M, Li X, You Z, Cai R, Chen C. Physiological Barriers and Strategies of Lipid-Based Nanoparticles for Nucleic Acid Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2303266. [PMID: 37792475 DOI: 10.1002/adma.202303266] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/21/2023] [Indexed: 10/06/2023]
Abstract
Lipid-based nanoparticles (LBNPs) are currently the most promising vehicles for nucleic acid drug (NAD) delivery. Although their clinical applications have achieved success, the NAD delivery efficiency and safety are still unsatisfactory, which are, to a large extent, due to the existence of multi-level physiological barriers in vivo. It is important to elucidate the interactions between these barriers and LBNPs, which will guide more rational design of efficient NAD vehicles with low adverse effects and facilitate broader applications of nucleic acid therapeutics. This review describes the obstacles and challenges of biological barriers to NAD delivery at systemic, organ, sub-organ, cellular, and subcellular levels. The strategies to overcome these barriers are comprehensively reviewed, mainly including physically/chemically engineering LBNPs and directly modifying physiological barriers by auxiliary treatments. Then the potentials and challenges for successful translation of these preclinical studies into the clinic are discussed. In the end, a forward look at the strategies on manipulating protein corona (PC) is addressed, which may pull off the trick of overcoming those physiological barriers and significantly improve the efficacy and safety of LBNP-based NADs delivery.
Collapse
Affiliation(s)
- Mingdi Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
- Sino-Danish Center for Education and Research, Beijing, 100049, China
| | - Xiaoyan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Zhen You
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Rong Cai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
- Sino-Danish Center for Education and Research, Beijing, 100049, China
- The GBA National Institute for Nanotechnology Innovation, Guangzhou, 510700, China
| |
Collapse
|
2
|
Szwed-Georgiou A, Płociński P, Kupikowska-Stobba B, Urbaniak MM, Rusek-Wala P, Szustakiewicz K, Piszko P, Krupa A, Biernat M, Gazińska M, Kasprzak M, Nawrotek K, Mira NP, Rudnicka K. Bioactive Materials for Bone Regeneration: Biomolecules and Delivery Systems. ACS Biomater Sci Eng 2023; 9:5222-5254. [PMID: 37585562 PMCID: PMC10498424 DOI: 10.1021/acsbiomaterials.3c00609] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/31/2023] [Indexed: 08/18/2023]
Abstract
Novel tissue regeneration strategies are constantly being developed worldwide. Research on bone regeneration is noteworthy, as many promising new approaches have been documented with novel strategies currently under investigation. Innovative biomaterials that allow the coordinated and well-controlled repair of bone fractures and bone loss are being designed to reduce the need for autologous or allogeneic bone grafts eventually. The current engineering technologies permit the construction of synthetic, complex, biomimetic biomaterials with properties nearly as good as those of natural bone with good biocompatibility. To ensure that all these requirements meet, bioactive molecules are coupled to structural scaffolding constituents to form a final product with the desired physical, chemical, and biological properties. Bioactive molecules that have been used to promote bone regeneration include protein growth factors, peptides, amino acids, hormones, lipids, and flavonoids. Various strategies have been adapted to investigate the coupling of bioactive molecules with scaffolding materials to sustain activity and allow controlled release. The current manuscript is a thorough survey of the strategies that have been exploited for the delivery of biomolecules for bone regeneration purposes, from choosing the bioactive molecule to selecting the optimal strategy to synthesize the scaffold and assessing the advantages and disadvantages of various delivery strategies.
Collapse
Affiliation(s)
- Aleksandra Szwed-Georgiou
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| | - Przemysław Płociński
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| | - Barbara Kupikowska-Stobba
- Biomaterials
Research Group, Lukasiewicz Research Network
- Institute of Ceramics and Building Materials, Krakow 31-983, Poland
| | - Mateusz M. Urbaniak
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
- The
Bio-Med-Chem Doctoral School, University of Lodz and Lodz Institutes
of the Polish Academy of Sciences, University
of Lodz, Lodz 90-237, Poland
| | - Paulina Rusek-Wala
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
- The
Bio-Med-Chem Doctoral School, University of Lodz and Lodz Institutes
of the Polish Academy of Sciences, University
of Lodz, Lodz 90-237, Poland
| | - Konrad Szustakiewicz
- Department
of Polymer Engineering and Technology, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw 50-370, Poland
| | - Paweł Piszko
- Department
of Polymer Engineering and Technology, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw 50-370, Poland
| | - Agnieszka Krupa
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| | - Monika Biernat
- Biomaterials
Research Group, Lukasiewicz Research Network
- Institute of Ceramics and Building Materials, Krakow 31-983, Poland
| | - Małgorzata Gazińska
- Department
of Polymer Engineering and Technology, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw 50-370, Poland
| | - Mirosław Kasprzak
- Biomaterials
Research Group, Lukasiewicz Research Network
- Institute of Ceramics and Building Materials, Krakow 31-983, Poland
| | - Katarzyna Nawrotek
- Faculty
of Process and Environmental Engineering, Lodz University of Technology, Lodz 90-924, Poland
| | - Nuno Pereira Mira
- iBB-Institute
for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de
Lisboa, Lisboa 1049-001, Portugal
- Associate
Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior
Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal
- Instituto
Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal
| | - Karolina Rudnicka
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| |
Collapse
|
3
|
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: 162] [Impact Index Per Article: 54.0] [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
|
4
|
Peng F, Zhang W, Qiu F. Self-assembling Peptides in Current Nanomedicine: Versatile Nanomaterials for Drug Delivery. Curr Med Chem 2020; 27:4855-4881. [PMID: 31309877 DOI: 10.2174/0929867326666190712154021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 05/27/2019] [Accepted: 06/11/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND The development of modern nanomedicine greatly depends on the involvement of novel materials as drug delivery system. In order to maximize the therapeutic effects of drugs and minimize their side effects, a number of natural or synthetic materials have been widely investigated for drug delivery. Among these materials, biomimetic self-assembling peptides (SAPs) have received more attention in recent years. Considering the rapidly growing number of SAPs designed for drug delivery, a summary of how SAPs-based drug delivery systems were designed, would be beneficial. METHOD We outlined research works on different SAPs that have been investigated as carriers for different drugs, focusing on the design of SAPs nanomaterials and how they were used for drug delivery in different strategies. RESULTS Based on the principle rules of chemical complementarity and structural compatibility, SAPs such as ionic self-complementary peptide, peptide amphiphile and surfactant-like peptide could be designed. Determined by the features of peptide materials and the drugs to be delivered, different strategies such as hydrogel embedding, hydrophobic interaction, electrostatic interaction, covalent conjugation or the combination of them could be employed to fabricate SAPs-drug complex, which could achieve slow release, targeted or environment-responsive delivery of drugs. Furthermore, some SAPs could also be combined with other types of materials for drug delivery, or even act as drug by themselves. CONCLUSION Various types of SAPs have been designed and used for drug delivery following various strategies, suggesting that SAPs as a category of versatile nanomaterials have promising potential in the field of nanomedicine.
Collapse
Affiliation(s)
- Fei Peng
- Laboratory of Anaesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wensheng Zhang
- Laboratory of Anaesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Feng Qiu
- Laboratory of Anaesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
5
|
Hamann A, Thomas AK, Kozisek T, Farris E, Lück S, Zhang Y, Pannier AK. Screening a chemically defined extracellular matrix mimetic substrate library to identify substrates that enhance substrate-mediated transfection. Exp Biol Med (Maywood) 2020; 245:606-619. [PMID: 32183552 DOI: 10.1177/1535370220913501] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Nonviral gene delivery, though limited by inefficiency, has extensive utility in cell therapy, tissue engineering, and diagnostics. Substrate-mediated gene delivery (SMD) increases efficiency and allows transfection at a cell-biomaterial interface, by immobilizing and concentrating nucleic acid complexes on a surface. Efficient SMD generally requires substrates to be coated with serum or other protein coatings to mediate nucleic acid complex immobilization, as well as cell adhesion and growth; however, this strategy limits reproducibility and may be difficult to translate for clinical applications. As an alternative, we screened a chemically defined combinatorial library of 20 different extracellular matrix mimetic substrates containing combinations of (1) different sulfated polysaccharides that are essential extracellular matrix glycosaminoglycans (GAGs), with (2) mimetic peptides derived from adhesion proteins, growth factors, and cell-penetrating domains, for use as SMD coatings. We identified optimal substrates for DNA lipoplex and polyplex SMD transfection of fibroblasts and human mesenchymal stem cells. Optimal extracellular matrix mimetic substrates varied between cell type, donor source, and transfection reagent, but typically contained Heparin GAG and an adhesion peptide. Multiple substrates significantly increased transgene expression (i.e. 2- to 20-fold) over standard protein coatings. Considering previous research of similar ligands, we hypothesize extracellular matrix mimetic substrates modulate cell adhesion, proliferation, and survival, as well as plasmid internalization and trafficking. Our results demonstrate the utility of screening combinatorial extracellular matrix mimetic substrates for optimal SMD transfection towards application- and patient-specific technologies. Impact statement Substrate-mediated gene delivery (SMD) approaches have potential for modification of cells in applications where a cell-material interface exists. Conventional SMD uses ill-defined serum or protein coatings to facilitate immobilization of nucleic acid complexes, cell attachment, and subsequent transfection, which limits reproducibility and clinical utility. As an alternative, we screened a defined library of extracellular matrix mimetic substrates containing combinations of different glycosaminoglycans and bioactive peptides to identify optimal substrates for SMD transfection of fibroblasts and human mesenchymal stem cells. This strategy could be utilized to develop substrates for specific SMD applications in which variability exists between different cell types and patient samples.
Collapse
Affiliation(s)
- Andrew Hamann
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Alvin K Thomas
- B CUBE - Center for Molecular Bioengineering, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 41, Dresden 01307, Germany
| | - Tyler Kozisek
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Eric Farris
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Steffen Lück
- B CUBE - Center for Molecular Bioengineering, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 41, Dresden 01307, Germany
| | - Yixin Zhang
- B CUBE - Center for Molecular Bioengineering, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 41, Dresden 01307, Germany
| | - Angela K Pannier
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| |
Collapse
|
6
|
Yao L, Wang X, Weng W, Fu Y, Cheng K. Bioactive nanocomposite coatings under visible light illumination promoted surface-mediated gene delivery. Biomater Sci 2020; 8:3685-3696. [DOI: 10.1039/d0bm00123f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bioactive coatings with photothermal conversion ability were used to spatially and temporally control surface-mediated gene delivery under visible light illumination.
Collapse
Affiliation(s)
- Lili Yao
- School of Materials Science and Engineering
- State Key Laboratory of Silicon Materials
- Cyrus Tang Center for Sensor Materials and Applications
- Zhejiang University
- Hangzhou
| | - Xiaozhao Wang
- Department of Orthopaedic Surgery
- Second Affiliated Hospital & Zhejiang University-University of Edinburgh Institute & School of Basic Medicine
- Zhejiang University School of Medicine
- Hangzhou 310058
- China
| | - Wenjian Weng
- School of Materials Science and Engineering
- State Key Laboratory of Silicon Materials
- Cyrus Tang Center for Sensor Materials and Applications
- Zhejiang University
- Hangzhou
| | - Yongqing Fu
- Faculty of Engineering & Environment
- Northumbria University
- Newcastle upon Tyne
- UK
| | - Kui Cheng
- School of Materials Science and Engineering
- State Key Laboratory of Silicon Materials
- Cyrus Tang Center for Sensor Materials and Applications
- Zhejiang University
- Hangzhou
| |
Collapse
|
7
|
Ni R, Feng R, Chau Y. Synthetic Approaches for Nucleic Acid Delivery: Choosing the Right Carriers. Life (Basel) 2019; 9:E59. [PMID: 31324016 PMCID: PMC6789897 DOI: 10.3390/life9030059] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 12/12/2022] Open
Abstract
The discovery of the genetic roots of various human diseases has motivated the exploration of different exogenous nucleic acids as therapeutic agents to treat these genetic disorders (inherited or acquired). However, the physicochemical properties of nucleic acids render them liable to degradation and also restrict their cellular entrance and gene translation/inhibition at the correct cellular location. Therefore, gene condensation/protection and guided intracellular trafficking are necessary for exogenous nucleic acids to function inside cells. Diversified cationic formulation materials, including natural and synthetic lipids, polymers, and proteins/peptides, have been developed to facilitate the intracellular transportation of exogenous nucleic acids. The chemical properties of different formulation materials determine their special features for nucleic acid delivery, so understanding the property-function correlation of the formulation materials will inspire the development of next-generation gene delivery carriers. Therefore, in this review, we focus on the chemical properties of different types of formulation materials and discuss how these formulation materials function as protectors and cellular pathfinders for nucleic acids, bringing them to their destination by overcoming different cellular barriers.
Collapse
Affiliation(s)
- Rong Ni
- Department of Chemical and Biological Engineering, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong 999077, China
- Institute for Advanced Study, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong 999077, China
| | - Ruilu Feng
- Department of Chemical and Biological Engineering, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong 999077, China
| | - Ying Chau
- Department of Chemical and Biological Engineering, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong 999077, China.
| |
Collapse
|
8
|
Mantz A, Rosenthal A, Farris E, Kozisek T, Bittrich E, Nazari S, Schubert E, Schubert M, Stamm M, Uhlmann P, Pannier AK. Free Polyethylenimine Enhances Substrate-Mediated Gene Delivery on Titanium Substrates Modified With RGD-Functionalized Poly(acrylic acid) Brushes. Front Chem 2019; 7:51. [PMID: 30792979 PMCID: PMC6374293 DOI: 10.3389/fchem.2019.00051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/18/2019] [Indexed: 01/08/2023] Open
Abstract
Substrate mediated gene delivery (SMD) is a method of immobilizing DNA complexes to a substrate via covalent attachment or nonspecific adsorption, which allows for increased transgene expression with less DNA compared to traditional bolus delivery. It may also increase cells receptivity to transfection via cell-material interactions. Substrate modifications with poly(acrylic) acid (PAA) brushes may improve SMD by enhancing substrate interactions with DNA complexes via tailored surface chemistry and increasing cellular adhesion via moieties covalently bound to the brushes. Previously, we described a simple method to graft PAA brushes to Ti and further demonstrated conjugation of cell adhesion peptides (i.e., RGD) to the PAA brushes to improve biocompatibility. The objective of this work was to investigate the ability of Ti substrates modified with PAA-RGD brushes (PAA-RGD) to immobilize complexes composed of branched polyethyleneimine and DNA plasmids (bPEI-DNA) and support SMD in NIH/3T3 fibroblasts. Transfection in NIH/3T3 cells cultured on bPEI-DNA complexes immobilized onto PAA-RGD substrates was measured and compared to transfection in cells cultured on control surfaces with immobilized complexes including Flat Ti, PAA brushes modified with a control peptide (RGE), and unmodified PAA. Transfection was two-fold higher in cells cultured on PAA-RGD compared to those cultured on all control substrates. While DNA immobilization measured with radiolabeled DNA indicated that all substrates (PAA-RGD, unmodified PAA, Flat Ti) contained nearly equivalent amounts of loaded DNA, ellipsometric measurements showed that more total mass (i.e., DNA and bPEI, both complexed and free) was immobilized to PAA and PAA-RGD compared to Flat Ti. The increase in adsorbed mass may be attributed to free bPEI, which has been shown to improve transfection. Further transfection investigations showed that removing free bPEI from the immobilized complexes decreased SMD transfection and negated any differences in transfection success between cells cultured on PAA-RGD and on control substrates, suggesting that free bPEI may be beneficial for SMD in cells cultured on bPEI-DNA complexes immobilized on PAA-RGD grafted to Ti. This work demonstrates that substrate modification with PAA-RGD is a feasible method to enhance SMD outcomes on Ti and may be used for future applications such as tissue engineering, gene therapy, and diagnostics.
Collapse
Affiliation(s)
- Amy Mantz
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
- Center for Nanohybrid Functional Materials, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Alice Rosenthal
- Leibniz-Institut für Polymerforschung Dresden e.V., Dresden, Germany
- Institute of Polymeric Materials, Technische Universität Dresden, Dresden, Germany
| | - Eric Farris
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Tyler Kozisek
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Eva Bittrich
- Leibniz-Institut für Polymerforschung Dresden e.V., Dresden, Germany
| | - Saghar Nazari
- Leibniz-Institut für Polymerforschung Dresden e.V., Dresden, Germany
| | - Eva Schubert
- Center for Nanohybrid Functional Materials, University of Nebraska-Lincoln, Lincoln, NE, United States
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Mathias Schubert
- Center for Nanohybrid Functional Materials, University of Nebraska-Lincoln, Lincoln, NE, United States
- Leibniz-Institut für Polymerforschung Dresden e.V., Dresden, Germany
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
- Department of Physics, Chemistry, and Biology, Linkoping University, Linkoping, Sweden
- Terahertz Materials Analysis Center (THeMAC), Linkoping University, Linkoping, Sweden
| | - Manfred Stamm
- Leibniz-Institut für Polymerforschung Dresden e.V., Dresden, Germany
- Institute of Polymeric Materials, Technische Universität Dresden, Dresden, Germany
| | - Petra Uhlmann
- Leibniz-Institut für Polymerforschung Dresden e.V., Dresden, Germany
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Angela K. Pannier
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
- Center for Nanohybrid Functional Materials, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
9
|
Mantz A, Pannier AK. Biomaterial substrate modifications that influence cell-material interactions to prime cellular responses to nonviral gene delivery. Exp Biol Med (Maywood) 2019; 244:100-113. [PMID: 30621454 PMCID: PMC6405826 DOI: 10.1177/1535370218821060] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
IMPACT STATEMENT This review summarizes how biomaterial substrate modifications (e.g. chemical modifications like natural coatings, ligands, or functional side groups, and/or physical modifications such as topography or stiffness) can prime the cellular response to nonviral gene delivery (e.g. affecting integrin binding and focal adhesion formation, cytoskeletal remodeling, endocytic mechanisms, and intracellular trafficking), to aid in improving gene delivery for applications where a cell-material interface might exist (e.g. tissue engineering scaffolds, medical implants and devices, sensors and diagnostics, wound dressings).
Collapse
Affiliation(s)
- Amy Mantz
- Department of Biological Systems Engineering,
University
of Nebraska-Lincoln, Lincoln, NE 68583,
USA
| | - Angela K Pannier
- Department of Biological Systems Engineering,
University
of Nebraska-Lincoln, Lincoln, NE 68583,
USA
| |
Collapse
|
10
|
Liu K, Zheng L, Ma C, Göstl R, Herrmann A. DNA-surfactant complexes: self-assembly properties and applications. Chem Soc Rev 2018; 46:5147-5172. [PMID: 28686247 DOI: 10.1039/c7cs00165g] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Over the last few years, DNA-surfactant complexes have gained traction as unique and powerful materials for potential applications ranging from optoelectronics to biomedicine because they self-assemble with outstanding flexibility spanning packing modes from ordered lamellar, hexagonal and cubic structures to disordered isotropic phases. These materials consist of a DNA backbone from which the surfactants protrude as non-covalently bound side chains. Their formation is electrostatically driven and they form bulk films, lyotropic as well as thermotropic liquid crystals and hydrogels. This structural versatility and their easy-to-tune properties render them ideal candidates for assembly in bulk films, for example granting directional conductivity along the DNA backbone, for dye dispersion minimizing fluorescence quenching allowing applications in lasing and nonlinear optics or as electron blocking and hole transporting layers, such as in LEDs or photovoltaic cells, owing to their extraordinary dielectric properties. However, they do not only act as host materials but also function as a chromophore itself. They can be employed within electrochromic DNA-surfactant liquid crystal displays exhibiting remarkable absorptivity in the visible range whose volatility can be controlled by the external temperature. Concomitantly, applications in the biological field based on DNA-surfactant bulk films, liquid crystals and hydrogels are rendered possible by their excellent gene and drug delivery capabilities. Beyond the mere exploitation of their material properties, DNA-surfactant complexes proved outstandingly useful for synthetic chemistry purposes when employed as scaffolds for DNA-templated reactions, nucleic acid modifications or polymerizations. These promising examples are by far not exhaustive but foreshadow their potential applications in yet unexplored fields. Here, we will give an insight into the peculiarities and perspectives of each material and are confident to inspire future developments and applications employing this emerging substance class.
Collapse
Affiliation(s)
- Kai Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry of Chinese Academy of Sciences, 130022, Changchun, China
| | | | | | | | | |
Collapse
|
11
|
Li BC, Chang H, Ren KF, Ji J. Substrate-mediated delivery of gene complex nanoparticles via polydopamine coating for enhancing competitiveness of endothelial cells. Colloids Surf B Biointerfaces 2016; 147:172-179. [PMID: 27500360 DOI: 10.1016/j.colsurfb.2016.07.063] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/28/2016] [Accepted: 07/29/2016] [Indexed: 11/27/2022]
Abstract
Substrate-mediated delivery of functional plasmid DNA (pDNA) has been proven to be a promising strategy to promote competitiveness of endothelial cells (ECs) over smooth muscle cells (SMCs), which is beneficial to inducing fast endothelialization of implanted vascular devices. Thus, it is of great importance to develop universal approaches with simplicity and easiness to immobilize DNA complex nanoparticles on substrates. In this study, the bioinspired polydopamine (PDA) coating was employed in immobilization of DNA complex nanoparticles, which were composed of protamine (PrS) and plasmid DNA encoding with hepatocyte growth factor (HGF-pDNA) gene. We demonstrated that the DNA complex nanoparticles can be successfully immobilized onto the PDA surface. Consequently, the HGF expression of both ECs and SMCs were significantly improved when they cultured on the DNA complex nanoparticles-immobilized substrates. Furthermore, EC proliferation was specifically promoted due to bioactivity of HGF, leading to an enhancement of EC competitiveness over SMCs. Our findings demonstrated the substrate-mediated functional gene nanoparticle delivery through PDA coating as a simple and efficient approach. It may hold great potential in the field of interventional cardiovascular implants.
Collapse
Affiliation(s)
- Bo-Chao Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Hao Chang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Ke-Feng Ren
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
12
|
Wang Z, Wu H, Shi H, Wang M, Huang C, Jia N. A novel multifunctional biomimetic Au@BSA nanocarrier as a potential siRNA theranostic nanoplatform. J Mater Chem B 2016; 4:2519-2526. [DOI: 10.1039/c5tb02326b] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A novel siRNA nanocarrier based on biomimetic Au@BSA nanoflowers is fabricated which could serve as a potential theranostic nanoplatform.
Collapse
Affiliation(s)
- Zhiming Wang
- The Education Ministry Key Laboratory of Resource Chemistry
- Shanghai Key Laboratory of Rare Earth Functional Materials and Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors
- Department of Chemistry
- Shanghai Normal University
- Shanghai 200234
| | - Hui Wu
- The Education Ministry Key Laboratory of Resource Chemistry
- Shanghai Key Laboratory of Rare Earth Functional Materials and Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors
- Department of Chemistry
- Shanghai Normal University
- Shanghai 200234
| | - Hongyuan Shi
- Department of Radiology
- Ruijin Hospital
- School of Medicine
- Shanghai Jiaotong University
- Shanghai 200025
| | - Mingliang Wang
- Department of Radiology
- Zhongshan Hospital
- School of Medicine
- Fudan University
- Shanghai 200032
| | - Chusen Huang
- The Education Ministry Key Laboratory of Resource Chemistry
- Shanghai Key Laboratory of Rare Earth Functional Materials and Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors
- Department of Chemistry
- Shanghai Normal University
- Shanghai 200234
| | - Nengqin Jia
- The Education Ministry Key Laboratory of Resource Chemistry
- Shanghai Key Laboratory of Rare Earth Functional Materials and Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors
- Department of Chemistry
- Shanghai Normal University
- Shanghai 200234
| |
Collapse
|
13
|
Studies of the physicochemical and structural properties of self-assembling cationic pyridine derivatives as gene delivery agents. Chem Phys Lipids 2015; 191:25-37. [PMID: 26272034 DOI: 10.1016/j.chemphyslip.2015.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/05/2015] [Accepted: 08/06/2015] [Indexed: 11/21/2022]
Abstract
New amphiphilic pyridine derivatives containing dodecyloxycarbonyl substituents at positions 3 and 5 and cationic moieties at positions 2 and 6 have been designed and synthesised. Compounds of this type can be considered as synthetic lipids. The corresponding 1,4-dihydropyridine (1,4-DHP) derivatives have earlier been proposed as a promising tool for plasmid DNA (pDNA) delivery in vitro. In this work studies of the self-assembling properties of amphiphilic pyridine derivatives leading to the formation of liposomes, determination of particle size, zeta-potential and critical micelle concentration (CMC) with dynamic light scattering (DLS) measurements are described. Furthermore, thermal analysis of pyridine derivatives was performed using thermogravimetry analysis (TGA) and differential thermal analysis (DTA) as well as the ability to deliver the pEGFP-C1 plasmid DNA (that encodes GFP reporter) into the Baby hamster kidney-derived (BHK-21) cell line was used for evaluation of gene delivery properties. We have revealed that the new pyridine derivatives possessed self-assembling properties which were proved by formation of nanoparticles with the average size from 115 to 743nm, the zeta-potentials in the range of 48-79mV and CMC values in the range of 2-67μM. DTA data showed that all processes were endothermic for all compounds. Additionally, we established that among the tested pyridines the representatives with N-methylpyrrolidinium or pyridinium moieties as cationic head-group at the positions 2 and 6 possessed higher pEGFP-C1 transfection activity into the BHK-21 cell line. Nevertheless, the obtained results indicated that correlation of the physicochemical, structural properties and gene delivery activities of the tested compounds were not completely elucidated yet. On the other hand, the synthesised pyridines as possible metabolites of promising delivery systems on the 1,4-DHP core possessed lower pDNA transfection activity than the corresponding 1,4-DHP amphiphiles.
Collapse
|
14
|
Zhao Y, Zhang S, Zhang Y, Cui S, Chen H, Zhi D, Zhen Y, Zhang S, Huang L. Tri-peptide cationic lipids for gene delivery. J Mater Chem B 2015; 3:119-126. [PMID: 25580248 PMCID: PMC4285367 DOI: 10.1039/c4tb01312c] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Several novel tri-peptide cationic lipids were designed and synthesized for delivering DNA and siRNA. They have tri-lysine and tri-ornithine as head groups, carbamate group as linker and 12 and 14 carbon atom alkyl groups as tails. These tri-peptide cationic lipids were prepared into cationic liposomes for the study of the physicochemical properties and gene delivery. Their particle size, Zeta potential and DNA-binding were characterized to show that they were suitable for gene transfection. The further results indicate that these lipids can transfer DNA and siRNA very efficiently into NCI-H460 and Hep-2 tumor cells. The selected lipid, CDO14, was able to deliver combined siRNAs against c-Myc and VEGF for silencing distinct oncogenic pathways in lung tumors of mice, with little in vitro and in vivo toxicity.
Collapse
Affiliation(s)
- Yinan Zhao
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116021, Liaoning, China
- SEAC-ME Key Laboratory of Biotechnology and Bio-resources Utilization, Dalian Nationalities University, Dalian 116600, Liaoning, China
| | - Shubiao Zhang
- SEAC-ME Key Laboratory of Biotechnology and Bio-resources Utilization, Dalian Nationalities University, Dalian 116600, Liaoning, China
| | - Yuan Zhang
- Department of Materials Science and Engineering, Department of Biological Engineering, The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shaohui Cui
- SEAC-ME Key Laboratory of Biotechnology and Bio-resources Utilization, Dalian Nationalities University, Dalian 116600, Liaoning, China
| | - Huiying Chen
- SEAC-ME Key Laboratory of Biotechnology and Bio-resources Utilization, Dalian Nationalities University, Dalian 116600, Liaoning, China
| | - Defu Zhi
- SEAC-ME Key Laboratory of Biotechnology and Bio-resources Utilization, Dalian Nationalities University, Dalian 116600, Liaoning, China
| | - Yuhong Zhen
- College of Phamacy, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Shufen Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116021, Liaoning, China
| | - Leaf Huang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
15
|
Monteiro N, Martins A, Reis RL, Neves NM. Liposomes in tissue engineering and regenerative medicine. J R Soc Interface 2014; 11:20140459. [PMID: 25401172 PMCID: PMC4223894 DOI: 10.1098/rsif.2014.0459] [Citation(s) in RCA: 216] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 10/02/2014] [Indexed: 01/13/2023] Open
Abstract
Liposomes are vesicular structures made of lipids that are formed in aqueous solutions. Structurally, they resemble the lipid membrane of living cells. Therefore, they have been widely investigated, since the 1960s, as models to study the cell membrane, and as carriers for protection and/or delivery of bioactive agents. They have been used in different areas of research including vaccines, imaging, applications in cosmetics and tissue engineering. Tissue engineering is defined as a strategy for promoting the regeneration of tissues for the human body. This strategy may involve the coordinated application of defined cell types with structured biomaterial scaffolds to produce living structures. To create a new tissue, based on this strategy, a controlled stimulation of cultured cells is needed, through a systematic combination of bioactive agents and mechanical signals. In this review, we highlight the potential role of liposomes as a platform for the sustained and local delivery of bioactive agents for tissue engineering and regenerative medicine approaches.
Collapse
Affiliation(s)
- Nelson Monteiro
- 3B's Research Group—Biomaterials, Biodegradables and Biomimetics Department of Polymer Engineering, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra S. Cláudio do Barco, 4806-909, Caldas das Taipas, Guimarães, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Albino Martins
- 3B's Research Group—Biomaterials, Biodegradables and Biomimetics Department of Polymer Engineering, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra S. Cláudio do Barco, 4806-909, Caldas das Taipas, Guimarães, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group—Biomaterials, Biodegradables and Biomimetics Department of Polymer Engineering, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra S. Cláudio do Barco, 4806-909, Caldas das Taipas, Guimarães, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno M. Neves
- 3B's Research Group—Biomaterials, Biodegradables and Biomimetics Department of Polymer Engineering, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra S. Cláudio do Barco, 4806-909, Caldas das Taipas, Guimarães, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
16
|
Wang C, Li L, Ma L, Li B, Gao C. Biotin-triggered release and transfection of DNA complexes immobilized on a substrate via biotin–avidin interaction. J BIOACT COMPAT POL 2014. [DOI: 10.1177/0883911514528147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In this study, the highly specific biotin–avidin interaction was used to immobilize DNA complexes to a substrate. An excess of biotin was added to trigger the dissociation of DNA complexes from the substrate to mediate their release and transfection. Biotin-grafted-polyethyleneimine/DNA complexes with N/P ratio of 5 were prepared with diameter of 170.2 nm and ζ-potential of 16.1 mV. The DNA immobilized substrates were fabricated using a biotin–avidin–biotin sandwich model, which were characterized by atom force microscope and fluorescent microscope. Compared to DNA immobilization by physical adsorption, a higher DNA density of 935 ng/cm2 was observed on biotinylated substrates. Based on the in vitro release profiles, the DNA complexes immobilized on silanized substrate released faster than those on biotinylated substrate. Triggered by the addition of extra biotin, more DNA complexes were released. The transfection efficiencies of the DNA complexes immobilized on different substrates were assayed on HEK-293T cells. The highest transfection efficiency was obtained in the group of biotinylated substrate with the trigger of extra biotin. Thus, the system of demobilized DNA complexes onto a substrate by the biotin–avidin interaction and the dissociation of DNA complexes from a substrate triggered by the extra biotin provides a promising strategy for the realization of the controlled release and enhanced transgene expression of genes.
Collapse
Affiliation(s)
- Chunfen Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Luyan Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Lie Ma
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Bo Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
Mahato M, Yadav S, Kumar P, Sharma AK. Synthesis and evaluation of tetramethylguanidinium-polyethylenimine polymers as efficient gene delivery vectors. BIOMED RESEARCH INTERNATIONAL 2014; 2014:459736. [PMID: 24864245 PMCID: PMC4017721 DOI: 10.1155/2014/459736] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 03/27/2014] [Indexed: 12/11/2022]
Abstract
Previously, we demonstrated that 6-(N,N,N',N'-tetramethylguanidinium chloride)-hexanoyl-polyethylenimine (THP) polymers exhibited significantly enhanced transfection efficiency and cell viability. Here, in the present study, we have synthesized a series of N,N,N',N'-tetramethylguanidinium-polyethylenimine (TP1-TP5) polymers via a single-step reaction involving peripheral primary amines of bPEI and varying amounts of 2-(1H-benzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HBTU). These polymers were found to interact efficiently with negatively charged pDNA and formed stable complexes in the size range of ~240-450 nm. Acid-base titration profiles revealed improved buffering capacity of TP polymers as compared to bPEI. Transfection and cytotoxicity assays performed with TP/pDNA complexes on HEK293, CHO, and HeLa cells showed significantly higher transfection efficiency and cell viability with one of the complexes, TP2/pDNA complex, exhibited the highest transfection efficiency (~1.4-2.3-fold) outcompeting native bPEI and the commercially available transfection reagent, Lipofectamine 2000. Compared to previously reported THP polymers, the transfection efficiency of TP/pDNA complexes was found to be lower, as examined by flow cytometry. These results highlight the importance of the hydrophobic C-6 linker in THP polymers in forming compact nanostructures with pDNA, which might lead to efficient uptake and internalization of the complexes; however, the projected TP polymers offer an advantage of their rapid and economical one-step synthesis.
Collapse
Affiliation(s)
- Manohar Mahato
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Delhi University Campus, Mall Road, Delhi 110 007, India
| | - Santosh Yadav
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Delhi University Campus, Mall Road, Delhi 110 007, India
| | - Pradeep Kumar
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Delhi University Campus, Mall Road, Delhi 110 007, India
| | - Ashwani Kumar Sharma
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Delhi University Campus, Mall Road, Delhi 110 007, India
| |
Collapse
|
18
|
Yadav S, Mahato M, Pathak R, Jha D, Kumar B, Deka SR, Gautam HK, Sharma AK. Multifunctional self-assembled cationic peptide nanostructures efficiently carry plasmid DNA in vitro and exhibit antimicrobial activity with minimal toxicity. J Mater Chem B 2014; 2:4848-4861. [DOI: 10.1039/c4tb00657g] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An amphiphilic peptide–aminoglycoside (Pep–Neo) conjugate has been synthesized, self-assembled into nanostructures and evaluated for its multifunctional properties.
Collapse
Affiliation(s)
- Santosh Yadav
- Nucleic Acids Research Laboratory
- CSIR-Institute of Genomics and Integrative Biology
- Delhi – 110007, India
| | - Manohar Mahato
- Nucleic Acids Research Laboratory
- CSIR-Institute of Genomics and Integrative Biology
- Delhi – 110007, India
| | - Rajiv Pathak
- Microbial Biotechnology Laboratory
- CSIR-Institute of Genomics and Integrative Biology
- Delhi – 110020, India
| | - Diksha Jha
- Microbial Biotechnology Laboratory
- CSIR-Institute of Genomics and Integrative Biology
- Delhi – 110020, India
| | - Bipul Kumar
- Microbial Biotechnology Laboratory
- CSIR-Institute of Genomics and Integrative Biology
- Delhi – 110020, India
| | - Smriti Rekha Deka
- Nucleic Acids Research Laboratory
- CSIR-Institute of Genomics and Integrative Biology
- Delhi – 110007, India
| | - Hemant Kumar Gautam
- Microbial Biotechnology Laboratory
- CSIR-Institute of Genomics and Integrative Biology
- Delhi – 110020, India
| | - Ashwani Kumar Sharma
- Nucleic Acids Research Laboratory
- CSIR-Institute of Genomics and Integrative Biology
- Delhi – 110007, India
| |
Collapse
|
19
|
Oh JS, Park M, Kim JS, Jang JH. Enhanced cellular transfection by ternary non-viral gene vectors coupled with adeno-associated virus-derived peptides. Macromol Biosci 2013; 14:121-30. [PMID: 23966357 DOI: 10.1002/mabi.201300296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 07/15/2013] [Indexed: 11/08/2022]
Abstract
The establishment of efficient and safe gene delivery systems is crucial for biomedical applications. To address this objective, novel, ternary hybrid gene vectors are designed with viral capsid peptides in non-viral gene carriers. The viral peptide, TQVGQKT, is coupled with a membrane active peptide, LK15. Which acts as a linker to tag peptide with plasmid DNA. Additionally, polyethylenimine (PEI) is employed to condense the complexes further, thereby forming ternary DNA/TQVGQKT-LK15/PEI complexes. The ternary complexes result in rapid internalization leading to significantly enhanced cellular transfection. The new moiety, TQVGQKT, as well as enhanced cellular transfection, will certainly provide crucial insights for the design of novel non-viral gene carriers with efficient and safe properties.
Collapse
Affiliation(s)
- Ji-Seon Oh
- Department of Chemical and Biomolecular Engineering, Yonsei University, 120-749, Seoul, Korea
| | | | | | | |
Collapse
|
20
|
Kim JS, Kim E, Oh JS, Jang JH. Integration of Adeno-Associated Virus-Derived Peptides into Nonviral Vectors to Synergistically Enhance Cellular Transfection. Biomacromolecules 2013; 14:2136-45. [DOI: 10.1021/bm4005854] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Jung-Suk Kim
- Department of Chemical
and Biomolecular
Engineering, Yonsei University, Seoul, Korea, 120-749
| | - Eunmi Kim
- Department of Chemical
and Biomolecular
Engineering, Yonsei University, Seoul, Korea, 120-749
| | - Ji-Seon Oh
- Department of Chemical
and Biomolecular
Engineering, Yonsei University, Seoul, Korea, 120-749
| | - Jae-Hyung Jang
- Department of Chemical
and Biomolecular
Engineering, Yonsei University, Seoul, Korea, 120-749
| |
Collapse
|
21
|
Holmes CA, Tabrizian M. Substrate-mediated gene delivery from glycol-chitosan/hyaluronic acid polyelectrolyte multilayer films. ACS APPLIED MATERIALS & INTERFACES 2013; 5:524-531. [PMID: 23281737 DOI: 10.1021/am303029k] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Substrate-mediated transfection is one of the key strategies for localized gene delivery. Layer-by-layer (LbL) polyelectrolyte deposition is a promising technique which enables controlled delivery of a number of biofactors, including nucleic acids. Here, we embed lipoplexes containing plasmid DNA within polyelectrolyte multilayers composed of glycol-chitosan (Glyc-CHI) and hyaluronic acid (HA) in order to produce a film system that enables localized, surface-based transfection. The topography and morphology of the resulting multilayers were characterized after lipoplex absorption and during subsequent film build-up via atomic force microscopy (AFM) and scanning electron microscopy (SEM), respectively. DNA embedding efficiency and release were then examined. Lipoplex-containing Glyc-CHI/HA films were found to successfully transfect NIH3T3 fibroblasts and HEK293 kidney cells in vitro, maintaining transfection levels of approximately 20% for a period of at least 7 days.
Collapse
Affiliation(s)
- Christina A Holmes
- Department of Biomedical Engineering, Duff Medical Science Building, 3775 University Street, McGill University, Montreal, H3A 2B4, Canada
| | | |
Collapse
|
22
|
Pajuste K, Hyvönen Z, Petrichenko O, Kaldre D, Rucins M, Cekavicus B, Ose V, Skrivele B, Gosteva M, Morin-Picardat E, Plotniece M, Sobolev A, Duburs G, Ruponen M, Plotniece A. Gene delivery agents possessing antiradical activity: self-assembling cationic amphiphilic 1,4-dihydropyridine derivatives. NEW J CHEM 2013. [DOI: 10.1039/c3nj00272a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
23
|
Mahato M, Kumar P, Sharma AK. Amphiphilic polyethylenimine polymers mediate efficient delivery of DNA and siRNA in mammalian cells. MOLECULAR BIOSYSTEMS 2013; 9:780-91. [DOI: 10.1039/c3mb25444e] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
24
|
Hu YL, Huang B, Zhang TY, Miao PH, Tang GP, Tabata Y, Gao JQ. Mesenchymal stem cells as a novel carrier for targeted delivery of gene in cancer therapy based on nonviral transfection. Mol Pharm 2012; 9:2698-709. [PMID: 22862421 DOI: 10.1021/mp300254s] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The success of gene therapy relies largely on an effective targeted gene delivery system. Till recently, more and more targeted delivery carriers, such as liposome, nanoparticles, microbubbles, etc., have been developed. However, the clinical applications of these systems were limited for their several disadvantages. Therefore, design and development of novel drug/gene delivery vehicles became a hot topic. Cell-based delivery systems are emerging as an alternative for the targeted delivery system as we described previously. Mesenchymal stem cells (MSCs) are an attractive cell therapy carrier for the delivery of therapeutic agents into tumor sites mainly for their tumor-targeting capacities. In the present study, a nonviral vector, PEI(600)-Cyd, prepared by linking low molecular weight polyethylenimine (PEI) and β-cyclodextrin (β-CD), was used to introduce the therapeutical gene, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), to MSCs. Meanwhile, the characterization, transfection efficiency, cytotoxicity, cellular internalization, and its mechanism of this nonviral vector were evaluated. The in vitro expression of TRAIL from MSCs-TRAIL was demonstrated by both enzyme-linked immunosorbent assay and Western blot analysis. The lung tumor homing ability of MSCs was further confirmed by the in vitro and in vivo model. Moreover, the therapeutic effects as well as the safety of MSCs-TRAIL on lung metastases bearing C57BL/6 mice and normal C57BL/6 mice were also demonstrated. Our results supported both the effectiveness of nonviral vectors in transferring the therapeutic gene to MSCs and the feasibility of using MSCs as a targeted gene delivery carrier, indicating that MSCs could be a promising tumor target delivery vehicle in cancer gene therapy based on nonviral gene recombination.
Collapse
Affiliation(s)
- Yu-Lan Hu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
25
|
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]
|
26
|
Xu L, Liu Y, Chen Z, Li W, Liu Y, Wang L, Liu Y, Wu X, Ji Y, Zhao Y, Ma L, Shao Y, Chen C. Surface-engineered gold nanorods: promising DNA vaccine adjuvant for HIV-1 treatment. NANO LETTERS 2012; 12:2003-12. [PMID: 22372996 DOI: 10.1021/nl300027p] [Citation(s) in RCA: 207] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
With the intense international response to the AIDS pandemic, HIV vaccines have been extensively investigated but have failed due to issues of safety or efficacy in humans. Adjuvants for HIV/AIDS vaccines are under intense research but a rational design approach is still lacking. Nanomaterials represent an obvious opportunity in this field due to their unique physicochemical properties. Gold nanostructures are being actively studied as a promising and versatile platform for biomedical application. Herein, we report novel surface-engineered gold nanorods (NRs) used as promising DNA vaccine adjuvant for HIV treatment. We have exploited the effects of surface chemistry on the adjuvant activity of the gold nanorod by placing three kinds of molecules, that is, cetyltrimethylammonium bromide (CTAB), poly(diallydimethylammonium chloride) (PDDAC), and polyethyleneimine (PEI) on the surface of the nanorod. These PDDAC- or PEI-modified Au NRs can significantly promote cellular and humoral immunity as well as T cell proliferation through activating antigen-presenting cells if compared to naked HIV-1 Env plasmid DNA treatment in vivo. These findings have shed light on the rational design of low-toxic nanomaterials as a versatile platform for vaccine nanoadjuvants/delivery systems.
Collapse
Affiliation(s)
- Ligeng Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Wang JL, Tang GP, Shen J, Hu QL, Xu FJ, Wang QQ, Li ZH, Yang WT. A gene nanocomplex conjugated with monoclonal antibodies for targeted therapy of hepatocellular carcinoma. Biomaterials 2012; 33:4597-607. [PMID: 22469295 DOI: 10.1016/j.biomaterials.2012.02.045] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 02/25/2012] [Indexed: 11/16/2022]
Abstract
To enhance tumor-targeting abilities and therapeutic efficiency, a monoclonal antibody-conjugated gene nanocomplex was herein designed. The biodegradable cationic polyethylenimine-grafted-α,β-poly(N-3-hydroxypropyl)-DL-aspartamide (PHPA-PEI) was used for complexing pDNA to form the PHPA-PEI/pDNA nanoparticle, and then 9B9 mAb, an anti-epidermal growth factor receptor (anti-EGFR) monoclonal antibody, was conjugated to produce the PHPA-PEI/pDNA/9B9 mAb (PP9mN) complex. The PP9mN complex with the diameter of around 300 nm at its optimal weight ratio could be uptaken effectively by SMMC-7721 cells. The cytotoxicity of the PP9mN complex was much lower than that of PEI 25 kD in SMMC-7721, HepG2, Bel-7404 and COS-7 cell lines. The PP9mN complex possessed the highly efficient in vitro gene delivery ability to the hepatocellular carcinoma cells. The in vivo gene expression indicated that PP9mN could target to the tumor tissues effectively. By using the therapeutic AChE gene, it was found that the PP9mN complexes significantly enhanced the anti-tumor effect on tumor-bearing nude mice. Such monoclonal antibody-conjugated gene complex should have great potential applications in liver cancer therapy.
Collapse
Affiliation(s)
- J L Wang
- Institute of Chemical Biology and Pharmaceutical Chemistry, Zhejiang University, Hangzhou 310028, PR China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
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]
|
29
|
Opanasopit P, Tragulpakseerojn J, Apirakaramwong A, Ngawhirunpat T, Rojanarata T, Ruktanonchai U. The development of poly-L-arginine-coated liposomes for gene delivery. Int J Nanomedicine 2011; 6:2245-52. [PMID: 22114488 PMCID: PMC3215165 DOI: 10.2147/ijn.s25336] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
In this study, liposomes coated with cationic polymers, poly-L-arginine (PLA), were assessed as a promising gene transfer system in human cervical carcinoma (HeLa) cells and human hepatoma cell line (Huh7) cells. The liposomes were prepared using egg yolk phosphatidylcholine and sodium oleate in the molar ratio of 10:2 with an ultrasonic generator and then coated with PLA. The PLA-coated liposomes (PCLs) formed complexes with plasmid DNA encoding green fluorescent protein. The complexes were characterized by agarose gel electrophoresis and investigated for their transfection efficiency in HeLa and Huh7 cells. The data were compared with PLA/DNA complexes and the positive control Lipofectamine 2000™. The results showed that complete PCL/DNA complexes were formed at weight ratios of more than 0.05. Efficient gene transfer by PCLs was dependent on the cell type. The transfection efficiency of PCLs was about two times higher than that of PLA/DNA complexes in both HeLa cells and Huh7 cells. Cytotoxicity was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay and showed that 80%–100% of both of the cells were viable after treating PCL/DNA complexes. The present results demonstrate that PCLs are a promising, nonviral gene carrier with low toxicity.
Collapse
|
30
|
Gene delivery to tumor cells by cationic polymeric nanovectors coupled to folic acid and the cell-penetrating peptide octaarginine. Biomaterials 2011; 32:7253-62. [DOI: 10.1016/j.biomaterials.2011.06.015] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 06/08/2011] [Indexed: 11/21/2022]
|
31
|
Gajria S, Neumann T, Tirrell M. Self‐assembly and applications of nucleic acid solid‐state films. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2011; 3:479-500. [DOI: 10.1002/wnan.148] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Surekha Gajria
- Department of Chemistry, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Thorsten Neumann
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - Matthew Tirrell
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
32
|
Adler AF, Leong KW. Emerging links between surface nanotechnology and endocytosis: impact on nonviral gene delivery. NANO TODAY 2010; 5:553-569. [PMID: 21383869 PMCID: PMC3048656 DOI: 10.1016/j.nantod.2010.10.007] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Significant effort continues to be exerted toward the improvement of transfection mediated by nonviral vectors. These endeavors are often focused on the design of particulate carriers with properties that encourage efficient accumulation at the membrane surface, particle uptake, and endosomal escape. Despite its demonstrated importance in successful nonviral transfection, relatively little investigation has been done to understand the pressures driving internalized vectors into favorable nondegradative endocytic pathways. Improvements in transfection efficiency have been noted for complexes delivered with a substrate-mediated approach, but the reasons behind such enhancements remain unclear. The phenotypic changes exhibited by cells interacting with nano- and micro-featured substrates offer hints that may explain these effects. This review describes nanoscale particulate and substrate parameters that influence both the uptake of nonviral gene carriers and the endocytic phenotype of interacting cells, and explores the molecular links that may mediate these interactions. Substrate-mediated control of endocytosis represents an exciting new design parameter that will guide the creation of efficient transgene carriers.
Collapse
Affiliation(s)
- Andrew F. Adler
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Box 90281, Durham, NC 27708, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Box 90281, Durham, NC 27708, USA
| |
Collapse
|
33
|
Zhang X, Kovtun A, Mendoza-Palomares C, Oulad-Abdelghani M, Fioretti F, Rinckenbach S, Mainard D, Epple M, Benkirane-Jessel N. SiRNA-loaded multi-shell nanoparticles incorporated into a multilayered film as a reservoir for gene silencing. Biomaterials 2010; 31:6013-8. [PMID: 20488536 DOI: 10.1016/j.biomaterials.2010.04.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Accepted: 04/12/2010] [Indexed: 01/09/2023]
Abstract
In this study, we presented a new type of coating based on polyelectrolyte multilayers containing sequentially adsorbed active shRNA calcium phosphate nanoparticles for locally defined and temporarily variable gene silencing. Therefore, we investigated multi-shell calcium phosphate-shRNA nanoparticles embedded into a polyelectrolyte multilayer for gene silencing. As model system, we synthesized triple-shell calcium phosphate-shRNA nanoparticles (NP) and prepared polyelectrolyte multilayers films made of nanoparticles and poly-(L-lysine) (PLL). The biological activities of these polyelectrolyte multilayers films were tested by the production of osteopontin and osteocalcin in the human osteoblasts (HOb) which were cultivated on the PEM films. This new strategy can be used to efficiently control the bone formation and could be applicable in tissue engineering.
Collapse
Affiliation(s)
- Xin Zhang
- Institut National de la Santé et de la Recherche Médicale, INSERM, Unité 977, Faculté de Médecine, Strasbourg, France
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Liposomal gene delivery mediated by tissue-engineered scaffolds. Trends Biotechnol 2010; 28:28-36. [DOI: 10.1016/j.tibtech.2009.10.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 10/06/2009] [Accepted: 10/07/2009] [Indexed: 12/15/2022]
|