51
|
Manturthi S, Bhattacharya D, Sakhare KR, Narayan KP, Patri SV. Nicotinic acid-based cationic vectors for efficient gene delivery to glioblastoma cells. NEW J CHEM 2022. [DOI: 10.1039/d2nj03207d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A tocopherol-conjugated nicotinic acid-based lipid (NGT) was used for liposomal formation with the co-lipid DOPE and exhibited enhanced transfection of glioblastoma cells for eGFP and β-galactosidase protein expression.
Collapse
Affiliation(s)
- Shireesha Manturthi
- Department of Chemistry, National Institute of Technology Warangal, Hanamkonda, Telangana-506004, India
| | - Dwaipayan Bhattacharya
- Department of Biological Science, Bits pilani-hyderabad, Hyderabad, Telangana-500078, India
| | - Kalyani Rajesh Sakhare
- Department of Biological Science, Bits pilani-hyderabad, Hyderabad, Telangana-500078, India
| | - Kumar Pranav Narayan
- Department of Biological Science, Bits pilani-hyderabad, Hyderabad, Telangana-500078, India
| | - Srilakshmi V. Patri
- Department of Chemistry, National Institute of Technology Warangal, Hanamkonda, Telangana-506004, India
| |
Collapse
|
52
|
Jerzykiewicz J, Czogalla A. Polyethyleneimine-Based Lipopolyplexes as Carriers in Anticancer Gene Therapies. MATERIALS 2021; 15:ma15010179. [PMID: 35009324 PMCID: PMC8746209 DOI: 10.3390/ma15010179] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/06/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022]
Abstract
Recent years have witnessed rapidly growing interest in application of gene therapies for cancer treatment. However, this strategy requires nucleic acid carriers that are both effective and safe. In this context, non-viral vectors have advantages over their viral counterparts. In particular, lipopolyplexes—nanocomplexes consisting of nucleic acids condensed with polyvalent molecules and enclosed in lipid vesicles—currently offer great promise. In this article, we briefly review the major aspects of developing such non-viral vectors based on polyethyleneimine and outline their properties in light of anticancer therapeutic strategies. Finally, examples of current in vivo studies involving such lipopolyplexes and possibilities for their future development are presented.
Collapse
|
53
|
Liao S, Wei L, Abriata LA, Stellacci F. Control and Characterization of the Compactness of Single-Chain Nanoparticles. Macromolecules 2021. [DOI: 10.1021/acs.macromol.1c02071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Suiyang Liao
- Institute of Materials, École Polytechnique Fédérale de Lausanne, Station 12, 1015 Lausanne, Switzerland
| | - Lixia Wei
- Institute of Materials, École Polytechnique Fédérale de Lausanne, Station 12, 1015 Lausanne, Switzerland
| | - Luciano A. Abriata
- Protein Production and Structure Core Facility, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Francesco Stellacci
- Institute of Materials, École Polytechnique Fédérale de Lausanne, Station 12, 1015 Lausanne, Switzerland
- Interfaculty Bioengineering Institute, École Polytechnique Fédérale de Lausanne, Station 12, 1015 Lausanne, Switzerland
| |
Collapse
|
54
|
Asfiya R, Maiti B, Kamra M, Karande AA, Bhattacharya S. Novel α-tocopherol-ferrocene conjugates for the specific delivery of transgenes in liver cancer cells under high serum conditions. Biomater Sci 2021; 9:7636-7647. [PMID: 34676384 DOI: 10.1039/d1bm00607j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The delivery of therapeutic genes to a specific organ has drawn significant research attention. Among the pool of various delivery vectors, cationic liposomes (non-viral) are potential candidates for delivering therapeutic genes due to their low immunogenic response. Here, we have developed novel ferrocene-conjugated cationic tocopheryl aggregates as non-viral vectors. These formulations can transfer a reporter gene (pGL3; encoded for luciferase protein) specifically to liver cancer cells (HepG2 and Huh7) instead of non-hepatic cancer cells, such as Caco-2 (human colon carcinoma) and HeLa (cervical cancer) cells. The transfection efficiency (TE) of the optimum liposomal formulation is more significant than commercially available Lipofectamine 2000 (L2K). Notably, it retains its TE under high serum conditions (up to 50% FBS). A coupled effect from conjugated ferrocene and tocopherol in the cationic liposomal formulation might be responsible for the cell-specific delivery and higher serum compatibility. Therefore, the present proposed delivery system may provide a platform for further progress in terms of developing hepatotropic gene delivery systems.
Collapse
Affiliation(s)
- Rahmat Asfiya
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Bappa Maiti
- Technical Research Centre, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| | - Mohini Kamra
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Anjali Anoop Karande
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Santanu Bhattacharya
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, India. .,Technical Research Centre, Indian Association for the Cultivation of Science, Kolkata 700 032, India.,School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700 032, India
| |
Collapse
|
55
|
Kos S, Bosnjak M, Jesenko T, Markelc B, Kamensek U, Znidar K, Matkovic U, Rencelj A, Sersa G, Hudej R, Tuljak A, Peterka M, Cemazar M. Non-Clinical In Vitro Evaluation of Antibiotic Resistance Gene-Free Plasmids Encoding Human or Murine IL-12 Intended for First-in-Human Clinical Study. Pharmaceutics 2021; 13:pharmaceutics13101739. [PMID: 34684032 PMCID: PMC8539770 DOI: 10.3390/pharmaceutics13101739] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 11/30/2022] Open
Abstract
Interleukin 12 (IL-12) is a key cytokine that mediates antitumor activity of immune cells. To fulfill its clinical potential, the development is focused on localized delivery systems, such as gene electrotransfer, which can provide localized delivery of IL-12 to the tumor microenvironment. Gene electrotransfer of the plasmid encoding human IL-12 is already in clinical trials in USA, demonstrating positive results in the treatment of melanoma patients. To comply with EU regulatory requirements for clinical application, which recommend the use of antibiotic resistance gene-free plasmids, we constructed and developed the production process for the clinical grade quality antibiotic resistance gene-free plasmid encoding human IL-12 (p21-hIL-12-ORT) and its ortholog encoding murine IL-12 (p21-mIL-12-ORT). To demonstrate the suitability of the p21-hIL-12-ORT or p21-mIL-12-ORT plasmid for the first-in-human clinical trial, the biological activity of the expressed transgene, its level of expression and plasmid copy number were determined in vitro in the human squamous cell carcinoma cell line FaDu and the murine colon carcinoma cell line CT26. The results of the non-clinical evaluation in vitro set the basis for further in vivo testing and evaluation of antitumor activity of therapeutic molecules in murine models as well as provide crucial data for further clinical trials of the constructed antibiotic resistance gene-free plasmid in humans.
Collapse
Affiliation(s)
- Spela Kos
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (S.K.); (M.B.); (T.J.); (B.M.); (U.K.); (K.Z.); (U.M.); (A.R.); (G.S.)
| | - Masa Bosnjak
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (S.K.); (M.B.); (T.J.); (B.M.); (U.K.); (K.Z.); (U.M.); (A.R.); (G.S.)
- Faculty of Pharmacy, University of Ljubljana, Aškerceva ulica 7, SI-1000 Ljubljana, Slovenia
| | - Tanja Jesenko
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (S.K.); (M.B.); (T.J.); (B.M.); (U.K.); (K.Z.); (U.M.); (A.R.); (G.S.)
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Bostjan Markelc
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (S.K.); (M.B.); (T.J.); (B.M.); (U.K.); (K.Z.); (U.M.); (A.R.); (G.S.)
- Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia
| | - Urska Kamensek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (S.K.); (M.B.); (T.J.); (B.M.); (U.K.); (K.Z.); (U.M.); (A.R.); (G.S.)
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva ulica 101, SI-1000 Ljubljana, Slovenia
| | - Katarina Znidar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (S.K.); (M.B.); (T.J.); (B.M.); (U.K.); (K.Z.); (U.M.); (A.R.); (G.S.)
| | - Urska Matkovic
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (S.K.); (M.B.); (T.J.); (B.M.); (U.K.); (K.Z.); (U.M.); (A.R.); (G.S.)
| | - Andrej Rencelj
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (S.K.); (M.B.); (T.J.); (B.M.); (U.K.); (K.Z.); (U.M.); (A.R.); (G.S.)
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Gregor Sersa
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (S.K.); (M.B.); (T.J.); (B.M.); (U.K.); (K.Z.); (U.M.); (A.R.); (G.S.)
- Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia
| | - Rosana Hudej
- Center Odličnosti za Biosenzoriko, Instrumentacijo in Procesno Kontrolo, Mirce 21, SI-5270 Ajdovscina, Slovenia; (R.H.); (A.T.); (M.P.)
| | - Aneja Tuljak
- Center Odličnosti za Biosenzoriko, Instrumentacijo in Procesno Kontrolo, Mirce 21, SI-5270 Ajdovscina, Slovenia; (R.H.); (A.T.); (M.P.)
| | - Matjaz Peterka
- Center Odličnosti za Biosenzoriko, Instrumentacijo in Procesno Kontrolo, Mirce 21, SI-5270 Ajdovscina, Slovenia; (R.H.); (A.T.); (M.P.)
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, SI-1000 Ljubljana, Slovenia; (S.K.); (M.B.); (T.J.); (B.M.); (U.K.); (K.Z.); (U.M.); (A.R.); (G.S.)
- Faculty of Health Sciences, University of Primorska, Polje 42, SI-6310 Izola, Slovenia
- Correspondence: ; Tel.: +386-1-5879-544
| |
Collapse
|
56
|
Chis AA, Dobrea CM, Rus LL, Frum A, Morgovan C, Butuca A, Totan M, Juncan AM, Gligor FG, Arseniu AM. Dendrimers as Non-Viral Vectors in Gene-Directed Enzyme Prodrug Therapy. Molecules 2021; 26:5976. [PMID: 34641519 PMCID: PMC8512881 DOI: 10.3390/molecules26195976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/19/2021] [Accepted: 09/29/2021] [Indexed: 01/02/2023] Open
Abstract
Gene-directed enzyme prodrug therapy (GDEPT) has been intensively studied as a promising new strategy of prodrug delivery, with its main advantages being represented by an enhanced efficacy and a reduced off-target toxicity of the active drug. In recent years, numerous therapeutic systems based on GDEPT strategy have entered clinical trials. In order to deliver the desired gene at a specific site of action, this therapeutic approach uses vectors divided in two major categories, viral vectors and non-viral vectors, with the latter being represented by chemical delivery agents. There is considerable interest in the development of non-viral vectors due to their decreased immunogenicity, higher specificity, ease of synthesis and greater flexibility for subsequent modulations. Dendrimers used as delivery vehicles offer many advantages, such as: nanoscale size, precise molecular weight, increased solubility, high load capacity, high bioavailability and low immunogenicity. The aim of the present work was to provide a comprehensive overview of the recent advances regarding the use of dendrimers as non-viral carriers in the GDEPT therapy.
Collapse
Affiliation(s)
| | | | | | - Adina Frum
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (A.A.C.); (C.M.D.); (L.-L.R.); (A.B.); (M.T.); (A.M.J.); (F.G.G.); (A.M.A.)
| | - Claudiu Morgovan
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (A.A.C.); (C.M.D.); (L.-L.R.); (A.B.); (M.T.); (A.M.J.); (F.G.G.); (A.M.A.)
| | | | | | | | | | | |
Collapse
|
57
|
Application of Non-Viral Vectors in Drug Delivery and Gene Therapy. Polymers (Basel) 2021; 13:polym13193307. [PMID: 34641123 PMCID: PMC8512075 DOI: 10.3390/polym13193307] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/15/2021] [Accepted: 09/18/2021] [Indexed: 12/13/2022] Open
Abstract
Vectors and carriers play an indispensable role in gene therapy and drug delivery. Non-viral vectors are widely developed and applied in clinical practice due to their low immunogenicity, good biocompatibility, easy synthesis and modification, and low cost of production. This review summarized a variety of non-viral vectors and carriers including polymers, liposomes, gold nanoparticles, mesoporous silica nanoparticles and carbon nanotubes from the aspects of physicochemical characteristics, synthesis methods, functional modifications, and research applications. Notably, non-viral vectors can enhance the absorption of cargos, prolong the circulation time, improve therapeutic effects, and provide targeted delivery. Additional studies focused on recent innovation of novel synthesis techniques for vector materials. We also elaborated on the problems and future research directions in the development of non-viral vectors, which provided a theoretical basis for their broad applications.
Collapse
|
58
|
Chytła A, Gajdzik-Nowak W, Biernatowska A, Sikorski AF, Czogalla A. High-Level Expression of Palmitoylated MPP1 Recombinant Protein in Mammalian Cells. MEMBRANES 2021; 11:membranes11090715. [PMID: 34564532 PMCID: PMC8470630 DOI: 10.3390/membranes11090715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 11/24/2022]
Abstract
Our recent studies have pointed to an important role of the MAGUK family member, MPP1, as a crucial molecule interacting with flotillins and involved in the lateral organization of the erythroid plasma membrane. The palmitoylation of MPP1 seems to be an important element in this process; however, studies on the direct effect of palmitoylation on protein–protein or protein–membrane interactions in vitro are still challenging due to the difficulties in obtaining functional post-translationally modified recombinant proteins and the lack of comprehensive protocols for the purification of palmitoylated proteins. In this work, we present an optimized approach for the high-yield overexpression and purification of palmitoylated recombinant MPP1 protein in mammalian HEK-293F cells. The presented approach facilitates further studies on the molecular mechanism of lateral membrane organization and the functional impact of the palmitoylation of MPP1, which could also be carried out for other palmitoylated proteins.
Collapse
Affiliation(s)
- Agnieszka Chytła
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.C.); (W.G.-N.)
| | - Weronika Gajdzik-Nowak
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.C.); (W.G.-N.)
| | - Agnieszka Biernatowska
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.C.); (W.G.-N.)
- Correspondence: (A.B.); (A.C.); Tel.: +48-7-1375-417 (A.B.); +48-7-1375-6356 (A.C.)
| | - Aleksander F. Sikorski
- Research and Development Center, Regional Specialist Hospital, Kamieńskiego 73a, 51-154 Wroclaw, Poland;
| | - Aleksander Czogalla
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.C.); (W.G.-N.)
- Correspondence: (A.B.); (A.C.); Tel.: +48-7-1375-417 (A.B.); +48-7-1375-6356 (A.C.)
| |
Collapse
|
59
|
Zohri M, Arefian E, Akbari Javar H, Gazori T, Aghaee-Bakhtiari SH, Taheri M, Fatahi Y, Azadi A, Khoshayand MR, Ghahremani MH. Potential of chitosan/alginate nanoparticles as a non-viral vector for gene delivery: Formulation and optimization using D-optimal design. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112262. [PMID: 34474821 DOI: 10.1016/j.msec.2021.112262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/03/2021] [Accepted: 06/11/2021] [Indexed: 12/24/2022]
Abstract
Chitosan/alginate (Chi/Alg) nanoparticles as a non-viral vector for the Smad4 encoding plasmid were optimized utilizing D-optimal design based on the nanoparticles/plasmid ratio, Chi/Alg MW, and preparation method type. Following the optimization and validation of the best formula, morphology studies and FTIR measurements were performed to evaluate the optimized Chi/Alg/S NPs. Toxicity (MTT assay) and transfection studies were performed for the best formula in comparison with Lipofectamine 2000, and Polyethyleneimine (PEI) and evaluated using Green Fluorescence Protein (GFP) assay, Flow cytometry, and RT-PCR. The model predicted a particle size of 111 nm, loading efficacy (LE) of 43%, cumulative release (CMR) of 39%, the ζ-potential of +50 mV, and PDI of 0.13. The predicted point condition was as follows: NP ratio = 13, Chi/Alg MW ratio = 2.35, and preparation method type = 1. Microscopic findings revealed that the shape of nanoparticles was spherical. The Chi/Alg/S nanoparticles showed no toxicity and transfection efficacy of 29.9% was observed in comparison with Lipofectamine (35.5%) and PEI (30.9%).
Collapse
Affiliation(s)
- Maryam Zohri
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Hamid Akbari Javar
- Departments of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Taraneh Gazori
- Trita Nanomedicine Research Center (TNRC), Trita Third Millennium Pharmaceuticals, 1917733831 Tehran, Iran
| | - Seyed Hamid Aghaee-Bakhtiari
- Bioinformatics Research Group, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yousef Fatahi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Azadi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Khoshayand
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
60
|
Niosomes-based gene delivery systems for effective transfection of human mesenchymal stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112307. [PMID: 34474858 DOI: 10.1016/j.msec.2021.112307] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/07/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023]
Abstract
Gene transfer to mesenchymal stem cells (MSCs) has arisen as a powerful approach to increase the therapeutic potential of this effective cell population. Over recent years, niosomes have emerged as self-assembled carriers with promising performance for gene delivery. The aim of our work was to develop effective niosomes-based DNA delivery platforms for targeting MSCs. Niosomes based on 1,2-di-O-octadecenyl-3-trimethylammonium propane (DOTMA; 0, 7 or 15%) as cationic lipid, cholesterol as helper lipid, and polysorbate 60 as non-ionic surfactant, were prepared using a reverse phase evaporation technique. Niosomes dispersions (filtered or not) and their corresponding nioplexes with a lacZ plasmid were characterized in terms of size, charge, protection, and complexation abilities. DOTMA concentration had a large influence on the physicochemical properties of resulting nioplexes. Transfection efficiency and cytotoxic profiles were assessed in two immortalized cell lines of MSCs. Niosomes formulated with 15% DOTMA provided the highest values of β-galactosidase activity, being similar to those achieved with Lipofectamine®, but showed less cytotoxicity. Filtration of niosomes dispersions before adding to the cells resulted in a loss of their biological activities. Storage of niosomes formulations (for 30 days at room temperature) caused minor modification of their physicochemical properties but also attenuated the transfection capability of the nioplexes. Differently, addition of the lysosomotropic agent sucrose into the culture medium during transfection or to the formulation itself improved the transfection performance of non-filtered niosomes. Indeed, steam heat-sterilized niosomes prepared in sucrose medium demonstrated transfection capability.
Collapse
|
61
|
Sattari S, Adeli M, Beyranvand S, Nemati M. Functionalized Graphene Platforms for Anticancer Drug Delivery. Int J Nanomedicine 2021; 16:5955-5980. [PMID: 34511900 PMCID: PMC8416335 DOI: 10.2147/ijn.s249712] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/17/2021] [Indexed: 12/24/2022] Open
Abstract
Two-dimensional nanomaterials are emerging as promising candidates for a wide range of biomedical applications including tissue engineering, biosensing, pathogen incapacitation, wound healing, and gene and drug delivery. Graphene, due to its high surface area, photothermal property, high loading capacity, and efficient cellular uptake, is at the forefront of these materials and plays a key role in this multidisciplinary research field. Poor water dispersibility and low functionality of graphene, however, hamper its hybridization into new nanostructures for future nanomedicine. Functionalization of graphene, either by covalent or non-covalent methods, is the most useful strategy to improve its dispersion in water and functionality as well as processability into new materials and devices. In this review, recent advances in functionalization of graphene derivatives by different (macro)molecules for future biomedical applications are reported and explained. In particular, hydrophilic functionalization of graphene and graphene oxide (GO) to improve their water dispersibility and physicochemical properties is discussed. We have focused on the anticancer drug delivery of polyfunctional graphene sheets.
Collapse
Affiliation(s)
- Shabnam Sattari
- Department of Chemistry, Faculty of Science, Lorestan University, Khorramabad, Iran
| | - Mohsen Adeli
- Department of Chemistry, Faculty of Science, Lorestan University, Khorramabad, Iran
| | - Siamak Beyranvand
- Department of Chemistry, Faculty of Science, Lorestan University, Khorramabad, Iran
| | - Mohammad Nemati
- Department of Chemistry, Faculty of Science, Lorestan University, Khorramabad, Iran
| |
Collapse
|
62
|
Functionalized PDA/DEX-PEI@HA nanoparticles combined with sleeping-beauty transposons for multistage targeted delivery of CRISPR/Cas9 gene. Biomed Pharmacother 2021; 142:112061. [PMID: 34449313 DOI: 10.1016/j.biopha.2021.112061] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/10/2021] [Accepted: 08/16/2021] [Indexed: 11/20/2022] Open
Abstract
CRISPR/Cas9 system has been used as the most powerful gene editing tool for precision medicine and advanced gene therapy. However, its wide applications are limited by the poor biosafety of lentivirus delivery vectors though with high-efficiency transduction. To construct a safer vector and promote genome integration, the CRISPR/Cas9 gene is cloned into a plasmid-based non-viral safe vector Sleeping-Beauty (SB) transposon in this study to obtain pT2SpCas9. Meanwhile, PDA/DEX-PEI@HA (PDPH) nanoparticles are constructed to facilitate the precise CRISPR/Cas9 targeting delivery, by using polydopamine (PDA) as the carrier, hyaluronic acid (HA) as the cell-targeting ligand and dexamethasone (DEX) as the nuclear localization signal (NLS). The results showed that PDPH could deliver pDNA efficiently into the cell and further into the nucleus. The transfection efficiency of PDPH is much higher than that of NPs without HA and DEX. Remarkably, the cytotoxicity of PDPH is negligible in comparison to PEI25k and PEI10k. Western blots showed that after the transfection of PDPH/pT2SpCas9-Nanog/SB11, Nanog protein in HeLa cells is knocked out, and the proliferation and migration abilities of tumor cells are significantly decreased. This study demonstrates that PDA/DEX-PEI25k@HA/pT2SpCas9 (PDPH25 K/pT2SpCas9) has the great potential as a non-viral gene vector for CRISPR/Cas9 delivery and clinical medication.
Collapse
|
63
|
Lin MHC, Lai PS, Chang LC, Huang WC, Lee MH, Chen KT, Chung CY, Yang JT. Characterization and Optimization of Chitosan-Coated Polybutylcyanoacrylate Nanoparticles for the Transfection-Guided Neural Differentiation of Mouse Induced Pluripotent Stem Cells. Int J Mol Sci 2021; 22:8741. [PMID: 34445447 PMCID: PMC8395893 DOI: 10.3390/ijms22168741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/07/2021] [Accepted: 08/11/2021] [Indexed: 01/04/2023] Open
Abstract
Gene transfection is a valuable tool for analyzing gene regulation and function, and providing an avenue for the genetic engineering of cells for therapeutic purposes. Though efficient, the potential concerns over viral vectors for gene transfection has led to research in non-viral alternatives. Cationic polyplexes such as those synthesized from chitosan offer distinct advantages such as enhanced polyplex stability, cellular uptake, endo-lysosomal escape, and release, but are limited by the poor solubility and viscosity of chitosan. In this study, the easily synthesized biocompatible and biodegradable polymeric polysorbate 80 polybutylcyanoacrylate nanoparticles (PS80 PBCA NP) are utilized as the backbone for surface modification with chitosan, in order to address the synthetic issues faced when using chitosan alone as a carrier. Plasmid DNA (pDNA) containing the brain-derived neurotrophic factor (BDNF) gene coupled to a hypoxia-responsive element and the cytomegalovirus promotor gene was selected as the genetic cargo for the in vitro transfection-guided neural-lineage specification of mouse induced pluripotent stem cells (iPSCs), which were assessed by immunofluorescence staining. The chitosan-coated PS80 PBCA NP/BDNF pDNA polyplex measured 163.8 ± 1.8 nm and zeta potential measured -34.8 ± 1.8 mV with 0.01% (w/v) high molecular weight chitosan (HMWC); the pDNA loading efficiency reached 90% at a nanoparticle to pDNA weight ratio of 15, which also corresponded to enhanced polyplex stability on the DNA stability assay. The HMWC-PS80 PBCA NP/BDNF pDNA polyplex was non-toxic to mouse iPSCs for up to 80 μg/mL (weight ratio = 40) and enhanced the expression of BDNF when compared with PS80 PBCA NP/BDNF pDNA polyplex. Evidence for neural-lineage specification of mouse iPSCs was observed by an increased expression of nestin, neurofilament heavy polypeptide, and beta III tubulin, and the effects appeared superior when transfection was performed with the chitosan-coated formulation. This study illustrates the versatility of the PS80 PBCA NP and that surface decoration with chitosan enabled this delivery platform to be used for the transfection-guided differentiation of mouse iPSCs.
Collapse
Affiliation(s)
- Martin Hsiu-Chu Lin
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan;
- PhD Programme of Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Ping-Shan Lai
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan;
| | - Li-Ching Chang
- Department of Dentistry, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan;
- Department of Nursing, Chang Gung University of Science and Technology, Chia-Yi 61363, Taiwan
| | - Wei-Chao Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
| | - Ming-Hsueh Lee
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
| | - Kuo-Tai Chen
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
| | - Chiu-Yen Chung
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
| | - Jen-Tsung Yang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
- College of Medicine, School of Traditional Chinese Medicine, Chang Gung University, Tao-Yuan 33302, Taiwan
| |
Collapse
|
64
|
Pires IS, Hammond PT, Irvine DJ. Engineering Strategies for Immunomodulatory Cytokine Therapies - Challenges and Clinical Progress. ADVANCED THERAPEUTICS 2021; 4:2100035. [PMID: 34734110 PMCID: PMC8562465 DOI: 10.1002/adtp.202100035] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Indexed: 12/15/2022]
Abstract
Cytokines are immunoregulatory proteins involved in many pathological states with promising potential as therapeutic agents. A diverse array of cytokines have been studied in preclinical disease models since the 1950s, some of which became successful biopharmaceutical products with the advancement of recombinant protein technology in the 1980s. However, following these early approvals, clinical translation of these natural immune signaling molecules has been limited due to their pleiotropic action in many cell types, and the fact that they have evolved to act primarily locally in tissues. These characteristics, combined with poor pharmacokinetics, have hindered the delivery of cytokines via systemic administration routes due to dose-limiting toxicities. However, given their clinical potential and recent clinical successes in cancer immunotherapy, cytokines continue to be extensively pursued in preclinical and clinical studies, and a range of molecular and formulation engineering strategies are being applied to reduce treatment toxicity while maintaining or enhancing therapeutic efficacy. This review provides a brief background on the characteristics of cytokines and their history as clinical therapeutics, followed by a deeper discussion on the engineering strategies developed for cytokine therapies with a focus on the translational relevance of these approaches.
Collapse
Affiliation(s)
- Ivan S Pires
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
65
|
Dielectrophoretic Manipulation of Cell Transfection Efficiency during Electroporation Using a Center Needle Electrode. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11157015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Long duration electric pulses are frequently used to facilitate DNA electrotransfer into cells and tissues, while electroporation pulses can be combined with electrophoresis to maximize the transfection efficiency. In this work, we present the dielectrophoresis (DEP)-assisted methodology for electrotransfer of plasmid DNA (3.5 kbp pmaxGFP) into mammalian cells (CHO-K1). A prototype of an electroporation cuvette with center needle electrode for DEP-assisted transfection is presented resulting in a 1.4-fold of transfection efficiency increase compared to the electroporation-only procedure (1.4 kV/cm × 100 µs × 8). The efficiency of transfection has been compared between three DEP frequencies of 1, 100, and 1 MHz. Lastly, the effects of exposure time (1, 3, and 5 min) during the DEP application step have been determined. It is concluded that the proposed methodology and exposure setup allow a significant improvement of transfection efficiency and could be used as an alternative to the currently popular electrotransfection techniques.
Collapse
|
66
|
Saeed RM, Abdullah M, Ahram M, Taha MO. Novel Ellipsoid Chitosan-Phthalate Lecithin Nanoparticles for siRNA Delivery. Front Bioeng Biotechnol 2021; 9:695371. [PMID: 34395401 PMCID: PMC8355739 DOI: 10.3389/fbioe.2021.695371] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/15/2021] [Indexed: 12/11/2022] Open
Abstract
Small interfering RNA (siRNA) has received increased interest as a gene therapeutic agent. However, instability and lack of safe, affordable, and effective carrier systems limit siRNA's widespread clinical use. To tackle this issue, synthetic vectors such as liposomes and polymeric nanoparticles have recently been extensively investigated. In this study, we exploited the advantages of reduced cytotoxicity and enhanced cellular penetration of chitosan-phthalate (CSP) together with the merits of lecithin (LC)-based nanoparticles (NPs) to create novel, ellipsoid, non-cytotoxic, tripolyphosphate (TPP)-crosslinked NPs capable of delivering siRNA efficiently. The resulting NPs were characterized by dynamic light scattering (DLS) and transmission electron microscopy (TEM), and were found to be ellipsoid in the shape of ca. 180 nm in size, exhibiting novel double-layer shells, with excellent stability at physiological pH and in serum solutions. MTT assay and confocal fluorescence microscopy showed that CSP-LC-TPP NPs are non-cytotoxic and efficiently penetrate cancer cells in vitro. They achieved 44% silencing against SLUG protein in MDA-MB-453 cancer cells and were significantly superior to a commercial liposome-based transfection agent that achieved only 30% silencing under comparable conditions. Moreover, the NPs protected their siRNA cargos in 50% serum and from being displaced by variable concentrations of heparin. In fact, CSP-LC-TPP NPs achieved 26% transfection efficiency in serum containing cell culture media. Real-time wide-field fluorescence microscopy showed siRNA-loaded CSP-LC-TPP NPs to successfully release their cargo intracellularly. We found that the amphoteric nature of chitosan-phthalate polymer promotes the endosomal escape of siRNA and improves the silencing efficiency.
Collapse
Affiliation(s)
- Ramzi Mukred Saeed
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Mohammed Abdullah
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| | - Mamoun Ahram
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| | - Mutasem Omar Taha
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Jordan, Amman, Jordan
| |
Collapse
|
67
|
Forced Suppression of let-7a-5p in Mouse Blastocysts Improves Implantation Rate. Reprod Sci 2021; 29:1730-1737. [PMID: 34254278 DOI: 10.1007/s43032-021-00659-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/10/2021] [Indexed: 10/20/2022]
Abstract
Embryo implantation requires appropriate communication between the blastocyst and endometrium. Recurrent implantation failure is an essential component of assisted reproductive technology. Also, miRNA-mediated gene expression impacts the implantation process, and the downregulation of some miRs, such as mmu-let-7a, improves this process. In the present study, we evaluated the effect of let-7a forced suppression on the mouse implantation rate. In total, 100 adult female mice and 10 adult male mice were included (Strain CD-1). We analysed the expression of let-7a and its potential mRNAs targets (Igf1, Il1a, Itgb3 and Tgfb1) in control, sham and antagomir-treated blastocysts using quantitative reverse transcription PCR (qRT-PCR). The control and treated blastocysts were transferred to the 20 pseudopregnant mice so that the effect of let-7a suppression on the rate of implantation could be determined. The expression level of let-7a in the treatment group was significantly downregulated (P=0.001) In contrast, no significant expression changes were observed for let-7a or mRNAs targets when the sham and control groups were compared (P>0.05). In comparison to the controls, the antagomir-treated group exhibited significantly upregulated expression levels of Igf1 (0.0167), Itgb3 (0.045) and Tgfb1 (0.0115). Additionally, the implantation rate was significantly higher in the treatment group (78%) than the control group (61%) (P=0.0098). We found that forced suppression of mmu-let-7a-5p through successful transfection of Anti-miR leads to upregulation of downstream genes, Igf1, Itgb3 and Tgfb1, which directly involved in the trophoblast-endometrium attachment and improve the implantation rate.
Collapse
|
68
|
Mezghrani B, Ali LMA, Richeter S, Durand JO, Hesemann P, Bettache N. Periodic Mesoporous Ionosilica Nanoparticles for Green Light Photodynamic Therapy and Photochemical Internalization of siRNA. ACS APPLIED MATERIALS & INTERFACES 2021; 13:29325-29339. [PMID: 34138540 DOI: 10.1021/acsami.1c05848] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
We report periodic mesoporous ionosilica nanoparticles (PMINPs) as versatile nano-objects for imaging, photodynamic therapy (PDT), and efficient adsorption and delivery of small interfering RNA (siRNA) into breast cancer cells. In order to endow these nanoparticles with PDT and siRNA photochemical internalization (PCI) properties, a porphyrin derivative was integrated into the ionosilica framework. For this purpose, we synthesized PMINPs via hydrolysis-cocondensation procedures from oligosilylated ammonium and porphyrin precursors. The formation of these nano-objects was proved by transmission electron microscopy. The formed nanoparticles were then thoroughly characterized via solid-state NMR, nitrogen sorption, dynamic light scattering, and UV-vis and fluorescence spectroscopies. Our results indicate the formation of highly porous nanorods with a length of 108 ± 9 nm and a width of 54 ± 4 nm. A significant PDT effect of type I mechanism (95 ± 2.8% of cell death) was observed upon green light irradiation in nanoparticle-treated breast cancer cells, while the blue light irradiation caused a significant phototoxic effect in non-treated cells. Furthermore, PMINPs formed stable complexes with siRNA (up to 24 h), which were efficiently internalized into the cells after 4 h of incubation mostly with the energy-dependent endocytosis process. The PCI effect was obvious with green light irradiation and successfully led to 83 ± 1.1% silencing of the luciferase gene in luciferase-expressing breast cancer cells, while no gene silencing effect was observed with blue light irradiation. The present work highlights the high potential of porphyrin-doped PMINPs as multifunctional nanocarriers for nucleic acids, such as siRNA, with a triple ability to perform imaging, PDT, and PCI.
Collapse
Affiliation(s)
- Braham Mezghrani
- IBMM, Univ. Montpellier, CNRS, ENSCM; Avenue Charles Flahault, CEDEX 05, 34093 Montpellier, France
- ICGM, Univ Montpellier-CNRS-ENSCM, 34090 Montpellier, France
| | - Lamiaa M A Ali
- IBMM, Univ. Montpellier, CNRS, ENSCM; Avenue Charles Flahault, CEDEX 05, 34093 Montpellier, France
- Department of Biochemistry, Medical Research Institute, University of Alexandria, Alexandria 21561, Egypt
| | | | | | - Peter Hesemann
- ICGM, Univ Montpellier-CNRS-ENSCM, 34090 Montpellier, France
| | - Nadir Bettache
- IBMM, Univ. Montpellier, CNRS, ENSCM; Avenue Charles Flahault, CEDEX 05, 34093 Montpellier, France
| |
Collapse
|
69
|
Stueber DD, Villanova J, Aponte I, Xiao Z, Colvin VL. Magnetic Nanoparticles in Biology and Medicine: Past, Present, and Future Trends. Pharmaceutics 2021; 13:943. [PMID: 34202604 PMCID: PMC8309177 DOI: 10.3390/pharmaceutics13070943] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/27/2021] [Accepted: 06/16/2021] [Indexed: 12/23/2022] Open
Abstract
The use of magnetism in medicine has changed dramatically since its first application by the ancient Greeks in 624 BC. Now, by leveraging magnetic nanoparticles, investigators have developed a range of modern applications that use external magnetic fields to manipulate biological systems. Drug delivery systems that incorporate these particles can target therapeutics to specific tissues without the need for biological or chemical cues. Once precisely located within an organism, magnetic nanoparticles can be heated by oscillating magnetic fields, which results in localized inductive heating that can be used for thermal ablation or more subtle cellular manipulation. Biological imaging can also be improved using magnetic nanoparticles as contrast agents; several types of iron oxide nanoparticles are US Food and Drug Administration (FDA)-approved for use in magnetic resonance imaging (MRI) as contrast agents that can improve image resolution and information content. New imaging modalities, such as magnetic particle imaging (MPI), directly detect magnetic nanoparticles within organisms, allowing for background-free imaging of magnetic particle transport and collection. "Lab-on-a-chip" technology benefits from the increased control that magnetic nanoparticles provide over separation, leading to improved cellular separation. Magnetic separation is also becoming important in next-generation immunoassays, in which particles are used to both increase sensitivity and enable multiple analyte detection. More recently, the ability to manipulate material motion with external fields has been applied in magnetically actuated soft robotics that are designed for biomedical interventions. In this review article, the origins of these various areas are introduced, followed by a discussion of current clinical applications, as well as emerging trends in the study and application of these materials.
Collapse
Affiliation(s)
- Deanna D. Stueber
- Center for Biomedical Engineering, School of Engineering, Brown University, 171 Meeting Street, Providence, RI 02912, USA; (D.D.S.); (J.V.); (I.A.)
| | - Jake Villanova
- Center for Biomedical Engineering, School of Engineering, Brown University, 171 Meeting Street, Providence, RI 02912, USA; (D.D.S.); (J.V.); (I.A.)
- Department of Chemistry, Brown University, 324 Brook Street, Providence, RI 02912, USA;
| | - Itzel Aponte
- Center for Biomedical Engineering, School of Engineering, Brown University, 171 Meeting Street, Providence, RI 02912, USA; (D.D.S.); (J.V.); (I.A.)
| | - Zhen Xiao
- Department of Chemistry, Brown University, 324 Brook Street, Providence, RI 02912, USA;
| | - Vicki L. Colvin
- Center for Biomedical Engineering, School of Engineering, Brown University, 171 Meeting Street, Providence, RI 02912, USA; (D.D.S.); (J.V.); (I.A.)
- Department of Chemistry, Brown University, 324 Brook Street, Providence, RI 02912, USA;
| |
Collapse
|
70
|
Ding Y, Wang C, Sun Z, Wu Y, You W, Mao Z, Wang W. Mesenchymal Stem Cells Engineered by Nonviral Vectors: A Powerful Tool in Cancer Gene Therapy. Pharmaceutics 2021; 13:pharmaceutics13060913. [PMID: 34205513 PMCID: PMC8235299 DOI: 10.3390/pharmaceutics13060913] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/09/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022] Open
Abstract
Due to their "tumor homing" and "immune privilege" characteristics, the use of mesenchymal stem cells (MSCs) has been proposed as a novel tool against cancer. MSCs are genetically engineered in vitro and then utilized to deliver tumoricidal agents, including prodrugs and bioactive molecules, to tumors. The genetic modification of MSCs can be achieved by various vectors, and in most cases viral vectors are used; however, viruses may be associated with carcinogenesis and immunogenicity, restricting their clinical translational potential. As such, nonviral vectors have emerged as a potential solution to address these limitations and have gradually attracted increasing attention. In this review, we briefly revisit the current knowledge about MSC-based cancer gene therapy. Then, we summarize the advantages and challenges of nonviral vectors for MSC transfection. Finally, we discuss recent advances in the development of new nonviral vectors, which have provided promising strategies to overcome obstacles in the gene modulation of MSCs.
Collapse
Affiliation(s)
- Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; (Y.D.); (C.W.); (Z.S.); (Y.W.); (W.Y.)
- Key Laboratory, Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou 310009, China
- Research Center, Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou 310009, China
- Clinical Medicine Innovation Center, Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou 310009, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, China
- Cancer Center, Zhejiang University, Hangzhou 310009, China
| | - Chenyang Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; (Y.D.); (C.W.); (Z.S.); (Y.W.); (W.Y.)
- Key Laboratory, Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou 310009, China
- Research Center, Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou 310009, China
- Clinical Medicine Innovation Center, Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou 310009, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, China
- Cancer Center, Zhejiang University, Hangzhou 310009, China
| | - Zhongquan Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; (Y.D.); (C.W.); (Z.S.); (Y.W.); (W.Y.)
- Key Laboratory, Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou 310009, China
- Research Center, Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou 310009, China
- Clinical Medicine Innovation Center, Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou 310009, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, China
- Cancer Center, Zhejiang University, Hangzhou 310009, China
| | - Yingsheng Wu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; (Y.D.); (C.W.); (Z.S.); (Y.W.); (W.Y.)
- Key Laboratory, Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou 310009, China
- Research Center, Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou 310009, China
- Clinical Medicine Innovation Center, Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou 310009, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, China
- Cancer Center, Zhejiang University, Hangzhou 310009, China
| | - Wanlu You
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; (Y.D.); (C.W.); (Z.S.); (Y.W.); (W.Y.)
- Key Laboratory, Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou 310009, China
- Research Center, Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou 310009, China
- Clinical Medicine Innovation Center, Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou 310009, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, China
- Cancer Center, Zhejiang University, Hangzhou 310009, China
| | - Zhengwei Mao
- Key Laboratory, Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou 310009, China
- MOE Key Laboratory, Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
- Correspondence: (Z.M.); (W.W.); Tel.: +86-15168215834 (Z.M.); +86-0571-87783820 (W.W.)
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China; (Y.D.); (C.W.); (Z.S.); (Y.W.); (W.Y.)
- Key Laboratory, Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou 310009, China
- Research Center, Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou 310009, China
- Clinical Medicine Innovation Center, Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou 310009, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310009, China
- Cancer Center, Zhejiang University, Hangzhou 310009, China
- Correspondence: (Z.M.); (W.W.); Tel.: +86-15168215834 (Z.M.); +86-0571-87783820 (W.W.)
| |
Collapse
|
71
|
Hörner M, Jerez-Longres C, Hudek A, Hook S, Yousefi OS, Schamel WWA, Hörner C, Zurbriggen MD, Ye H, Wagner HJ, Weber W. Spatiotemporally confined red light-controlled gene delivery at single-cell resolution using adeno-associated viral vectors. SCIENCE ADVANCES 2021; 7:7/25/eabf0797. [PMID: 34134986 PMCID: PMC8208708 DOI: 10.1126/sciadv.abf0797] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 05/04/2021] [Indexed: 05/15/2023]
Abstract
Methodologies for the controlled delivery of genetic information into target cells are of utmost importance for genetic engineering in both fundamental and applied research. However, available methods for efficient gene transfer into user-selected or even single cells suffer from low throughput, the need for complicated equipment, high invasiveness, or side effects by off-target viral uptake. Here, we engineer an adeno-associated viral (AAV) vector system that transfers genetic information into native target cells upon illumination with cell-compatible red light. This OptoAAV system allows adjustable and spatially resolved gene transfer down to single-cell resolution and is compatible with different cell lines and primary cells. Moreover, the sequential application of multiple OptoAAVs enables spatially resolved transduction with different transgenes. The approach presented is likely extendable to other classes of viral vectors and is expected to foster advances in basic and applied genetic research.
Collapse
Affiliation(s)
- Maximilian Hörner
- Faculty of Biology, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Carolina Jerez-Longres
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Anna Hudek
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Sebastian Hook
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - O Sascha Yousefi
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - Wolfgang W A Schamel
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - Cindy Hörner
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, Langen, Germany
| | - Matias D Zurbriggen
- Institute of Synthetic Biology and CEPLAS, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Haifeng Ye
- Synthetic Biology and Biomedical Engineering Laboratory, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Hanna J Wagner
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Wilfried Weber
- Faculty of Biology, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| |
Collapse
|
72
|
Arima H. Twenty Years of Research on Cyclodextrin Conjugates with PAMAM Dendrimers. Pharmaceutics 2021; 13:697. [PMID: 34064866 PMCID: PMC8151880 DOI: 10.3390/pharmaceutics13050697] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
Recently, the number of gene and oligonucleotide drugs are increasing. Of various drug delivery systems (DDSs) for gene and oligonucleotide drugs, few examples of the clinical application of polymer as drug carriers are known, despite development of the novel polymers has been progressing. Cyclodextrin (CD) conjugates with starburst polyamidoamine (PAMAM) dendrimer (CDEs), as a new type of polymer-based carriers, were first published in 2001. After that, galactose-, lactose-, mannose-, fucose-, folate-, and polyethyleneglycol (PEG)-appended CDEs have been prepared for passive and active targeting for gene, oligonucleotide, and low-molecular-weight drugs. PEG-appended CDE formed polypsuedorotaxanes with α-CD and γ-CD, which are useful for a sustained release system of gene and oligonucleotide drugs. Interestingly, CDEs were found to have anti-inflammatory effects and anti-amyloid effects themselves, which have potential as active pharmaceutical ingredients. Most recently, CDE is reported to be a useful Cas9-RNA ribonucleoproteins (Cas9 RNP) carrier that induces genome editing in the neuron and brain. In this review, the history and progression of CDEs are overviewed.
Collapse
Affiliation(s)
- Hidetoshi Arima
- School of Pharmacy, Daiichi University of Pharmacy, Fukuoka 815-8511, Japan
| |
Collapse
|
73
|
Bono N, Coloma Smith B, Moreschi F, Redaelli A, Gautieri A, Candiani G. In silico prediction of the in vitro behavior of polymeric gene delivery vectors. NANOSCALE 2021; 13:8333-8342. [PMID: 33900339 DOI: 10.1039/d0nr09052b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Non-viral gene delivery vectors have increasingly come under the spotlight, but their performaces are still far from being satisfactory. Therefore, there is an urgent need for forecasting tools and screening methods to enable the development of ever more effective transfectants. Here, coarse-grained (CG) models of gold standard transfectant poly(ethylene imine)s (PEIs) have been profitably used to investigate and highlight the effect of experimentally-relevant parameters, namely molecular weight (2 vs. 10 kDa) and topologies (linear vs. branched), protonation state, and ammine-to-phosphate ratios (N/Ps), on the complexation and the gene silencing efficiency of siRNA molecules. The results from the in vitro screening of cationic polymers and conditions were used to validate the in silico platform that we developed, such that the hits which came out of the CG models were of high practical relevance. We show that our in silico platform enables to foresee the most suitable conditions for the complexation of relevant siRNA-polycation assemblies, thereby providing a reliable predictive tool to test bench transfectants in silico, and foster the design and development of gene delivery vectors.
Collapse
Affiliation(s)
- Nina Bono
- GenT LΛB, Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, 20131 Milan, Italy.
| | | | | | | | | | | |
Collapse
|
74
|
Kavanagh H, Dunne S, Martin DS, McFadden E, Gallagher L, Schwaber J, Leonard S, O'Dea S. A novel non-viral delivery method that enables efficient engineering of primary human T cells for ex vivo cell therapy applications. Cytotherapy 2021; 23:852-860. [PMID: 33941482 PMCID: PMC8386197 DOI: 10.1016/j.jcyt.2021.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/02/2021] [Accepted: 03/12/2021] [Indexed: 11/28/2022]
Abstract
Background aims Next-generation immune cell therapy products will require complex modifications using engineering technologies that can maintain high levels of cell functionality. Non-viral engineering methods have the potential to address limitations associated with viral vectors. However, while electroporation is the most widely used non-viral modality, concerns about its effects on cell functionality have led to the exploration of alternative approaches. Here the authors have examined the suitability of the Solupore non-viral delivery system for engineering primary human T cells for cell therapy applications. Methods The Solupore system was used to deliver messenger RNA (mRNA) and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) guide RNA ribonucleoprotein (RNP) cargos to T cells, and efficiency was measured by flow cytometry. Cell perturbation was assessed by immune gene expression profiling, including an electroporation comparator. In vitro and in vivo cytotoxicity of chimeric antigen receptor (CAR) T cells generated using the Solupore system was evaluated using a real-time cellular impedance assay and a Raji-luciferase mouse tumor model, respectively. Results Efficient transfection was demonstrated through delivery of mRNA and CRISPR CAS9 RNP cargos individually, simultaneously and sequentially using the Solupore system while consistently maintaining high levels of cell viability. Gene expression profiling revealed minimal alteration in immune gene expression, demonstrating the low level of perturbation experienced by the cells during this transfection process. By contrast, electroporation resulted in substantial changes in immune gene expression in T cells. CAR T cells generated using the Solupore system exhibited efficient cytotoxicity against target cancer cells in vitro and in vivo. Conclusions The Solupore system is a non-viral means of simply, rapidly and efficiently delivering cargos to primary human immune cells with retention of high cell viability and functionality.
Collapse
|
75
|
Sizikov AA, Kharlamova MV, Nikitin MP, Nikitin PI, Kolychev EL. Nonviral Locally Injected Magnetic Vectors for In Vivo Gene Delivery: A Review of Studies on Magnetofection. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1078. [PMID: 33922066 PMCID: PMC8143545 DOI: 10.3390/nano11051078] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 12/20/2022]
Abstract
Magnetic nanoparticles have been widely used in nanobiomedicine for diagnostics and the treatment of diseases, and as carriers for various drugs. The unique magnetic properties of "magnetic" drugs allow their delivery in a targeted tumor or tissue upon application of a magnetic field. The approach of combining magnetic drug targeting and gene delivery is called magnetofection, and it is very promising. This method is simple and efficient for the delivery of genetic material to cells using magnetic nanoparticles controlled by an external magnetic field. However, magnetofection in vivo has been studied insufficiently both for local and systemic routes of magnetic vector injection, and the relevant data available in the literature are often merely descriptive and contradictory. In this review, we collected and systematized the data on the efficiency of the local injections of magnetic nanoparticles that carry genetic information upon application of external magnetic fields. We also investigated the efficiency of magnetofection in vivo, depending on the structure and coverage of magnetic vectors. The perspectives of the development of the method were also considered.
Collapse
Affiliation(s)
- Artem A. Sizikov
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.A.S.); (M.V.K.); (M.P.N.)
| | - Marianna V. Kharlamova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.A.S.); (M.V.K.); (M.P.N.)
| | - Maxim P. Nikitin
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.A.S.); (M.V.K.); (M.P.N.)
- Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Petr I. Nikitin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 117942 Moscow, Russia
| | - Eugene L. Kolychev
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.A.S.); (M.V.K.); (M.P.N.)
| |
Collapse
|
76
|
Aránega AE, Lozano-Velasco E, Rodriguez-Outeiriño L, Ramírez de Acuña F, Franco D, Hernández-Torres F. MiRNAs and Muscle Regeneration: Therapeutic Targets in Duchenne Muscular Dystrophy. Int J Mol Sci 2021; 22:ijms22084236. [PMID: 33921834 PMCID: PMC8072594 DOI: 10.3390/ijms22084236] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/15/2021] [Indexed: 12/17/2022] Open
Abstract
microRNAs (miRNAs) are small non-coding RNAs required for the post-transcriptional control of gene expression. MicroRNAs play a critical role in modulating muscle regeneration and stem cell behavior. Muscle regeneration is affected in muscular dystrophies, and a critical point for the development of effective strategies for treating muscle disorders is optimizing approaches to target muscle stem cells in order to increase the ability to regenerate lost tissue. Within this framework, miRNAs are emerging as implicated in muscle stem cell response in neuromuscular disorders and new methodologies to regulate the expression of key microRNAs are coming up. In this review, we summarize recent advances highlighting the potential of miRNAs to be used in conjunction with gene replacement therapies, in order to improve muscle regeneration in the context of Duchenne Muscular Dystrophy (DMD).
Collapse
Affiliation(s)
- Amelia Eva Aránega
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
- Correspondence:
| | - Estefanía Lozano-Velasco
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
| | - Lara Rodriguez-Outeiriño
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
| | - Felicitas Ramírez de Acuña
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
| | - Francisco Hernández-Torres
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Avda. de la Investigación 11, 18016 Granada, Spain
| |
Collapse
|
77
|
Singh R, Kumar P. Disaccharide-polyethylenimine organic nanoparticles as non-toxic in vitro gene transporters and their anticancer potential. Bioorg Chem 2021; 112:104918. [PMID: 33932768 DOI: 10.1016/j.bioorg.2021.104918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 12/26/2022]
Abstract
Polyethylenimines (PEIs) have been shown as efficient gene delivery vectors due to their unique properties, however, toxicity as well as non-specific interactions with the tissues/cells because of high charge density have hampered their use in clinical applications. To counter these concerns, here, we have prepared disachharide-PEI organic nanoparticles by mixing PEIs with non-reducing disaccharides, i.e. trehalose (TPONs) and sucrose (SPONs), under mild conditions. The fabricated nanoparticles were complexed with pDNA and size of these complexes was found in the range of ~130-162 nm with zeta potential ~ +8-25 mV. Further evaluation of these nanoparticles revealed that substitution of disaccharides on PEIs successfully augmented cell viability. Transfection efficiency exhibited by these complexes was significantly higher than the unmodified polymer and the standard, Lipofectamine, complexes. Fabrication of organic nanoparticles did not alter the buffering capacity considerably which was found to be instrumental during endosomal escape of the complexes. Among both the series of nanoparticles, trehalose-PEI organic nanoparticles (TPONs) exhibited greater pDNA transportation potential than sucrose-PEI organic nanoparticles (SPONs) which was also established by flow cytometric data, wherein percent cells expressing GFP was higher in case of TP/pDNA complexes as compared to SP/pDNA complexes. Interestingly, TPONs also showed promising anticancer activity on cancer cell lines i.e. Mg63, MCF-7 and HepG2. Overall, the results advocate promising potential of disaccharide-PEI organic nanoparticles as efficient gene delivery agents which can be used effectively in future gene therapy applications along with anti-cancer competence of TPONs.
Collapse
Affiliation(s)
- Reena Singh
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pradeep Kumar
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
78
|
Mdlovu NV, Lin KS, Chen Y, Wu CM. Formulation of magnetic nanocomposites for intracellular delivery of micro-RNA for MYCN inhibition in neuroblastoma. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.126264] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
79
|
Richter F, Mapfumo P, Martin L, Solomun JI, Hausig F, Frietsch JJ, Ernst T, Hoeppener S, Brendel JC, Traeger A. Improved gene delivery to K-562 leukemia cells by lipoic acid modified block copolymer micelles. J Nanobiotechnology 2021; 19:70. [PMID: 33676500 PMCID: PMC7936509 DOI: 10.1186/s12951-021-00801-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/09/2021] [Indexed: 12/23/2022] Open
Abstract
Although there has been substantial progress in the research field of gene delivery, there are some challenges remaining, e.g. there are still cell types such as primary cells and suspension cells (immune cells) known to be difficult to transfect. Cationic polymers have gained increasing attention due to their ability to bind, condense and mask genetic material, being amenable to scale up and highly variable in their composition. In addition, they can be combined with further monomers exhibiting desired biological and chemical properties, such as antioxidative, pH- and redox-responsive or biocompatible features. By introduction of hydrophobic monomers, in particular as block copolymers, cationic micelles can be formed possessing an improved chance of transfection in otherwise challenging cells. In this study, the antioxidant biomolecule lipoic acid, which can also be used as crosslinker, was incorporated into the hydrophobic block of a diblock copolymer, poly{[2-(dimethylamino)ethyl methacrylate]101-b-[n-(butyl methacrylate)124-co-(lipoic acid methacrylate)22]} (P(DMAEMA101-b-[nBMA124-co-LAMA22])), synthesized by RAFT polymerization and assembled into micelles (LAMA-mic). These micelles were investigated regarding their pDNA binding, cytotoxicity mechanisms and transfection efficiency in K-562 and HEK293T cells, the former representing a difficult to transfect, suspension leukemia cell line. The LAMA-mic exhibited low cytotoxicity at applied concentrations but demonstrated superior transfection efficiency in HEK293T and especially K-562 cells. In-depth studies on the transfection mechanism revealed that transfection efficiency in K-562 cells does not depend on the specific oncogenic fusion gene BCR-ABL alone. It is independent of the cellular uptake of polymer-pDNA complexes but correlates with the endosomal escape of the LAMA-mic. A comparison of the transfection efficiency of the LAMA-mic with structurally comparable micelles without lipoic acid showed that lipoic acid is not solely responsible for the superior transfection efficiency of the LAMA-mic. More likely, a synergistic effect of the antioxidative lipoic acid and the micellar architecture was identified. Therefore, the incorporation of lipoic acid into the core of hydrophobic-cationic micelles represents a promising tailor-made transfer strategy, which can potentially be beneficial for other difficult to transfect cell types.
Collapse
Affiliation(s)
- Friederike Richter
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Prosper Mapfumo
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Liam Martin
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Jana I Solomun
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Franziska Hausig
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Jochen J Frietsch
- Klinik für Innere Medizin II, Abteilung Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Thomas Ernst
- Klinik für Innere Medizin II, Abteilung Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Stephanie Hoeppener
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Johannes C Brendel
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany.
| |
Collapse
|
80
|
Valente J, Pereira P, Sousa A, Queiroz J, Sousa F. Effect of Plasmid DNA Size on Chitosan or Polyethyleneimine Polyplexes Formulation. Polymers (Basel) 2021; 13:793. [PMID: 33807586 PMCID: PMC7962013 DOI: 10.3390/polym13050793] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 01/22/2023] Open
Abstract
Gene therapy could be simply defined as a strategy for the introduction of a functional copy of desired genes in patients, to correct some specific mutation and potentially treat the respective disorder. However, this straightforward definition hides very complex processes related to the design and preparation of the therapeutic genes, as well as the development of suitable gene delivery systems. Within non-viral vectors, polymeric nanocarriers have offered an ideal platform to be applied as gene delivery systems. Concerning this, the main goal of the study was to do a systematic evaluation on the formulation of pDNA delivery systems based on the complexation of different sized plasmids with chitosan (CH) or polyethyleneimine (PEI) polymers to search for the best option regarding encapsulation efficiency, surface charge, size, and delivery ability. The cytotoxicity and the transfection efficiency of these systems were accessed and, for the best p53 encoding pDNA nanosystems, the ability to promote protein expression was also evaluated. Overall, it was showed that CH polyplexes are more efficient on transfection when compared with the PEI polyplexes, resulting in higher P53 protein expression. Cells transfected with CH/p53-pDNA polyplexes presented an increase of around 54.2% on P53 expression, while the transfection with the PEI/p53-pDNA polyplexes resulted in a 32% increase.
Collapse
Affiliation(s)
- J.F.A. Valente
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (P.P.); (A.S.); (J.A.Q.)
- CDRsp—Centre Rapid and Sustainable Product Development, Polytechnic Institute of Leiria, 2411-901 Leiria, Portugal
| | - P. Pereira
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (P.P.); (A.S.); (J.A.Q.)
- CEMMPRE, Department of Chemical Engineering, University of Coimbra, Rua Sílvio Lima-Pólo II, 3030-790 Coimbra, Portugal
| | - A. Sousa
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (P.P.); (A.S.); (J.A.Q.)
| | - J.A. Queiroz
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (P.P.); (A.S.); (J.A.Q.)
| | - F. Sousa
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (P.P.); (A.S.); (J.A.Q.)
| |
Collapse
|
81
|
Egorova KS, Posvyatenko AV, Larin SS, Ananikov V. Ionic liquids: prospects for nucleic acid handling and delivery. Nucleic Acids Res 2021; 49:1201-1234. [PMID: 33476366 PMCID: PMC7897475 DOI: 10.1093/nar/gkaa1280] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/18/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022] Open
Abstract
Operations with nucleic acids are among the main means of studying the mechanisms of gene function and developing novel methods of molecular medicine and gene therapy. These endeavours usually imply the necessity of nucleic acid storage and delivery into eukaryotic cells. In spite of diversity of the existing dedicated techniques, all of them have their limitations. Thus, a recent notion of using ionic liquids in manipulations of nucleic acids has been attracting significant attention lately. Due to their unique physicochemical properties, in particular, their micro-structuring impact and tunability, ionic liquids are currently applied as solvents and stabilizing media in chemical synthesis, electrochemistry, biotechnology, and other areas. Here, we review the current knowledge on interactions between nucleic acids and ionic liquids and discuss potential advantages of applying the latter in delivery of the former into eukaryotic cells.
Collapse
Affiliation(s)
- Ksenia S Egorova
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| | - Alexandra V Posvyatenko
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
- Molecular Immunology Laboratory, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Samory Mashela St 1, Moscow 117997, Russia
| | - Sergey S Larin
- Molecular Immunology Laboratory, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Samory Mashela St 1, Moscow 117997, Russia
| | - Valentine P Ananikov
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, Moscow 119991, Russia
| |
Collapse
|
82
|
Blokpoel Ferreras LA, Chan SY, Vazquez Reina S, Dixon JE. Rapidly Transducing and Spatially Localized Magnetofection Using Peptide-Mediated Non-Viral Gene Delivery Based on Iron Oxide Nanoparticles. ACS APPLIED NANO MATERIALS 2021; 4:167-181. [PMID: 33763629 PMCID: PMC7978400 DOI: 10.1021/acsanm.0c02465] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/06/2020] [Indexed: 05/03/2023]
Abstract
Non-viral delivery systems are generally of low efficiency, which limits their use in gene therapy and editing applications. We previously developed a technology termed glycosaminoglycan (GAG)-binding enhanced transduction (GET) to efficiently deliver a variety of cargos intracellularly; our system employs GAG-binding peptides, which promote cell targeting, and cell penetrating peptides (CPPs), which enhance endocytotic cell internalization. Herein, we describe a further modification by combining gene delivery and magnetic targeting with the GET technology. We associated GET peptides, plasmid (p)DNA, and iron oxide superparamagnetic nanoparticles (MNPs), allowing rapid and targeted GET-mediated uptake by application of static magnetic fields in NIH3T3 cells. This produced effective transfection levels (significantly higher than the control) with seconds to minutes of exposure and localized gene delivery two orders of magnitude higher in targeted over non-targeted cell monolayers using magnetic fields (in 15 min exposure delivering GFP reporter pDNA). More importantly, high cell membrane targeting by GET-DNA and MNP co-complexes and magnetic fields allowed further enhancement to endocytotic uptake, meaning that the nucleic acid cargo was rapidly internalized beyond that of GET complexes alone (GET-DNA). Magnetofection by MNPs combined with GET-mediated delivery allows magnetic field-guided local transfection in vitro and could facilitate focused gene delivery for future regenerative and disease-targeted therapies in vivo.
Collapse
Affiliation(s)
- Lia A. Blokpoel Ferreras
- Regenerative
Medicine & Cellular Therapies Division, The University of Nottingham
Biodiscovery Institute (BDI), School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
| | - Sze Yan Chan
- Regenerative
Medicine & Cellular Therapies Division, The University of Nottingham
Biodiscovery Institute (BDI), School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
| | - Saul Vazquez Reina
- School
of Veterinary Sciences, University of Nottingham, Nottingham NG7 2RD, U.K.
| | - James E. Dixon
- Regenerative
Medicine & Cellular Therapies Division, The University of Nottingham
Biodiscovery Institute (BDI), School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
| |
Collapse
|
83
|
Pennetta C, Bono N, Ponti F, Bellucci MC, Viani F, Candiani G, Volonterio A. Multifunctional Neomycin-Triazine-Based Cationic Lipids for Gene Delivery with Antibacterial Properties. Bioconjug Chem 2021; 32:690-701. [PMID: 33470802 PMCID: PMC8154203 DOI: 10.1021/acs.bioconjchem.0c00616] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
![]()
Cationic
lipids (CLs) have gained significant attention among nonviral
gene delivery vectors due to their ease of synthesis and functionalization
with multivalent moieties. In particular, there is an increasing request
for multifunctional CLs having gene delivery capacity and antibacterial
activity. Herein, we describe the design and synthesis of a novel
class of aminoglycoside (AG)-based multifunctional vectors with high
transfection efficiency and noticeable antibacterial properties. Specifically,
cationic amphiphiles were built on a triazine scaffold, allowing for
an easy derivatization with up to three potentially different substituents,
such as neomycin (Neo) that serves as the polar head and one or two
lipophilic tails, namely stearyl (ST) and oleyl (OL) alkyl chains
and cholesteryl (Chol) tail. With the aim to shed more light on the
effect of different types and numbers of lipophilic moieties on the
ability of CLs to condense and transfect cells, the performance of
Neo–triazine-based derivatives as gene delivery vectors was
evaluated and compared. The ability of Neo–triazine-based derivatives
to act as antimicrobial agents was evaluated as well. Neo–triazine-based
CLs invariably exhibited excellent DNA condensation ability, even
at a low charge ratio (CR, +/−). Besides, each derivative showed
very good transfection performance at its optimal CR on two different
cell lines, along with negligible cytotoxicity. CLs bearing symmetric
two-tailed OL proved to be the most effective in transfection. Interestingly,
Neo–triazine-based derivatives, used as either free lipids
or lipoplexes, exhibited strong antibacterial activity against Gram-negative
bacteria, especially in the case of CLs bearing one or two aliphatic
chains. Altogether, these results highlight the potential of Neo–triazine-based
derivatives as effective multifunctional nonviral gene delivery vectors.
Collapse
Affiliation(s)
- Chiara Pennetta
- Department of Chemistry, Materials, and Chemical Engineering "G. Natta", Politecnico di Milano, Via Mancinelli 7, Milan 20131, Italy
| | - Nina Bono
- Department of Chemistry, Materials, and Chemical Engineering "G. Natta", Politecnico di Milano, Via Mancinelli 7, Milan 20131, Italy
| | - Federica Ponti
- Department of Chemistry, Materials, and Chemical Engineering "G. Natta", Politecnico di Milano, Via Mancinelli 7, Milan 20131, Italy.,Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met-Materials Engineering & Research Center of CHU de Quebec, Division of Regenerative Medicine, Laval University, Quebec City, Quebec G1 V 0A6, Canada
| | - Maria Cristina Bellucci
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, Via Celoria 2, Milan 20133, Italy
| | - Fiorenza Viani
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimiche "G. Natta" (SCITEC), Via Mario Bianco 9, Milan 20131, Italy
| | - Gabriele Candiani
- Department of Chemistry, Materials, and Chemical Engineering "G. Natta", Politecnico di Milano, Via Mancinelli 7, Milan 20131, Italy
| | - Alessandro Volonterio
- Department of Chemistry, Materials, and Chemical Engineering "G. Natta", Politecnico di Milano, Via Mancinelli 7, Milan 20131, Italy.,Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimiche "G. Natta" (SCITEC), Via Mario Bianco 9, Milan 20131, Italy
| |
Collapse
|
84
|
Riva L, Fiorati A, Punta C. Synthesis and Application of Cellulose-Polyethyleneimine Composites and Nanocomposites: A Concise Review. MATERIALS (BASEL, SWITZERLAND) 2021; 14:473. [PMID: 33498164 PMCID: PMC7863743 DOI: 10.3390/ma14030473] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/05/2021] [Accepted: 01/15/2021] [Indexed: 12/11/2022]
Abstract
Cellulose/polyethyleneimine composites have increasingly attracted the attention of scientific community, devoted to the design and development of new synthetic strategies and materials for different application fields. In this review, after introducing the main characteristics of the two polymeric components, we provide in the second section a critical overview on the main protocols for the synthesis of these composites, considering both the several cellulose sources and forms, and the different cross-linkers and cross-linking procedures developed for this purpose, outlining advantages and limits for the reported approaches. The last section analyses the principal results obtained in different application fields. A wide discussion is dedicated to the principal use of cellulose/polyethyleneimine composites as sorbents for water remediation from heavy metal ions and organic contaminants. Subsequently, we introduce the literature describing the use of these composites, functionalized appropriately, where necessary, as drug delivery systems, sensors, and heterogeneous catalysts for organic reactions. Finally, after a brief description of other random applications, we furnish a personal analysis of actual limits and potentialities for these systems.
Collapse
Affiliation(s)
| | | | - Carlo Punta
- Department of Chemistry, Materials, and Chemical Engineering “G. Natta” and INSTM Local Unit, Politecnico di Milano, 20131 Milano, Italy; (L.R.); (A.F.)
| |
Collapse
|
85
|
Shaabani E, Sharifiaghdam M, De Keersmaecker H, De Rycke R, De Smedt S, Faridi-Majidi R, Braeckmans K, Fraire JC. Layer by Layer Assembled Chitosan-Coated Gold Nanoparticles for Enhanced siRNA Delivery and Silencing. Int J Mol Sci 2021; 22:E831. [PMID: 33467656 PMCID: PMC7830320 DOI: 10.3390/ijms22020831] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 12/14/2022] Open
Abstract
Delivery of small interfering RNA (siRNA) provides one of the most powerful strategies for downregulation of therapeutic targets. Despite the widely explored capabilities of this strategy, intracellular delivery is hindered by a lack of carriers that have high stability, low toxicity and high transfection efficiency. Here we propose a layer by layer (LBL) self-assembly method to fabricate chitosan-coated gold nanoparticles (CS-AuNPs) as a more stable and efficient siRNA delivery system. Direct reduction of HAuCl4 in the presence of chitosan led to the formation of positively charged CS-AuNPs, which were subsequently modified with a layer of siRNA cargo molecules and a final chitosan layer to protect the siRNA and to have a net positive charge for good interaction with cells. Cytotoxicity, uptake, and downregulation of enhanced Green Fluorescent Protein (eGFP) in H1299-eGFP lung epithelial cells indicated that LBL-CS-AuNPs provided excellent protection of siRNA against enzymatic degradation, ensured good uptake in cells by endocytosis, facilitated endosomal escape of siRNA, and improved the overall silencing effect in comparison with commercial transfection reagents Lipofectamine and jetPEI®. Therefore, this work shows that LBL assembled CS-AuNPs are promising nanocarriers for enhanced intracellular siRNA delivery and silencing.
Collapse
Affiliation(s)
- Elnaz Shaabani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; (E.S.); (M.S.)
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, B-9000 Ghent, Belgium; (H.D.K.); (S.D.S.); (J.C.F.)
| | - Maryam Sharifiaghdam
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; (E.S.); (M.S.)
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, B-9000 Ghent, Belgium; (H.D.K.); (S.D.S.); (J.C.F.)
| | - Herlinde De Keersmaecker
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, B-9000 Ghent, Belgium; (H.D.K.); (S.D.S.); (J.C.F.)
- Center for Advanced Light Microscopy, Ghent University, 9000 Ghent, Belgium
| | - Riet De Rycke
- Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium and VIB Center for Inflammation Research, 9052 Ghent, Belgium;
- Ghent University Expertise Centre for Transmission Electron Microscopy and VIB BioImaging Core, 9052 Ghent, Belgium
| | - Stefaan De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, B-9000 Ghent, Belgium; (H.D.K.); (S.D.S.); (J.C.F.)
- Center for Advanced Light Microscopy, Ghent University, 9000 Ghent, Belgium
| | - Reza Faridi-Majidi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; (E.S.); (M.S.)
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, B-9000 Ghent, Belgium; (H.D.K.); (S.D.S.); (J.C.F.)
- Center for Advanced Light Microscopy, Ghent University, 9000 Ghent, Belgium
| | - Juan C. Fraire
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, B-9000 Ghent, Belgium; (H.D.K.); (S.D.S.); (J.C.F.)
| |
Collapse
|
86
|
Progress of cationic gene delivery reagents for non-viral vector. Appl Microbiol Biotechnol 2021; 105:525-538. [PMID: 33394152 DOI: 10.1007/s00253-020-11028-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/18/2020] [Accepted: 11/22/2020] [Indexed: 12/13/2022]
Abstract
Gene delivery systems play a vital role in gene therapy and recombinant protein production. The advantages of using gene delivery reagents for non-viral vector include the capacity to accommodate a large packaging load and their low or absent immunogenicity. Furthermore, they are easy to produce at a large scale and preserve. Gene delivery reagents for non-viral vector are commonly used for transfecting a variety of cells and tissues. It is mainly composed of liposomes and non-liposome cationic polymers. According to the different head structures used, the non-viral cationic transfection reagents include a quaternary ammonium salt, amine, amino acid or polypeptide, guanidine salt, and a heterocyclic ring. This article summarizes these approaches and developments of types and components of transfection reagents and optimization of gene delivery. The optimization of mammalian cell transient recombinant protein expression system and cationic reagents for clinical or clinical trials are also discussed.
Collapse
|
87
|
Hosseinpour S, Walsh LJ. Laser-assisted nucleic acid delivery: A systematic review. JOURNAL OF BIOPHOTONICS 2021; 14:e202000295. [PMID: 32931155 DOI: 10.1002/jbio.202000295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/26/2020] [Accepted: 09/13/2020] [Indexed: 06/11/2023]
Abstract
Gene therapy has become an effective treatment modality for some conditions. Laser light may augment or enhance gene therapy through photomechanical, photothermal, and photochemical. This review examined the evidence base for laser therapy to enhance nucleic acid transfection in mammalian cells. An electronic search of MEDLINE, Scopus, EMBASE, Web of Science, and Google Scholar was performed, covering all available years. The preferred reporting items for systematic reviews and meta-analyses guideline for systematic reviews was used for designing the study and analyzing the results. In total, 49 studies of laser irradiation for nucleic acid delivery were included. Key approaches were optoporation, photomechanical gene transfection, and photochemical internalization. Optoporation is better suited to cells in culture, photomechanical and photochemical approaches appear well suited to in vivo use. Additional studies explored the impact of photothermal for enhancing gene transfection. Each approach has merits and limitations. Augmenting nucleic acid delivery using laser irradiation is a promising method for improving gene therapy. Laser protocols can be non-invasive because of the penetration of desirable wavelengths of light, but it depends on various parameters such as power density, treatment duration, irradiation mode, etc. The current protocols show low efficiency, and there is a need for further work to optimize irradiation parameters.
Collapse
Affiliation(s)
- Sepanta Hosseinpour
- School of Dentistry, Oral Health Centre, The University of Queensland, Brisbane, Australia
| | - Laurence J Walsh
- School of Dentistry, Oral Health Centre, The University of Queensland, Brisbane, Australia
| |
Collapse
|
88
|
Costard LS, Kelly DC, Power RN, Hobbs C, Jaskaniec S, Nicolosi V, Cavanagh BL, Curtin CM, O’Brien FJ. Layered Double Hydroxide as a Potent Non-viral Vector for Nucleic Acid Delivery Using Gene-Activated Scaffolds for Tissue Regeneration Applications. Pharmaceutics 2020; 12:pharmaceutics12121219. [PMID: 33339452 PMCID: PMC7765978 DOI: 10.3390/pharmaceutics12121219] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/02/2020] [Accepted: 12/10/2020] [Indexed: 02/04/2023] Open
Abstract
Nonviral vectors offer a safe alternative to viral vectors for gene therapy applications, albeit typically exhibiting lower transfection efficiencies. As a result, there remains a significant need for the development of a nonviral delivery system with low cytotoxicity and high transfection efficacy as a tool for safe and transient gene delivery. This study assesses MgAl-NO3 layered double hydroxide (LDH) as a nonviral vector to deliver nucleic acids (pDNA, miRNA and siRNA) to mesenchymal stromal cells (MSCs) in 2D culture and using a 3D tissue engineering scaffold approach. Nanoparticles were formulated by complexing LDH with pDNA, microRNA (miRNA) mimics and inhibitors, and siRNA at varying mass ratios of LDH:nucleic acid. In 2D monolayer, pDNA delivery demonstrated significant cytotoxicity issues, and low cellular transfection was deemed to be a result of the poor physicochemical properties of the LDH–pDNA nanoparticles. However, the lower mass ratios required to successfully complex with miRNA and siRNA cargo allowed for efficient delivery to MSCs. Furthermore, incorporation of LDH–miRNA nanoparticles into collagen-nanohydroxyapatite scaffolds resulted in successful overexpression of miRNA in MSCs, demonstrating the development of an efficacious miRNA delivery platform for gene therapy applications in regenerative medicine.
Collapse
Affiliation(s)
- Lara S. Costard
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St Stephen’s Green, D02 YN77 Dublin, Ireland; (L.S.C.); (D.C.K.); (R.N.P.)
| | - Domhnall C. Kelly
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St Stephen’s Green, D02 YN77 Dublin, Ireland; (L.S.C.); (D.C.K.); (R.N.P.)
- Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland, Galway (NUI, Galway), H91 TK33 Galway, Ireland
| | - Rachael N. Power
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St Stephen’s Green, D02 YN77 Dublin, Ireland; (L.S.C.); (D.C.K.); (R.N.P.)
| | - Christopher Hobbs
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and Trinity College Dublin (TCD), College Green, D02 PN40 Dublin, Ireland; (C.H.); (S.J.); (V.N.)
- School of Chemistry and Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, College Green, D02 PN40 Dublin, Ireland
| | - Sonia Jaskaniec
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and Trinity College Dublin (TCD), College Green, D02 PN40 Dublin, Ireland; (C.H.); (S.J.); (V.N.)
- School of Chemistry and Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, College Green, D02 PN40 Dublin, Ireland
| | - Valeria Nicolosi
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and Trinity College Dublin (TCD), College Green, D02 PN40 Dublin, Ireland; (C.H.); (S.J.); (V.N.)
- School of Chemistry and Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, College Green, D02 PN40 Dublin, Ireland
| | - Brenton L. Cavanagh
- Cellular and Molecular Imaging Core, RCSI, 123 St Stephen’s Green, D02 YN77 Dublin, Ireland;
| | - Caroline M. Curtin
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St Stephen’s Green, D02 YN77 Dublin, Ireland; (L.S.C.); (D.C.K.); (R.N.P.)
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and Trinity College Dublin (TCD), College Green, D02 PN40 Dublin, Ireland; (C.H.); (S.J.); (V.N.)
- Trinity Centre for BioMedical Engineering, Trinity Biomedical Sciences Institute, TCD, College Green, D02 PN40 Dublin, Ireland
- Correspondence: (C.M.C.); (F.J.O.); Tel.: +353-1-4028620 (C.M.C.); +353-1-4028533 (F.J.O.)
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), 123 St Stephen’s Green, D02 YN77 Dublin, Ireland; (L.S.C.); (D.C.K.); (R.N.P.)
- Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland, Galway (NUI, Galway), H91 TK33 Galway, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and Trinity College Dublin (TCD), College Green, D02 PN40 Dublin, Ireland; (C.H.); (S.J.); (V.N.)
- Trinity Centre for BioMedical Engineering, Trinity Biomedical Sciences Institute, TCD, College Green, D02 PN40 Dublin, Ireland
- Correspondence: (C.M.C.); (F.J.O.); Tel.: +353-1-4028620 (C.M.C.); +353-1-4028533 (F.J.O.)
| |
Collapse
|
89
|
Rivero-Barbarroja G, Benito JM, Ortiz Mellet C, García Fernández JM. Cyclodextrin-Based Functional Glyconanomaterials. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2517. [PMID: 33333914 PMCID: PMC7765426 DOI: 10.3390/nano10122517] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 12/29/2022]
Abstract
Cyclodextrins (CDs) have long occupied a prominent position in most pharmaceutical laboratories as "off-the-shelve" tools to manipulate the pharmacokinetics of a broad range of active principles, due to their unique combination of biocompatibility and inclusion abilities. The development of precision chemical methods for their selective functionalization, in combination with "click" multiconjugation procedures, have further leveraged the nanoscaffold nature of these oligosaccharides, creating a direct link between the glyco and the nano worlds. CDs have greatly contributed to understand and exploit the interactions between multivalent glycodisplays and carbohydrate-binding proteins (lectins) and to improve the drug-loading and functional properties of nanomaterials through host-guest strategies. The whole range of capabilities can be enabled through self-assembly, template-assisted assembly or covalent connection of CD/glycan building blocks. This review discusses the advancements made in this field during the last decade and the amazing variety of functional glyconanomaterials empowered by the versatility of the CD component.
Collapse
Affiliation(s)
- Gonzalo Rivero-Barbarroja
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, 41012 Seville, Spain; (G.R.-B.); (C.O.M.)
| | - Juan Manuel Benito
- Instituto de Investigaciones Químicas (IIQ), CSIC, Universidad de Sevilla, 41092 Sevilla, Spain;
| | - Carmen Ortiz Mellet
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, 41012 Seville, Spain; (G.R.-B.); (C.O.M.)
| | | |
Collapse
|
90
|
Yang R, Chen F, Guo J, Zhou D, Luan S. Recent advances in polymeric biomaterials-based gene delivery for cartilage repair. Bioact Mater 2020; 5:990-1003. [PMID: 32671293 PMCID: PMC7338882 DOI: 10.1016/j.bioactmat.2020.06.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/29/2020] [Accepted: 06/10/2020] [Indexed: 12/16/2022] Open
Abstract
Untreated articular cartilage damage normally results in osteoarthritis and even disability that affects millions of people. However, both the existing surgical treatment and tissue engineering approaches are unable to regenerate the original structures of articular cartilage durably, and new strategies for integrative cartilage repair are needed. Gene therapy provides local production of therapeutic factors, especially guided by biomaterials can minimize the diffusion and loss of the genes or gene complexes, achieve accurate spatiotemporally release of gene products, thus provideing long-term treatment for cartilage repair. The widespread application of gene therapy requires the development of safe and effective gene delivery vectors and supportive gene-activated matrices. Among them, polymeric biomaterials are particularly attractive due to their tunable physiochemical properties, as well as excellent adaptive performance. This paper reviews the recent advances in polymeric biomaterial-guided gene delivery for cartilage repair, with an emphasis on the important role of polymeric biomaterials in delivery systems.
Collapse
Affiliation(s)
- Ran Yang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
- College of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Fei Chen
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Jinshan Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Dongfang Zhou
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Shifang Luan
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
- College of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| |
Collapse
|
91
|
Gantenbein B, Tang S, Guerrero J, Higuita-Castro N, Salazar-Puerta AI, Croft AS, Gazdhar A, Purmessur D. Non-viral Gene Delivery Methods for Bone and Joints. Front Bioeng Biotechnol 2020; 8:598466. [PMID: 33330428 PMCID: PMC7711090 DOI: 10.3389/fbioe.2020.598466] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
Viral carrier transport efficiency of gene delivery is high, depending on the type of vector. However, viral delivery poses significant safety concerns such as inefficient/unpredictable reprogramming outcomes, genomic integration, as well as unwarranted immune responses and toxicity. Thus, non-viral gene delivery methods are more feasible for translation as these allow safer delivery of genes and can modulate gene expression transiently both in vivo, ex vivo, and in vitro. Based on current studies, the efficiency of these technologies appears to be more limited, but they are appealing for clinical translation. This review presents a summary of recent advancements in orthopedics, where primarily bone and joints from the musculoskeletal apparatus were targeted. In connective tissues, which are known to have a poor healing capacity, and have a relatively low cell-density, i.e., articular cartilage, bone, and the intervertebral disk (IVD) several approaches have recently been undertaken. We provide a brief overview of the existing technologies, using nano-spheres/engineered vesicles, lipofection, and in vivo electroporation. Here, delivery for microRNA (miRNA), and silencing RNA (siRNA) and DNA plasmids will be discussed. Recent studies will be summarized that aimed to improve regeneration of these tissues, involving the delivery of bone morphogenic proteins (BMPs), such as BMP2 for improvement of bone healing. For articular cartilage/osteochondral junction, non-viral methods concentrate on targeted delivery to chondrocytes or MSCs for tissue engineering-based approaches. For the IVD, growth factors such as GDF5 or GDF6 or developmental transcription factors such as Brachyury or FOXF1 seem to be of high clinical interest. However, the most efficient method of gene transfer is still elusive, as several preclinical studies have reported many different non-viral methods and clinical translation of these techniques still needs to be validated. Here we discuss the non-viral methods applied for bone and joint and propose methods that can be promising in clinical use.
Collapse
Affiliation(s)
- Benjamin Gantenbein
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Shirley Tang
- Department of Biomedical Engineering and Department of Orthopaedics, Spine Research Institute Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Julien Guerrero
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering and Department of Surgery, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Ana I Salazar-Puerta
- Department of Biomedical Engineering and Department of Surgery, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Andreas S Croft
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Amiq Gazdhar
- Department of Pulmonary Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Devina Purmessur
- Department of Biomedical Engineering and Department of Orthopaedics, Spine Research Institute Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
92
|
Neva T, Carbajo-Gordillo AI, Benito JM, Lana H, Marcelo G, Ortiz Mellet C, Tros de Ilarduya C, Mendicuti F, García Fernández JM. Tuning the Topological Landscape of DNA-Cyclodextrin Nanocomplexes by Molecular Design. Chemistry 2020; 26:15259-15269. [PMID: 32710799 DOI: 10.1002/chem.202002951] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Indexed: 12/25/2022]
Abstract
Original molecular vectors that ensure broad flexibility to tune the shape and surface properties of plasmid DNA (pDNA) condensates are reported herein. The prototypic design involves a cyclodextrin (CD) platform bearing a polycationic cluster at the primary face and a doubly linked aromatic module bridging two consecutive monosaccharide units at the secondary face that behaves as a topology-encoding element. Subtle differences at the molecular level then translate into disparate morphologies at the nanoscale, including rods, worms, toroids, globules, ellipsoids, and spheroids. In vitro evaluation of the transfection capabilities revealed marked selectivity differences as a function of nanocomplex morphology. Remarkably high transfection efficiencies were associated with ellipsoidal or spherical shapes with a lamellar internal arrangement of pDNA chains and CD bilayers. Computational studies support that the stability of such supramolecular edifices is directly related to the tendency of the molecular vector to form noncovalent dimers upon DNA templating. Because the stability of the dimers depends on the protonation state of the polycationic clusters, the coaggregates display pH responsiveness, which facilitates endosomal escape and timely DNA release, a key step in successful transfection. The results provide a versatile strategy for the construction of fully synthetic and perfectly monodisperse nonviral gene delivery systems uniquely suited for optimization schemes.
Collapse
Affiliation(s)
- Tania Neva
- Institute for Chemical Research, IIQ, CSIC-Univ. Sevilla, C/ Américo Vespucio 49, 41092, Sevilla, Spain
| | - Ana I Carbajo-Gordillo
- Institute for Chemical Research, IIQ, CSIC-Univ. Sevilla, C/ Américo Vespucio 49, 41092, Sevilla, Spain
| | - Juan M Benito
- Institute for Chemical Research, IIQ, CSIC-Univ. Sevilla, C/ Américo Vespucio 49, 41092, Sevilla, Spain
| | - Hugo Lana
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, 31080, Pamplona, Spain
| | - Gema Marcelo
- Department of Analytical Chemistry, Physical Chemistry and Chemical Engineering, Instituto de Investigación Química, "Andrés M. del Rio" (IQAR), University of Alcalá, Campus Universitario Ctra. Madrid-Barcelona, Km 33.600, 28871, Alcalá de Henares, Spain
| | - Carmen Ortiz Mellet
- Department of Organic Chemistry, Faculty of Chemistry, University of Sevilla, C/ Prof García González 1, 41012, Sevilla, Spain
| | - Conchita Tros de Ilarduya
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, 31080, Pamplona, Spain
| | - Francisco Mendicuti
- Department of Analytical Chemistry, Physical Chemistry and Chemical Engineering, Instituto de Investigación Química, "Andrés M. del Rio" (IQAR), University of Alcalá, Campus Universitario Ctra. Madrid-Barcelona, Km 33.600, 28871, Alcalá de Henares, Spain
| | - José M García Fernández
- Institute for Chemical Research, IIQ, CSIC-Univ. Sevilla, C/ Américo Vespucio 49, 41092, Sevilla, Spain
| |
Collapse
|
93
|
Egorova A, Selutin A, Maretina M, Selkov S, Baranov V, Kiselev A. Characterization of iRGD-Ligand Modified Arginine-Histidine-Rich Peptides for Nucleic Acid Therapeutics Delivery to αvβ3 Integrin-Expressing Cancer Cells. Pharmaceuticals (Basel) 2020; 13:E300. [PMID: 33050526 PMCID: PMC7601072 DOI: 10.3390/ph13100300] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/30/2020] [Accepted: 10/03/2020] [Indexed: 12/20/2022] Open
Abstract
Efficient and specific delivery of nucleic acid (NA) therapeutics to tumor cells is extremely important for cancer gene therapy. Various therapeutic strategies include delivery of DNA-therapeutics such as immunostimulatory or suicide genes and delivery of siRNA-therapeutics able to silence expression of cancer-related genes. Peptides are a promising class of non-viral vehicles which are biodegradable and can efficiently condense, protect and specifically deliver NA to the cells. Here we designed arginine-histidine-rich peptide carriers consisting of an iRGD ligand to target αvβ3 integrins and studied them as vehicles for DNA and siRNA delivery to cancer cells. Combination of iRGD-modified and unmodified arginine-histidine-rich peptides during NA complexation resulted in carriers with different ligand contents. The NA-binding and protecting properties in vitro transfection efficiency and cytotoxicity of the DNA- and siRNA-polyplexes were studied and the most efficient carrier RGD1 was determined. The ability of the peptides to mediate specific intracellular uptake was confirmed inhuman cervical carcinoma (HeLa), human kidney (293T) and human pancreatic (PANC-1) cell lines with different αvβ3 integrins surface expression. By means of RGD1 carrier, efficient delivery of the herpes simplex virus (HSV-1) thymidine kinase gene to PANC-1 cells was demonstrated. Subsequent ganciclovir treatment led to a reduction of PANC-1 cells' viability by up to 54%. Efficient RNAi-mediated down-regulation of GFP and VEGFA gene expression was achieved in MDA-MB-231-GFP+ breast cancer and EA.hy926 endothelial cells, respectively, by means of RGD1/siRNA polyplexes. Here we demonstrated that the peptide carrier RGD1 can be considered as promising candidate for development of NA therapeutics delivery systems useful in cancer gene therapy.
Collapse
Affiliation(s)
- Anna Egorova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia; (A.E.); (M.M.); (V.B.)
- Institute of Chemistry, Saint Petersburg State University, Universitetskii pr. 26, 198504 Peterhoff, Russia
| | - Alexander Selutin
- Department of Immunology and Intercellular Interactions, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia; (A.S.); (S.S.)
| | - Marianna Maretina
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia; (A.E.); (M.M.); (V.B.)
- Institute of Chemistry, Saint Petersburg State University, Universitetskii pr. 26, 198504 Peterhoff, Russia
| | - Sergei Selkov
- Department of Immunology and Intercellular Interactions, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia; (A.S.); (S.S.)
| | - Vladislav Baranov
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia; (A.E.); (M.M.); (V.B.)
| | - Anton Kiselev
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia; (A.E.); (M.M.); (V.B.)
| |
Collapse
|
94
|
Madry H, Venkatesan JK, Carballo-Pedrares N, Rey-Rico A, Cucchiarini M. Scaffold-Mediated Gene Delivery for Osteochondral Repair. Pharmaceutics 2020; 12:pharmaceutics12100930. [PMID: 33003607 PMCID: PMC7601511 DOI: 10.3390/pharmaceutics12100930] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022] Open
Abstract
Osteochondral defects involve both the articular cartilage and the underlying subchondral bone. If left untreated, they may lead to osteoarthritis. Advanced biomaterial-guided delivery of gene vectors has recently emerged as an attractive therapeutic concept for osteochondral repair. The goal of this review is to provide an overview of the variety of biomaterials employed as nonviral or viral gene carriers for osteochondral repair approaches both in vitro and in vivo, including hydrogels, solid scaffolds, and hybrid materials. The data show that a site-specific delivery of therapeutic gene vectors in the context of acellular or cellular strategies allows for a spatial and temporal control of osteochondral neotissue composition in vitro. In vivo, implantation of acellular hydrogels loaded with nonviral or viral vectors has been reported to significantly improve osteochondral repair in translational defect models. These advances support the concept of scaffold-mediated gene delivery for osteochondral repair.
Collapse
Affiliation(s)
- Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany; (H.M.); (J.K.V.)
| | - Jagadeesh Kumar Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany; (H.M.); (J.K.V.)
| | - Natalia Carballo-Pedrares
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, S-15071 A Coruña, Spain; (N.C.-P.); (A.R.-R.)
| | - Ana Rey-Rico
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, S-15071 A Coruña, Spain; (N.C.-P.); (A.R.-R.)
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany; (H.M.); (J.K.V.)
- Correspondence: ; Tel.: +49-684-1162-4987; Fax: +49-684-1162-4988
| |
Collapse
|
95
|
Efficient Non-Viral Gene Modification of Mesenchymal Stromal Cells from Umbilical Cord Wharton's Jelly with Polyethylenimine. Pharmaceutics 2020; 12:pharmaceutics12090896. [PMID: 32971730 PMCID: PMC7559368 DOI: 10.3390/pharmaceutics12090896] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stromal cells (MSC) derived from human umbilical cord Wharton’s jelly (WJ) have a wide therapeutic potential in cell therapy and tissue engineering because of their multipotential capacity, which can be reinforced through gene therapy in order to modulate specific responses. However, reported methodologies to transfect WJ-MSC using cationic polymers are scarce. Here, WJ-MSC were transfected using 25 kDa branched- polyethylenimine (PEI) and a DNA plasmid encoding GFP. PEI/plasmid complexes were characterized to establish the best transfection efficiencies with lowest toxicity. Expression of MSC-related cell surface markers was evaluated. Likewise, immunomodulatory activity and multipotential capacity of transfected WJ-MSC were assessed by CD2/CD3/CD28-activated peripheral blood mononuclear cells (PBMC) cocultures and osteogenic and adipogenic differentiation assays, respectively. An association between cell number, PEI and DNA content, and transfection efficiency was observed. The highest transfection efficiency (15.3 ± 8.6%) at the lowest toxicity was achieved using 2 ng/μL DNA and 3.6 ng/μL PEI with 45,000 WJ-MSC in a 24-well plate format (200 μL). Under these conditions, there was no significant difference between the expression of MSC-identity markers, inhibitory effect on CD3+ T lymphocytes proliferation and osteogenic/adipogenic differentiation ability of transfected WJ-MSC, as compared with non-transfected cells. These results suggest that the functional properties of WJ-MSC were not altered after optimized transfection with PEI.
Collapse
|
96
|
Wang Y, Wagner E. Non-Viral Targeted Nucleic Acid Delivery: Apply Sequences for Optimization. Pharmaceutics 2020; 12:E888. [PMID: 32961908 PMCID: PMC7559072 DOI: 10.3390/pharmaceutics12090888] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/09/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
In nature, genomes have been optimized by the evolution of their nucleic acid sequences. The design of peptide-like carriers as synthetic sequences provides a strategy for optimizing multifunctional targeted nucleic acid delivery in an iterative process. The optimization of sequence-defined nanocarriers differs for different nucleic acid cargos as well as their specific applications. Supramolecular self-assembly enriched the development of a virus-inspired non-viral nucleic acid delivery system. Incorporation of DNA barcodes presents a complementary approach of applying sequences for nanocarrier optimization. This strategy may greatly help to identify nucleic acid carriers that can overcome pharmacological barriers and facilitate targeted delivery in vivo. Barcode sequences enable simultaneous evaluation of multiple nucleic acid nanocarriers in a single test organism for in vivo biodistribution as well as in vivo bioactivity.
Collapse
Affiliation(s)
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, D-81377 Munich, Germany;
| |
Collapse
|
97
|
Sharpe M, Barry J, Kefalas P. Clinical Adoption of Advanced Therapies: Challenges and Opportunities. J Pharm Sci 2020; 110:1877-1884. [PMID: 32918916 DOI: 10.1016/j.xphs.2020.08.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/25/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022]
Abstract
As the cell and gene therapy field matures the powerful therapeutic potential of these innovative therapies is starting to be shown, particularly in the fields of oncology and childhood immune deficiency diseases. However, as more therapies enter late stage clinical trials, advances and innovation are required in manufacturing, logistics, regulation, reimbursement and the healthcare setting to ensure that systems are in place to support wider clinical adoption of these promising treatments. A window of opportunity exists to implement new methodologies for best practice in both the ability to manufacture products reproducibly at scale, as well as ensuring healthcare systems are not overwhelmed by the variety and complexity of these new therapies and the additional burden they will place on already stretched facilities. If all interested parties work together it will be possible for the sector to develop the necessary processes, skilled staff and infrastructure needed as more treatments move from clinical trial to marketed products.
Collapse
Affiliation(s)
- Michaela Sharpe
- Moare Solutions Ltd, 99 Canterbury Road, Whitstable, Kent CT5 4HG, UK.
| | - Jacqueline Barry
- Cell and Gene Therapy Catapult, Guys Hospital, 12th Floor Tower Wing, Great Maze Pond, London SE1 9RT, UK
| | - Panos Kefalas
- Cell and Gene Therapy Catapult, Guys Hospital, 12th Floor Tower Wing, Great Maze Pond, London SE1 9RT, UK
| |
Collapse
|
98
|
Ma LL, Tang Q, Liu MX, Liu XY, Liu JY, Lu ZL, Gao YG, Wang R. [12]aneN 3-Based Gemini-Type Amphiphiles with Two-Photon Absorption Properties for Enhanced Nonviral Gene Delivery and Bioimaging. ACS APPLIED MATERIALS & INTERFACES 2020; 12:40094-40107. [PMID: 32805811 DOI: 10.1021/acsami.0c10718] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Although a plethora of nonviral gene vectors have been developed for potential gene therapy, imageable gemini surfactants with stimuli-responsiveness and high transfection efficiency are still scarce for gene delivery. Herein, three gemini amphiphiles (DEDPP-4/8/12) consisting of an aggregation-induced emission (AIE) central fluorophore: 5,6-diphenylpyrazine-2,3-diester (DEDPP), decorated with triazole-[12]aneN3 as the hydrophilic moiety and alkyl chains of various lengths as the hydrophobic moiety, were designed and synthesized for trackable gene delivery via optical imaging. All three amphiphiles exhibited ultralow critical micelle concentrations (CMCs) (up to 3.40 × 10-6 M), prominent two-photon absorption properties, and solvatochromic fluorescence. Gel electrophoresis assays demonstrated that the migration of plasmid DNA was completely retarded after condensation with these gemini amphiphiles at low concentrations (up to 10 μM). In addition, the ester bond in these amphiphiles may facilitate vector degradation and DNA release, in response to esterase and the acidic environment inside cells. Upon self-assembly with DOPE to form liposomes, DEDPP-8/DOPE achieved the best transfection efficiency in four cell lines, and the transfection efficiency of DEDPP-8/DOPE in HeLa cell lines was 23.5-fold higher than that of Lipo2000, which is unusually high for small organic molecule-based nonviral vectors. Furthermore, excellent transfection efficiency of DEDPP-8/DOPE was obtained in the presence of serum, and the red fluorescence protein (RFP) gene was successfully transfected in zebrafish embryos. Both one- and two-photon fluorescence imaging clearly demonstrated the delivery process of plasmid DNA. This study demonstrated that gemini-type amphiphiles composed of a two-photon fluorophore core conjugated with triazole-[12]aneN3 via an ester bond afforded an unprecedentedly high transfection efficiency with excellent biocompatibility, which may provide new insights for the design and development of multifunctional nonviral gene vectors for imageable gene delivery.
Collapse
Affiliation(s)
- Le-Le Ma
- Key Laboratory of Radiopharmaceutics, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Quan Tang
- Key Laboratory of Radiopharmaceutics, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ming-Xuan Liu
- Key Laboratory of Radiopharmaceutics, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Xu-Ying Liu
- Key Laboratory of Radiopharmaceutics, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Jin-Yu Liu
- Key Laboratory of Radiopharmaceutics, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Zhong-Lin Lu
- Key Laboratory of Radiopharmaceutics, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Yong-Guang Gao
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR 9990078, China
| |
Collapse
|
99
|
Carballo-Pedrares N, Fuentes-Boquete I, Díaz-Prado S, Rey-Rico A. Hydrogel-Based Localized Nonviral Gene Delivery in Regenerative Medicine Approaches-An Overview. Pharmaceutics 2020; 12:E752. [PMID: 32785171 PMCID: PMC7464633 DOI: 10.3390/pharmaceutics12080752] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 12/11/2022] Open
Abstract
Hydrogel-based nonviral gene delivery constitutes a powerful strategy in various regenerative medicine scenarios, as those concerning the treatment of musculoskeletal, cardiovascular, or neural tissues disorders as well as wound healing. By a minimally invasive administration, these systems can provide a spatially and temporarily defined supply of specific gene sequences into the target tissue cells that are overexpressing or silencing the original gene, which can promote natural repairing mechanisms to achieve the desired effect. In the present work, we provide an overview of the most avant-garde approaches using various hydrogels systems for controlled delivery of therapeutic nucleic acid molecules in different regenerative medicine approaches.
Collapse
Affiliation(s)
- Natalia Carballo-Pedrares
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, 15071 A Coruña, Spain; (N.C.-P.); (I.F.-B.); (S.D.-P.)
| | - Isaac Fuentes-Boquete
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, 15071 A Coruña, Spain; (N.C.-P.); (I.F.-B.); (S.D.-P.)
- Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15071 A Coruña, Galicia, Spain
| | - Silvia Díaz-Prado
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, 15071 A Coruña, Spain; (N.C.-P.); (I.F.-B.); (S.D.-P.)
- Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), 15071 A Coruña, Galicia, Spain
| | - Ana Rey-Rico
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, 15071 A Coruña, Spain; (N.C.-P.); (I.F.-B.); (S.D.-P.)
| |
Collapse
|
100
|
Song J, Ma P, Huang S, Wang J, Xie H, Jia B, Zhang W. Acylation of the antimicrobial peptide CAMEL for cancer gene therapy. Drug Deliv 2020; 27:964-973. [PMID: 32611259 PMCID: PMC8216477 DOI: 10.1080/10717544.2020.1787556] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Obtaining ideal gene delivery vectors is still a major goal in cancer gene therapy. CAMEL, a short hybrid antimicrobial peptide, can kill cancer cells by membrane lysis. In this study, we constructed a series of non-viral vectors by attaching fatty acids with different chain lengths to the N-terminus of CAMEL. Our results showed that the cellular uptake and transfection efficiency of acyl-CAMEL started to significantly increase from a chain length of 12 carbons. C18-CAMEL was screened for gene delivery because it had the highest transfection efficiency. Surprisingly, C18-CAMEL/plasmid complexes displayed strong endosomal escape activity after entering cells via endocytosis. Importantly, C18-CAMEL could deliver p53 plasmids to cancer cells and significantly inhibited cell proliferation by the expression of p53. In addition, the C18-CAMEL/p53 plasmid complexes and the MDM2 inhibitor nutlin-3a showed significantly synergistic anticancer activity against MCF-7 cells expressing wild-type p53. Conclusively, our study demonstrated that conjugation of stearic acid to antimicrobial peptides is a simple and successful approach for constructing efficient and economical non-viral vectors for cancer gene therapy.
Collapse
Affiliation(s)
- Jingjing Song
- The Institute of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Panpan Ma
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Sujie Huang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Juanli Wang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Huan Xie
- The Institute of Pharmacology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Bo Jia
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Wei Zhang
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|