1
|
De Rubis G, Paudel KR, Corrie L, Mehndiratta S, Patel VK, Kumbhar PS, Manjappa AS, Disouza J, Patravale V, Gupta G, Manandhar B, Rajput R, Robinson AK, Reyes RJ, Chakraborty A, Chellappan DK, Singh SK, Oliver BGG, Hansbro PM, Dua K. Applications and advancements of nanoparticle-based drug delivery in alleviating lung cancer and chronic obstructive pulmonary disease. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2793-2833. [PMID: 37991539 DOI: 10.1007/s00210-023-02830-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/30/2023] [Indexed: 11/23/2023]
Abstract
Lung cancer (LC) and chronic obstructive pulmonary disease (COPD) are among the leading causes of mortality worldwide. Cigarette smoking is among the main aetiologic factors for both ailments. These diseases share common pathogenetic mechanisms including inflammation, oxidative stress, and tissue remodelling. Current therapeutic approaches are limited by low efficacy and adverse effects. Consequentially, LC has a 5-year survival of < 20%, while COPD is incurable, underlining the necessity for innovative treatment strategies. Two promising emerging classes of therapy against these diseases include plant-derived molecules (phytoceuticals) and nucleic acid-based therapies. The clinical application of both is limited by issues including poor solubility, poor permeability, and, in the case of nucleic acids, susceptibility to enzymatic degradation, large size, and electrostatic charge density. Nanoparticle-based advanced drug delivery systems are currently being explored as flexible systems allowing to overcome these limitations. In this review, an updated summary of the most recent studies using nanoparticle-based advanced drug delivery systems to improve the delivery of nucleic acids and phytoceuticals for the treatment of LC and COPD is provided. This review highlights the enormous relevance of these delivery systems as tools that are set to facilitate the clinical application of novel categories of therapeutics with poor pharmacokinetic properties.
Collapse
Affiliation(s)
- Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Keshav Raj Paudel
- Centre of Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2007, Australia
| | - Leander Corrie
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Samir Mehndiratta
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Vyoma K Patel
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Popat S Kumbhar
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur, Maharashtra, 416113, India
| | - Arehalli Sidramappa Manjappa
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur, Maharashtra, 416113, India
- Department of Pharmaceutics, Vasantidevi Patil Institute of Pharmacy, Kodoli, Kolkapur, Maharashtra, 416114, India
| | - John Disouza
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist: Kolhapur, Maharashtra, 416113, India
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai, 400019, Maharashtra, India
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India, Chennai, India
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, 302017, India
| | - Bikash Manandhar
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Rashi Rajput
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Alexandra Kailie Robinson
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Ruby-Jean Reyes
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Amlan Chakraborty
- Division of Immunology, Immunity to Infection and Respiratory Medicine (DIIIRM), School of Biological Sciences I Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Dinesh Kumar Chellappan
- School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Sachin Kumar Singh
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Brian Gregory George Oliver
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Woolcock Institute of Medical Research, Macquarie University, Sydney, New South Wales, Australia
| | - Philip Michael Hansbro
- Centre of Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2007, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia.
| |
Collapse
|
2
|
Safety and Immunogenicity of Combined DNA-Polyethylenimine and Oral Bacterial Idiotypic Vaccine for Patients with B-Cell Non-Hodgkin Lymphoma: A Pilot Study. Cancers (Basel) 2022; 14:cancers14143298. [PMID: 35884359 PMCID: PMC9320637 DOI: 10.3390/cancers14143298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Immunoglobulin variable domains, or idiotypes, have been used as lymphoma-specific antigens for therapeutic vaccination against B-cell lymphomas in a number of clinical trials. The effectiveness of DNA vaccines significantly depends on the chosen method of DNA delivery. In this study, we applied the intramuscular injection of a DNA–PEI vaccine followed by an oral vaccine-carrying Salmonella boost for lymphoma patients, which was safe and well tolerated. The observed remission was accompanied by T-cell but not an antibody response to the vaccine in most of the patients. Abstract We report, in brief, the results of a phase I, non-randomized study of idiotypic DNA vaccination in patients with B-cell non-Hodgkin’s lymphoma (ISRCTN31090206). The DNA sequence of lymphoma-derived immunoglobulin variable regions was used as a tumor-specific antigen fused to the potato virus X coat protein. A conjugate of plasmid DNA with polyethylenimine was used for the intramuscular injections, followed by a boost with an oral live-attenuated Salmonella vaccine carrying the same plasmid. The patients with a complete or partial response to previous chemotherapy received one or two courses of vaccination, including four injections at monthly intervals. The vaccine was well tolerated, with low-grade adverse events. The T-cell immune responses were assessed by ELISpot, at last vaccine, one week and one month post-vaccination, and were detected in 11/14 (78.6%) of the patients. In cases of progression requiring chemotherapy, or the presence of a positive MRD after the first course of vaccination, the patients underwent a second course of vaccination. At the end point, 6/19 vaccinated patients had disease stabilization, while 13/19 were in complete remission. The overall survival was 100% at follow-up, of a median of 2.3 years.
Collapse
|
3
|
Lipid Nanoparticles for Organ-Specific mRNA Therapeutic Delivery. Pharmaceutics 2021; 13:pharmaceutics13101675. [PMID: 34683969 PMCID: PMC8538155 DOI: 10.3390/pharmaceutics13101675] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary This article belongs to the Special Issue mRNA Therapeutics: A Themed Issue in Honor of Professor Katalin Karikó. Abstract Advances in the using in vitro transcribed (IVT) modRNA in the past two decades, especially the tremendous recent success of mRNA vaccines against SARS-CoV-2, have brought increased attention to IVT mRNA technology. Despite its well-known use in infectious disease vaccines, IVT modRNA technology is being investigated mainly in cancer immunotherapy and protein replacement therapy, with ongoing clinical trials in both areas. One of the main barriers to progressing mRNA therapeutics to the clinic is determining how to deliver mRNA to target cells and protect it from degradation. Over the years, many different vehicles have been developed to tackle this issue. Desirable vehicles must be safe, stable and preferably organ specific for successful mRNA delivery to clinically relevant cells and tissues. In this review we discuss various mRNA delivery platforms, with particular focus on attempts to create organ-specific vehicles for therapeutic mRNA delivery.
Collapse
|
4
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
5
|
Rai N, Shihan M, Seeger W, Schermuly RT, Novoyatleva T. Genetic Delivery and Gene Therapy in Pulmonary Hypertension. Int J Mol Sci 2021; 22:ijms22031179. [PMID: 33503992 PMCID: PMC7865388 DOI: 10.3390/ijms22031179] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive complex fatal disease of multiple etiologies. Hyperproliferation and resistance to apoptosis of vascular cells of intimal, medial, and adventitial layers of pulmonary vessels trigger excessive pulmonary vascular remodeling and vasoconstriction in the course of pulmonary arterial hypertension (PAH), a subgroup of PH. Multiple gene mutation/s or dysregulated gene expression contribute to the pathogenesis of PAH by endorsing the proliferation and promoting the resistance to apoptosis of pulmonary vascular cells. Given the vital role of these cells in PAH progression, the development of safe and efficient-gene therapeutic approaches that lead to restoration or down-regulation of gene expression, generally involved in the etiology of the disease is the need of the hour. Currently, none of the FDA-approved drugs provides a cure against PH, hence innovative tools may offer a novel treatment paradigm for this progressive and lethal disorder by silencing pathological genes, expressing therapeutic proteins, or through gene-editing applications. Here, we review the effectiveness and limitations of the presently available gene therapy approaches for PH. We provide a brief survey of commonly existing and currently applicable gene transfer methods for pulmonary vascular cells in vitro and describe some more recent developments for gene delivery existing in the field of PH in vivo.
Collapse
Affiliation(s)
- Nabham Rai
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Mazen Shihan
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ralph T. Schermuly
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Tatyana Novoyatleva
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Correspondence:
| |
Collapse
|
6
|
Chauhan DS, Prasad R, Srivastava R, Jaggi M, Chauhan SC, Yallapu MM. Comprehensive Review on Current Interventions, Diagnostics, and Nanotechnology Perspectives against SARS-CoV-2. Bioconjug Chem 2020; 31:2021-2045. [PMID: 32680422 PMCID: PMC7425040 DOI: 10.1021/acs.bioconjchem.0c00323] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/16/2020] [Indexed: 02/06/2023]
Abstract
The coronavirus disease 2019 (COVID-19) has dramatically challenged the healthcare system of almost all countries. The authorities are struggling to minimize the mortality along with ameliorating the economic downturn. Unfortunately, until now, there has been no promising medicine or vaccine available. Herein, we deliver perspectives of nanotechnology for increasing the specificity and sensitivity of current interventional platforms toward the urgent need of quickly deployable solutions. This review summarizes the recent involvement of nanotechnology from the development of a biosensor to fabrication of a multifunctional nanohybrid system for respiratory and deadly viruses, along with the recent interventions and current understanding about severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
Collapse
Affiliation(s)
- Deepak S. Chauhan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Rajendra Prasad
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, USA
| | - Subhash C. Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, USA
| | - Murali M. Yallapu
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, USA
| |
Collapse
|
7
|
Hossian AKMN, Jois SD, Jonnalagadda SC, Mattheolabakis G. Nucleic Acid Delivery with α-Tocopherol-Polyethyleneimine-Polyethylene Glycol Nanocarrier System. Int J Nanomedicine 2020; 15:6689-6703. [PMID: 32982227 PMCID: PMC7494428 DOI: 10.2147/ijn.s259724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/30/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose Nucleic acid-based therapies are a promising therapeutic tool. The major obstacle in their clinical translation is their efficient delivery to the desired tissue. We developed a novel nanosized delivery system composed of conjugates of α-tocopherol, polyethyleneimine, and polyethylene glycol (TPP) to deliver nucleic acids. Methods We synthesized a panel of TPP molecules using different molecular weights of PEG and PEI and analyzed with various analytical approaches. The optimized version of TPP (TPP111 - the 1:1:1 molecular ratio) was self-assembled in water to produce nanostructures and then evaluated in diversified in vitro and in vivo studies. Results Through a panel of synthesized molecules, TPP111 conjugate components self-assembled in water, forming globular shaped nanostructures of ~90 nm, with high nucleic acid entrapment efficiency. The polymer had low cytotoxicity in vitro and protected nucleic acids from nucleases. Using a luciferase-expressing plasmid, TPP111-plasmid nano-complexes were rapidly up-taken by cancer cells in vitro and induced strong transfection, comparable to PEI. Colocalization of the nano-complexes and endosomes/lysosomes suggested an endosome-mediated uptake. Using a subcutaneous tumor model, intravenously injected nano-complexes preferentially accumulated to the tumor area over 24 h. Conclusion These results indicate that we successfully synthesized the TPP111 nanocarrier system, which can deliver nucleic acids in vitro and in vivo and merits further evaluation.
Collapse
Affiliation(s)
- A K M Nawshad Hossian
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Seetharama D Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | | | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| |
Collapse
|
8
|
Taschauer A, Polzer W, Pöschl S, Metz S, Tepe N, Decker S, Cyran N, Scholda J, Maier J, Bloß H, Anton M, Hofmann T, Ogris M, Sami H. Combined Chemisorption and Complexation Generate siRNA Nanocarriers with Biophysics Optimized for Efficient Gene Knockdown and Air-Blood Barrier Crossing. ACS APPLIED MATERIALS & INTERFACES 2020; 12:30095-30111. [PMID: 32515194 PMCID: PMC7467563 DOI: 10.1021/acsami.0c06608] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Current nucleic acid (NA) nanotherapeutic approaches face challenges because of shortcomings such as limited control on loading efficiency, complex formulation procedure involving purification steps, low load of NA cargo per nanoparticle, endosomal trapping, and hampered release inside the cell. When combined, these factors significantly limit the amount of biologically active NA delivered per cell in vitro, delivered dosages in vivo for a prolonged biological effect, and the upscalability potential, thereby warranting early consideration in the design and developmental phase. Here, we report a versatile nanotherapeutic platform, termed auropolyplexes, for improved and efficient delivery of small interfering RNA (siRNA). Semitelechelic, thiolated linear polyethylenimine (PEI) was chemisorbed onto gold nanoparticles to endow them with positive charge. A simple two-step complexation method offers tunable loading of siRNA at concentrations relevant for in vivo studies and the flexibility for inclusion of multiple functionalities without any purification steps. SiRNA was electrostatically complexed with these cationic gold nanoparticles and further condensed with polycation or polyethyleneglycol-polycation conjugates. The resulting auropolyplexes ensured complete complexation of siRNA into nanoparticles with a high load of ∼15,500 siRNA molecules/nanoparticle. After efficient internalization into the tumor cell, an 80% knockdown of the luciferase reporter gene was achieved. Auropolyplexes were applied intratracheally in Balb/c mice for pulmonary delivery, and their biodistribution were studied spatio-temporally and quantitatively by optical tomography. Auropolyplexes were well tolerated with ∼25% of the siRNA dose remaining in the lungs after 24 h. Importantly, siRNA was released from auropolyplexes in vivo and a fraction also crossed the air-blood barrier, which was then excreted via kidneys, whereas >97% of gold nanoparticles were retained in the lung. Linear PEI-based auropolyplexes offer a combination of successful endosomal escape and better biocompatibility profile in vivo. Taken together, combined chemisorption and complexation endow auropolyplexes with crucial biophysical attributes, enabling a versatile and upscalable nanogold-based platform for siRNA delivery in vitro and in vivo.
Collapse
Affiliation(s)
- Alexander Taschauer
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Wolfram Polzer
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Stefan Pöschl
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Slavica Metz
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Nathalie Tepe
- Department of Environmental Geosciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| | - Simon Decker
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Norbert Cyran
- Core Facility Cell
Imaging and Ultrastructure Research (CIUS), University of Vienna, 1090 Vienna, Austria
| | - Julia Scholda
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Julia Maier
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Hermann Bloß
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Martina Anton
- Institutes of Molecular Immunology and Experimental Oncology, Klinikum
rechts der Isar, Technische Universität
München, 81675 Munich, Germany
| | - Thilo Hofmann
- Department of Environmental Geosciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| | - Manfred Ogris
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
- Center for NanoScience (CeNS), Ludwig Maximilians
University, 80539 Munich, Germany
| | - Haider Sami
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| |
Collapse
|
9
|
Patiño Vargas MI, Mesa Cadavid M, Arenas Gómez CM, Diosa Arango J, Restrepo Múnera LM, Becerra Colorado NY. Polyplexes System to Enhance the LL-37 Antimicrobial Peptide Expression in Human Skin Cells. Tissue Eng Part A 2020; 26:400-410. [PMID: 31805827 DOI: 10.1089/ten.tea.2019.0196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Inefficient autologous tissue recovery in diverse skin injuries increases the susceptibility of patients to infections caused by multiresistant microorganisms, resulting in a high mortality rate. Nonviral transfection is an attractive alternative for these patients, where genetically modified cells incorporated into skin substitutes could release additional antimicrobial agents into the native skin. In this work, we have modulated the conditions of using a nonviral system for transfection of primary human keratinocytes and fibroblasts, consisting of a polymer/plasmid DNA (pDNA) complex called polyplex and its effects on the expression of LL-37 antimicrobial peptide. Linear and branched polyethylenimine (PEI) polymers in different weight concentrations were varied for evaluating the formation and colloidal characteristics of the polyplexes. The PEI/pDNA polyplexes with 19 nitrogen/phosphate ratio are nanometric particles (400 and 250 nm with linear and branched PEI, respectively) exhibiting positive surface (+30 ± 2 mV). Both kinds of polyplexes allowed the expression of a reporter gene and increased the human cathelicidin antimicrobial peptide gene expression in transfected keratinocytes and fibroblasts; however, greater cytotoxicity was observed when polyplexes formed with branched PEI were used. Moreover, cell culture supernatants from transfected cells with linear PEI/pDNA polyplexes showed enhanced antimicrobial activity (decrease of bacterial growth in 95.8%) against a Staphylococcus aureus strain in vitro. The study of the PEI/pDNA polyplexes formation allowed us to develop an improved transfection strategy of skin cells, promoting the production of LL-37 antimicrobial peptide. In the future, this strategy could be used for the construction of skin substitutes to prevent, reduce, or eliminate bacterial infections. Impact statement The results of this study contribute to the understanding of the polyplexes system in the genetic modification of skin cells and its effects on the expression of the LL-37 antimicrobial peptide. In the future, three-dimensional skin substitutes built with these cells could be an efficient way to decrease bacterial growth and prevent the infections in skin wounds.
Collapse
Affiliation(s)
- Maria Isabel Patiño Vargas
- Tissue Engineering and Cell Therapy Group, Faculty of Medicine, University of Antioquia, Medellín, Colombia
| | - Mónica Mesa Cadavid
- Materials Science Group, Faculty of Exact and Natural Sciences, The University Research Headquarters (SIU), University of Antioquia, Medellín, Colombia
| | - Claudia Marcela Arenas Gómez
- Marine Biological Laboratory, Eugene Bell Center for Regenerative Biology and Tissue Engineering, Woods Hole, Massachusetts
| | - Johnatan Diosa Arango
- Materials Science Group, Faculty of Exact and Natural Sciences, The University Research Headquarters (SIU), University of Antioquia, Medellín, Colombia
| | - Luz Marina Restrepo Múnera
- Tissue Engineering and Cell Therapy Group, Faculty of Medicine, University of Antioquia, Medellín, Colombia
| | | |
Collapse
|
10
|
Chen Z, Krishnamachary B, Pachecho-Torres J, Penet MF, Bhujwalla ZM. Theranostic small interfering RNA nanoparticles in cancer precision nanomedicine. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 12:e1595. [PMID: 31642207 DOI: 10.1002/wnan.1595] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/24/2022]
Abstract
Due to their ability to effectively downregulate the expression of target genes, small interfering RNA (siRNA) have emerged as promising candidates for precision medicine in cancer. Although some siRNA-based treatments have advanced to clinical trials, challenges such as poor stability during circulation, and less than optimal pharmacokinetics and biodistribution of siRNA in vivo present barriers to the systemic delivery of siRNA. In recent years, theranostic nanomedicine integrating siRNA delivery has attracted significant attention for precision medicine. Theranostic nanomedicine takes advantage of the high capacity of nanoplatforms to ferry cargo with imaging and therapeutic capabilities. These theranostic nanoplatforms have the potential to play a major role in gene specific treatments. Here we have reviewed recent advances in the use of theranostic nanoplatforms to deliver siRNA, and discussed the opportunities as well as challenges associated with this exciting technology. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Implantable Materials and Surgical Technologies > Nanomaterials and Implants.
Collapse
Affiliation(s)
- Zhihang Chen
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jesus Pachecho-Torres
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marie-France Penet
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
11
|
Garcia-Hernandez C, Freese AK, Rodriguez-Mendez ML, Wanekaya AK. In situ synthesis, stabilization and activity of protein-modified gold nanoparticles for biological applications. Biomater Sci 2019; 7:2511-2519. [DOI: 10.1039/c9bm00129h] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We demonstrate an in situ synthesis, stabilization and activity of a nanoparticle-based protein carrier platform via the Layer-by-Layer (LbL) technology.
Collapse
Affiliation(s)
- Celia Garcia-Hernandez
- Chemistry Department
- Missouri State University
- 901 S. National Ave
- USA
- Group UVASENS. Chemistry Department
| | | | | | - Adam K. Wanekaya
- Chemistry Department
- Missouri State University
- 901 S. National Ave
- USA
| |
Collapse
|
12
|
Cao W, Mishina M, Amoah S, Mboko WP, Bohannon C, McCoy J, Mittal SK, Gangappa S, Sambhara S. Nasal delivery of H5N1 avian influenza vaccine formulated with GenJet™ or in vivo-jetPEI ® induces enhanced serological, cellular and protective immune responses. Drug Deliv 2018. [PMID: 29542358 PMCID: PMC6058713 DOI: 10.1080/10717544.2018.1450909] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Avian influenza virus infection is a serious public health threat and preventive vaccination is the most cost-effective public health intervention strategy. Unfortunately, currently available unadjuvanted avian influenza vaccines are poorly immunogenic and alternative vaccine formulations and delivery strategies are in urgent need to reduce the high risk of avian influenza pandemics. Cationic polymers have been widely used as vectors for gene delivery in vitro and in vivo. In this study, we formulated H5N1 influenza vaccines with GenJet™ or in vivo-jetPEI®, and showed that these formulations significantly enhanced the immunogenicity of H5N1 vaccines and conferred protective immunity in a mouse model. Detailed analyses of adaptive immune responses revealed that both formulations induced mixed TH1/TH2 antigen-specific CD4 T-cell responses, antigen-specific cytotoxic CD8 T-cell and memory B-cell responses. Our findings suggest that cationic polymers merit future development as potential adjuvants for mucosal delivery of poorly immunogenic vaccines.
Collapse
Affiliation(s)
- Weiping Cao
- a Immunology and Pathogenesis Branch , National Center for Immunization and Respiratory Diseases , Atlanta , GA , USA.,b Influenza Division , Centers for Disease Control and Prevention , Atlanta , GA , USA
| | - Margarita Mishina
- a Immunology and Pathogenesis Branch , National Center for Immunization and Respiratory Diseases , Atlanta , GA , USA.,b Influenza Division , Centers for Disease Control and Prevention , Atlanta , GA , USA.,c Battelle Memorial Institute , Atlanta , GA , USA
| | - Samuel Amoah
- a Immunology and Pathogenesis Branch , National Center for Immunization and Respiratory Diseases , Atlanta , GA , USA.,b Influenza Division , Centers for Disease Control and Prevention , Atlanta , GA , USA.,c Battelle Memorial Institute , Atlanta , GA , USA
| | - Wadzanai P Mboko
- d Department of Comparative Pathobiology , Purdue University , West Lafayette , IN , USA
| | - Caitlin Bohannon
- a Immunology and Pathogenesis Branch , National Center for Immunization and Respiratory Diseases , Atlanta , GA , USA.,b Influenza Division , Centers for Disease Control and Prevention , Atlanta , GA , USA.,e Oak Ridge Institute for Science and Education (ORISE), CDC Fellowship Program , Oak Ridge , TN , USA
| | - James McCoy
- f Department of Pathology and Laboratory Medicine , Emory University , Atlanta , GA , USA
| | - Suresh K Mittal
- d Department of Comparative Pathobiology , Purdue University , West Lafayette , IN , USA
| | - Shivaprakash Gangappa
- a Immunology and Pathogenesis Branch , National Center for Immunization and Respiratory Diseases , Atlanta , GA , USA.,b Influenza Division , Centers for Disease Control and Prevention , Atlanta , GA , USA
| | - Suryaprakash Sambhara
- a Immunology and Pathogenesis Branch , National Center for Immunization and Respiratory Diseases , Atlanta , GA , USA.,b Influenza Division , Centers for Disease Control and Prevention , Atlanta , GA , USA
| |
Collapse
|
13
|
Robinson E, MacDonald KD, Slaughter K, McKinney M, Patel S, Sun C, Sahay G. Lipid Nanoparticle-Delivered Chemically Modified mRNA Restores Chloride Secretion in Cystic Fibrosis. Mol Ther 2018; 26:2034-2046. [PMID: 29910178 PMCID: PMC6094356 DOI: 10.1016/j.ymthe.2018.05.014] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 05/12/2018] [Accepted: 05/12/2018] [Indexed: 12/14/2022] Open
Abstract
The promise of gene therapy for the treatment of cystic fibrosis has yet to be fully clinically realized despite years of effort toward correcting the underlying genetic defect in the cystic fibrosis transmembrane conductance regulator (CFTR). mRNA therapy via nanoparticle delivery represents a powerful technology for the transfer of genetic material to cells with large, widespread populations, such as airway epithelia. We deployed a clinically relevant lipid-based nanoparticle (LNP) for packaging and delivery of large chemically modified CFTR mRNA (cmCFTR) to patient-derived bronchial epithelial cells, resulting in an increase in membrane-localized CFTR and rescue of its primary function as a chloride channel. Furthermore, nasal application of LNP-cmCFTR restored CFTR-mediated chloride secretion to conductive airway epithelia in CFTR knockout mice for at least 14 days. On day 3 post-transfection, CFTR activity peaked, recovering up to 55% of the net chloride efflux characteristic of healthy mice. This magnitude of response is superior to liposomal CFTR DNA delivery and is comparable with outcomes observed in the currently approved drug ivacaftor. LNP-cmRNA-based systems represent a powerful platform technology for correction of cystic fibrosis and other monogenic disorders.
Collapse
Affiliation(s)
- Ema Robinson
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Kelvin D MacDonald
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA; Department of Pediatrics, School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Kai Slaughter
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Madison McKinney
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Conroy Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA; Department of Radiation Medicine, School of Medicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA; Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97201, USA.
| |
Collapse
|
14
|
Zimmermann M, John D, Grigoriev D, Puretskiy N, Böker A. From 2D to 3D patches on multifunctional particles: how microcontact printing creates a new dimension of functionality. SOFT MATTER 2018; 14:2301-2309. [PMID: 29504010 PMCID: PMC5870046 DOI: 10.1039/c8sm00163d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 02/14/2018] [Indexed: 05/13/2023]
Abstract
A straightforward approach for the precise multifunctional surface modification of particles with three-dimensional patches using microcontact printing is presented. By comparison to previous works it was possible to not only control the diameter, but also to finely tune the thickness of the deposited layer, opening up the way for three-dimensional structures and orthogonal multifunctionality. The use of PEI as polymeric ink, PDMS stamps for microcontact printing on silica particles and the influence of different solvents during particle release on the creation of functional particles with three-dimensional patches are described. Finally, by introducing fluorescent properties by incorporation of quantum dots into patches and by particle self-assembly via avidin-biotin coupling, the versatility of this novel modification method is demonstrated.
Collapse
Affiliation(s)
- Marc Zimmermann
- Fraunhofer Institute for Applied Polymer Research IAP, D-14476 Potsdam-Golm, Germany. and Chair of Polymer Materials and Polymer Technologies, University Potsdam, D-14476 Potsdam-Golm, Germany
| | - Daniela John
- Fraunhofer Institute for Applied Polymer Research IAP, D-14476 Potsdam-Golm, Germany.
| | - Dmitry Grigoriev
- Fraunhofer Institute for Applied Polymer Research IAP, D-14476 Potsdam-Golm, Germany.
| | - Nikolay Puretskiy
- Fraunhofer Institute for Applied Polymer Research IAP, D-14476 Potsdam-Golm, Germany.
| | - Alexander Böker
- Fraunhofer Institute for Applied Polymer Research IAP, D-14476 Potsdam-Golm, Germany. and Chair of Polymer Materials and Polymer Technologies, University Potsdam, D-14476 Potsdam-Golm, Germany
| |
Collapse
|
15
|
Schlosser K, Taha M, Deng Y, Stewart DJ. Systemic delivery of MicroRNA mimics with polyethylenimine elevates pulmonary microRNA levels, but lacks pulmonary selectivity. Pulm Circ 2017; 8:2045893217750613. [PMID: 29251557 PMCID: PMC5753929 DOI: 10.1177/2045893217750613] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Reversing pathologic alterations in vascular microRNA (miRNA) expression represents a potential therapeutic strategy for pulmonary hypertension. While polyethylenimine (PEI) has previously been shown to be an effective vehicle for vascular lung-directed delivery of plasmid DNA, it remains unclear whether this utility is generalizable to miRNAs. Here we show that despite elevated lung levels, the intravenous infusion of PEI-miRNA mimic complexes fails to provide lung-selective delivery in rats.
Collapse
Affiliation(s)
- Kenny Schlosser
- 1 10055 Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Mohamad Taha
- 1 10055 Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,2 Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Yupu Deng
- 1 10055 Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Duncan J Stewart
- 1 10055 Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,2 Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,3 Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
16
|
Forcato DO, Fili AE, Alustiza FE, Lázaro Martínez JM, Bongiovanni Abel S, Olmos Nicotra MF, Alessio AP, Rodríguez N, Barbero C, Bosch P. Transfection of bovine fetal fibroblast with polyethylenimine (PEI) nanoparticles: effect of particle size and presence of fetal bovine serum on transgene delivery and cytotoxicity. Cytotechnology 2017; 69:655-665. [PMID: 28321779 DOI: 10.1007/s10616-017-0075-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 02/09/2017] [Indexed: 11/29/2022] Open
Abstract
The development of efficient transfection protocols for livestock cells is crucial for implementation of cell-based transgenic methods to produce genetically modified animals. We synthetized fully deacylated linear 22, 87 and 217 kDa polyethylenimine (PEI) nanoparticles and compared their transfection efficiency and cytotoxicity to commercial branched 25 kDa PEI and linear 58 kDa poly(allylamine) hydrochloride. We studied the effect of PEI size and presence of serum on transfection efficiency on primary cultures of bovine fetal fibroblasts and established cells lines (HEK 293 and Hep G2). We found that transfection efficiency was affected mainly by polymer/pDNA ratio and DNA concentration and in less extent by PEI MW. In bovine fibroblast, preincubation of PEI nanoparticles with fetal bovine serum (FBS) greatly increased percentage of cells expressing the transgene (up to 82%) while significantly decreased the polymer cytotoxic effect. 87 and 217 kDa PEI rendered the highest transfection rates in HEK 293 and Hep G2 cell lines (>50% transfected cells) with minimal cell toxicity. In conclusion, our results indicate that fully deacylated PEI of 87 and 217 kDa are useful DNA vehicles for non-viral transfection of primary cultures of bovine fetal fibroblast and HEK 293 and Hep G2 cell lines.
Collapse
Affiliation(s)
- D O Forcato
- Department of Molecular Biology, FCEFQyN, Universidad Nacional de Río Cuarto, Córdoba, Argentina.,National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - A E Fili
- Department of Molecular Biology, FCEFQyN, Universidad Nacional de Río Cuarto, Córdoba, Argentina.,National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - F E Alustiza
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina.,Department of Chemistry, FCEFQyN, Universidad Nacional de Río Cuarto, Córdoba, Argentina
| | - J M Lázaro Martínez
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina.,IQUIFIB-FFyB, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - S Bongiovanni Abel
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina.,Department of Chemistry, FCEFQyN, Universidad Nacional de Río Cuarto, Córdoba, Argentina
| | - M F Olmos Nicotra
- Department of Molecular Biology, FCEFQyN, Universidad Nacional de Río Cuarto, Córdoba, Argentina.,National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - A P Alessio
- Department of Molecular Biology, FCEFQyN, Universidad Nacional de Río Cuarto, Córdoba, Argentina.,National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - N Rodríguez
- Department of Molecular Biology, FCEFQyN, Universidad Nacional de Río Cuarto, Córdoba, Argentina
| | - C Barbero
- Department of Chemistry, FCEFQyN, Universidad Nacional de Río Cuarto, Córdoba, Argentina
| | - P Bosch
- Department of Molecular Biology, FCEFQyN, Universidad Nacional de Río Cuarto, Córdoba, Argentina. .,National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
17
|
Meleshko AN, Petrovskaya NA, Savelyeva N, Vashkevich KP, Doronina SN, Sachivko NV. Phase I clinical trial of idiotypic DNA vaccine administered as a complex with polyethylenimine to patients with B-cell lymphoma. Hum Vaccin Immunother 2017; 13:1-6. [PMID: 28272989 DOI: 10.1080/21645515.2017.1285477] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
We report on the design of a phase I, non-randomized, open-label study of idiotypic DNA vaccination in patients with B-cell non-Hodgkin's lymphoma (ISRCTN31090206). The study uses DNA fusion gene vaccination encoding patient-specific single chain variable fragment, or idiotype, linked to an immunostimulatory sequence. Two types of immunostimulatory sequence are being explored: potato virus X coat protein and human chemokine MIP3α. Linear polyethylenimine with low molecular weight (8 kDa) is used as a synthetic vehicle for vaccine delivery. Humoral and T-cellular immune responses to vaccination will be measured by ELISA and ELISPOT, respectively. The primary study endpoints are safety, tolerability and immunogenicity of DNA-PEI vaccination.
Collapse
Affiliation(s)
- A N Meleshko
- a Belarusian Research Center for Pediatric Oncology, Hematology and Immunology , Minsk , Belarus
| | - N A Petrovskaya
- b N.N. Alexandrov National Cancer Centre of Belarus , Minsk , Belarus
| | - N Savelyeva
- c Cancer Sciences Unit, Faculty of Medicine, University of Southampton , Southampton , UK
| | - K P Vashkevich
- a Belarusian Research Center for Pediatric Oncology, Hematology and Immunology , Minsk , Belarus
| | - S N Doronina
- a Belarusian Research Center for Pediatric Oncology, Hematology and Immunology , Minsk , Belarus
| | - N V Sachivko
- b N.N. Alexandrov National Cancer Centre of Belarus , Minsk , Belarus
| |
Collapse
|
18
|
Abstract
The use of gene delivery systems for the expression of antigenic proteins is an established means for activating a patient’s own immune system against the cancer they carry. Since tumor cells are poor antigen-presenting cells, cross-presentation of tumor antigens by dendritic cells (DCs) is essential for the generation of tumor-specific cytotoxic T-lymphocyte responses. A number of polymer-based nanomedicines have been developed to deliver genes into DCs, primarily by incorporating tumor-specific, antigen-encoding plasmid DNA with polycationic molecules to facilitate DNA loading and intracellular trafficking. Direct in vivo targeting of plasmid DNA to DC surface receptors can induce high transfection efficiency and long-term gene expression, essential for antigen loading onto major histocompatibility complex molecules and stimulation of T-cell responses. This chapter summarizes the physicochemical properties and biological information on polymer-based non-viral vectors used for targeting DCs, and discusses the main challenges for successful in vivo gene transfer into DCs.
Collapse
Affiliation(s)
- Kenneth A. Howard
- Department of Molecular Biology and Gen, Interdisciplinary Nanoscience Center (i, Aarhus, Denmark
| | - Thomas Vorup-Jensen
- Department of Biomedicine, Biophysical I, Aarhus University, Aarhus, Denmark
| | - Dan Peer
- Britannia Bldg, 2nd Fl, Rm 226, Tel-Aviv Univ, Dept Cell Research, Tel-Aviv, Israel
| |
Collapse
|
19
|
Kim N, Duncan GA, Hanes J, Suk JS. Barriers to inhaled gene therapy of obstructive lung diseases: A review. J Control Release 2016; 240:465-488. [PMID: 27196742 DOI: 10.1016/j.jconrel.2016.05.031] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/11/2016] [Accepted: 05/12/2016] [Indexed: 12/29/2022]
Abstract
Knowledge of genetic origins of obstructive lung diseases has made inhaled gene therapy an attractive alternative to the current standards of care that are limited to managing disease symptoms. Initial lung gene therapy clinical trials occurred in the early 1990s following the discovery of the genetic defect responsible for cystic fibrosis (CF), a monogenic disorder. However, despite over two decades of intensive effort, gene therapy has yet to help patients with CF or any other obstructive lung disease. The slow progress is due in part to poor understanding of the biological barriers to inhaled gene therapy. Encouragingly, clinical trials have shown that inhaled gene therapy with various viral vectors and non-viral gene vectors is well tolerated by patients, and continued research has provided valuable lessons and resources that may lead to future success of this therapeutic strategy. In this review, we first introduce representative obstructive lung diseases and examine limitations of currently available therapeutic options. We then review key components for successful execution of inhaled gene therapy, including gene delivery systems, primary physiological barriers and strategies to overcome them, and advances in preclinical disease models with which the most promising systems may be identified for human clinical trials.
Collapse
Affiliation(s)
- Namho Kim
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Gregg A Duncan
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Justin Hanes
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Environmental and Health Sciences, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Neurosurgery, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jung Soo Suk
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
20
|
Golan M, Feinshtein V, Polyak D, Scomparin A, Satchi-Fainaro R, David A. Inhibition of Gene Expression and Cancer Cell Migration by CD44v3/6-Targeted Polyion Complexes. Bioconjug Chem 2016; 27:947-60. [DOI: 10.1021/acs.bioconjchem.6b00020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
| | | | - Dina Polyak
- Department
of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Anna Scomparin
- Department
of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ronit Satchi-Fainaro
- Department
of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | | |
Collapse
|
21
|
Saffari M, Moghimi HR, Dass CR. Barriers to Liposomal Gene Delivery: from Application Site to the Target. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2016; 15:3-17. [PMID: 28228799 PMCID: PMC5242347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Gene therapy is a therapeutic approach to deliver genetic material into cells to alter their function in entire organism. One promising form of gene delivery system (DDS) is liposomes. The success of liposome-mediated gene delivery is a multifactorial issue and well-designed liposomal systems might lead to optimized gene transfection particularly in vivo. Liposomal gene delivery systems face different barriers from their site of application to their target, which is inside the cells. These barriers include presystemic obstacles (epithelial barriers), systemic barriers in blood circulation and cellular barriers. Epithelial barriers differ depending on the route of administration. Systemic barriers include enzymatic degradation, binding and opsonisation. Both of these barriers can act as limiting hurdles that genetic material and their vector should overcome before reaching the cells. Finally liposomes should overcome cellular barriers that include cell entrance, endosomal escape and nuclear uptake. These barriers and their impact on liposomal gene delivery will be discussed in this review.
Collapse
Affiliation(s)
- Mostafa Saffari
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran. ,Current Address: Department of Pharmaceutics, School of Pharmacy, Islamic Azad University, Tehran, Iran.
| | - Hamid Reza Moghimi
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran. ,
| | - Crispin R Dass
- School of Pharmacy, Faculty of Health Sciences, Curtin University, Perth, Australia
| |
Collapse
|
22
|
Gene delivery potential of biofunctional carbonate apatite nanoparticles in lungs. BIOMED RESEARCH INTERNATIONAL 2014; 2014:646787. [PMID: 25143941 PMCID: PMC4131073 DOI: 10.1155/2014/646787] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 06/03/2014] [Indexed: 11/30/2022]
Abstract
Existing nonviral gene delivery systems to lungs are inefficient and associated with dose limiting toxicity in mammalian cells. Therefore, carbonate apatite (CO3Ap) nanoparticles were examined as an alternative strategy for effective gene delivery to the lungs. This study aimed to (1) assess the gene delivery efficiency of CO3Ap in vitro and in mouse lungs, (2) evaluate the cytotoxicity effect of CO3Ap/pDNA in vitro, and (3) characterize the CO3Ap/pDNA complex formulations. A significantly high level of reporter gene expression was detected from the lung cell line transfected with CO3Ap/pDNA complex prepared in both serum and serum-free medium. Cytotoxicity analysis revealed that the percentage of the viable cells treated with CO3Ap to be almost similar to the untreated cells. Characterization analyses showed that the CO3Ap/pDNA complexes are in a nanometer range with aggregated spherical structures and tended to be more negatively charged. In the lung of mice, highest level of transgene expression was observed when CO3Ap (8 μL) was complexed with 40 μg of pDNA at day 1 after administration. Although massive reduction of gene expression was seen beyond day 1 post administration, the level of expression remained significant throughout the study period.
Collapse
|
23
|
Sun JY, Sun Y, Wu HJ, Zhang HX, Zhao ZH, Chen Q, Zhang ZG. Transgene therapy for rat anti-Thy1.1 glomerulonephritis via mesangial cell vector with a polyethylenimine/decorin nanocomplex. NANOSCALE RESEARCH LETTERS 2012; 7:451. [PMID: 22876812 PMCID: PMC3629717 DOI: 10.1186/1556-276x-7-451] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 07/09/2012] [Indexed: 06/01/2023]
Abstract
Polyethylenimine (PEI), a cationic polymer, is one of the most efficient non-viral vectors for transgene therapy. Decorin (DCN), a leucine-rich proteoglycan secreted by glomerular mesangial cells (MC), is a promising anti-fibrotic agent for the treatment of glomerulonephritis. In this study, we used PEI-DCN nanocomplexes with different N/P ratios to transfect MC in vitro and deliver the MC vector with PEI-DCN expressing into rat anti-Thy1.1 nephritis kidney tissue via injection into the left renal artery in vivo. The PEI-plasmid DNA complex at N/P 20 had the highest level of transfection efficiency and the lowest level of cytotoxicity in cultured MC. Following injection, the ex vivo gene was transferred successfully into the glomeruli of the rat anti-Thy1.1 nephritis model by the MC vector with the PEI-DCN complex. The exogenous MC with DCN expression was located mainly in the mesangium and the glomerular capillary. Over-expression of DCN in diseased glomeruli could result in the inhibition of collagen IV deposition and MC proliferation. The pathological changes of rat nephritis were alleviated following injection of the vector. These findings demonstrate that the DCN gene delivered by the PEI-DNA nanocomplex with the MC vector is a promising therapeutic method for the treatment of glomerulonephritis.
Collapse
Affiliation(s)
- Jian-Yong Sun
- Department of Pathology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Sun
- Department of Pathology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui-Juan Wu
- Department of Pathology, Shanghai Medical College, Fudan University, Shanghai, China
- Key Laboratory of Molecular Medicine, Ministry of Education of China, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong-Xia Zhang
- Department of Pathology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhong-Hua Zhao
- Department of Pathology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qi Chen
- Department of Pathology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Gang Zhang
- Department of Pathology, Shanghai Medical College, Fudan University, Shanghai, China
- Key Laboratory of Molecular Medicine, Ministry of Education of China, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Grandinetti G, Smith AE, Reineke TM. Membrane and nuclear permeabilization by polymeric pDNA vehicles: efficient method for gene delivery or mechanism of cytotoxicity? Mol Pharm 2012; 9:523-38. [PMID: 22175236 PMCID: PMC3524998 DOI: 10.1021/mp200368p] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The aim of this study is to compare the cytotoxicity mechanisms of linear PEI to two analogous polymers synthesized by our group: a hydroxyl-containing poly(l-tartaramidoamine) (T4) and a version containing an alkyl chain spacer poly(adipamidopentaethylenetetramine) (A4) by studying the cellular responses to polymer transfection. We have also synthesized analogues of T4 with different molecular weights (degrees of polymerization of 6, 12, and 43) to examine the role of molecular weight on the cytotoxicity mechanisms. Several mechanisms of polymer-induced cytotoxicity are investigated, including plasma membrane permeabilization, the formation of potentially harmful polymer degradation products during transfection including reactive oxygen species, and nuclear membrane permeabilization. We hypothesized that since cationic polymers are capable of disrupting the plasma membrane, they may also be capable of disrupting the nuclear envelope, which could be a potential mechanism of how the pDNA is delivered into the nucleus (other than nuclear envelope breakdown during mitosis). Using flow cytometry and confocal microscopy, we show that the polycations with the highest amount of protein expression and toxicity, PEI and T4(43), are capable of inducing nuclear membrane permeability. This finding is important for the field of nucleic acid delivery in that direct nucleus permeabilization could be not only a mechanism for pDNA nuclear import but also a potential mechanism of cytotoxicity and cell death. We also show that the production of reactive oxygen species is not a main mechanism of cytotoxicity, and that the presence or absence of hydroxyl groups and polymer length play a role in polyplex size and charge in addition to protein expression efficiency and toxicity.
Collapse
Affiliation(s)
- Giovanna Grandinetti
- Department of Chemistry, Virginia Polytechnic Institute and State University. Blacksburg, VA 24061
| | - Adam E. Smith
- Department of Chemistry, Virginia Polytechnic Institute and State University. Blacksburg, VA 24061
| | - Theresa M. Reineke
- Department of Chemistry, Virginia Polytechnic Institute and State University. Blacksburg, VA 24061
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
25
|
Catanese DJ, Fogg JM, Schrock DE, Gilbert BE, Zechiedrich L. Supercoiled Minivector DNA resists shear forces associated with gene therapy delivery. Gene Ther 2012; 19:94-100. [PMID: 21633394 PMCID: PMC3252587 DOI: 10.1038/gt.2011.77] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 03/04/2011] [Accepted: 03/09/2011] [Indexed: 02/08/2023]
Abstract
Supercoiled DNAs varying from 281 to 5302 bp were subjected to shear forces generated by aerosolization or sonication. DNA shearing strongly correlated with length. Typical sized plasmids (≥ 3000 bp) degraded rapidly. DNAs 2000-3000 bp persisted ~10 min. Even in the absence of condensing agents, supercoiled DNA <1200 bp survived nebulization, and increased forces of sonication were necessary to shear it. Circular vectors were considerably more resistant to shearing than linear vectors of the same length. DNA supercoiling afforded additional protection. These results show the potential of shear-resistant Minivector DNAs to overcome one of the major challenges associated with gene therapy delivery.
Collapse
Affiliation(s)
- D J Catanese
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - J M Fogg
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - D E Schrock
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- University of Texas MD Anderson Cancer Center School of Health Sciences, Houston, TX, USA
| | - B E Gilbert
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - L Zechiedrich
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Pharmacology, Baylor College of Medicine, Houston, TX, USA
- University of Texas MD Anderson Cancer Center School of Health Sciences, Houston, TX, USA
| |
Collapse
|
26
|
Grandinetti G, Ingle NP, Reineke TM. Interaction of poly(ethylenimine)-DNA polyplexes with mitochondria: implications for a mechanism of cytotoxicity. Mol Pharm 2011; 8:1709-19. [PMID: 21699201 DOI: 10.1021/mp200078n] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Poly(ethylenimine) (PEI) and PEI-based systems have been widely studied for use as nucleic acid delivery vehicles. However, many of these vehicles display high cytotoxicity, rendering them unfit for therapeutic use. By exploring the mechanisms that cause cytotoxicity, and through understanding structure-function relationships between polymers and intracellular interactions, nucleic acid delivery vehicles with precise intracellular properties can be tailored for specific function. Previous research has shown that PEI is able to depolarize mitochondria, but the exact mechanism as to how depolarization is induced remains elusive and therefore is the focus of the current study. Potential mechanisms for mitochondrial depolarization include direct mitochondrial membrane permeabilization by PEI or PEI polyplexes, activation of the mitochondrial permeability transition pore, and interference with mitochondrial membrane proton pumps, specifically Complex I of the electron transport chain and F(0)F(1)-ATPase. Herein, confocal microscopy and live cell imaging showed that PEI polyplexes do colocalize to some degree with mitochondria early in transfection, and the degree of colocalization increases over time. Cyclosporin a was used to prevent activation of the mitochondrial membrane permeability transition pore, and it was found that early in transfection cyclosporin a was unable to prevent the loss of mitochondrial membrane potential. Further studies done using rotenone and oligomycin to inhibit Complex I of the electron transport chain and F(0)F(1)-ATPase, respectively, indicate that both of these mitochondrial proton pumps are functioning during PEI transfection. Overall, we conclude that direct interaction between polyplexes and mitochondria may be the reason why mitochondrial function is impaired during PEI transfection.
Collapse
Affiliation(s)
- Giovanna Grandinetti
- Department of Chemistry & Macromolecules & Interfaces Institute, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061, United States
| | | | | |
Collapse
|
27
|
Impact of lentiviral vector-mediated transduction on the tightness of a polarized model of airway epithelium and effect of cationic polymer polyethylenimine. J Biomed Biotechnol 2010; 2010:103976. [PMID: 20617131 PMCID: PMC2896616 DOI: 10.1155/2010/103976] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Revised: 03/31/2010] [Accepted: 05/09/2010] [Indexed: 11/17/2022] Open
Abstract
Lentiviral (LV) vectors are promising agents for efficient and long-lasting gene transfer into the lung and for gene therapy of genetically determined pulmonary diseases, such as cystic fibrosis, however, they have not been evaluated for cytotoxicity and impact on the tightness of the airway epithelium. In this study, we evaluated the transduction efficiency of a last-generation LV vector bearing Green Fluorescent Protein (GFP) gene as well as cytotoxicity and tight junction (TJ) integrity in a polarized model of airway epithelial cells. High multiplicities of infection (MOI) showed to be cytotoxic, as assessed by increase in propidium iodide staining and decrease in cell viability, and harmful for the epithelial tightness, as demonstrated by the decrease of transepithelial resistance (TER) and delocalization of occludin from the TJs. To increase LV efficiency at low LV:cell ratio, we employed noncovalent association with the polycation branched 25 kDa polyethylenimine (PEI). Transduction of cells with PEI/LV particles resulted in 2.5–3.6-fold increase of percentage of GFP-positive cells only at the highest PEI:LV ratios (1×107 PEI molecules/transducing units with 50 MOI LV) as compared to plain LV. At this dose PEI/LV transduction resulted in 6.5 ± 2.4% of propidium iodide-positive cells. On the other hand, PEI/LV particles did not determine any alteration of TER and occludin localization. We conclude that PEI may be useful for improving the efficiency of gene transfer mediated by LV vectors in airway epithelial cells, in the absence of high acute cytotoxicity and alteration in epithelial tightness.
Collapse
|