1
|
Vasukutty A, Chahal S, Lee KH, Park SH, Kim SM, Shin E, Kim YJ, Kim D, Lee JA, Jeong HS, Park IK. Dual-Mechanism mRNA Delivery via Fluorinated-Sorbitol Polyplexes: Enhancing Cellular Uptake and Endosomal Escape for COVID-19 Vaccination. Adv Healthc Mater 2025; 14:e2403374. [PMID: 39723670 DOI: 10.1002/adhm.202403374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Advancements in mRNA delivery nanoparticles have significantly improved the potential for treating challenging diseases. Due to the inherent immunogenicity and rapid degradation of mRNA, specialized nanoparticles are required for efficient intracellular uptake, endosomal escape, and protection from lysosomal degradation. Although current methods enable transgene expression but achieving a balance between efficiency and toxicity remains challenging. In this study, an effective mRNA delivery system is developed by modifying a cationic polymer with sorbitol and fluorine, resulting in fluorinated polyethyleneimine with sorbitol functionalization (PFS). This polyplex enhances mRNA delivery through improved cellular uptake via sorbitol channels and caveolae-mediated endocytosis, while fluorination facilitates endosomal escape and mitigates toxicity. The formulation demonstrated successful expression of Gaussian luciferase mRNA in both Raw 264.7 cells and Balb/c mice. Additionally, intramuscular administration of the SARS-CoV-2 spike mRNA vaccine elicited robust immune responses comparable to Moderna's LNP formulation. The vaccine effectively neutralized the Wuhan variant strain of SARS-CoV-2, as shown by PRNT50 testing. These findings suggest that the PFS formulation is a promising candidate for developing polymeric mRNA vaccines targeting various infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Arathy Vasukutty
- Department of Biomedical Sciences, Biomedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, 322 Seoyang-ro, Hwasun, 58128, Republic of Korea
| | - Sahil Chahal
- Department of Biomedical Sciences, Biomedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, 322 Seoyang-ro, Hwasun, 58128, Republic of Korea
| | - Kyung-Hwa Lee
- Department of Pathology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, 322 Seoyang-ro, Hwasun, 58128, Republic of Korea
| | - So-Hee Park
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Health, Cheong-Ju, CheongJu-si, Chungbuk, 28160, Republic of Korea
| | - Soo-Min Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Health, Cheong-Ju, CheongJu-si, Chungbuk, 28160, Republic of Korea
| | - Eunkyung Shin
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Health, Cheong-Ju, CheongJu-si, Chungbuk, 28160, Republic of Korea
| | - You-Jin Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Health, Cheong-Ju, CheongJu-si, Chungbuk, 28160, Republic of Korea
| | - Dokeun Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Health, Cheong-Ju, CheongJu-si, Chungbuk, 28160, Republic of Korea
| | - Jung-Ah Lee
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Health, Cheong-Ju, CheongJu, Chungbuk, 28160, Republic of Korea
| | - Hye-Sook Jeong
- Division of Vaccine Clinical Research, Center for Vaccine Research, National Institute of Health, Cheong-Ju, CheongJu, Chungbuk, 28160, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences, Biomedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, 322 Seoyang-ro, Hwasun, 58128, Republic of Korea
- DR Cure Inc., Hwasun, 58128, Republic of Korea
| |
Collapse
|
2
|
Prasher P, Sharma M, Agarwal V, Singh SK, Gupta G, Dureja H, Dua K. Cationic cycloamylose based nucleic acid nanocarriers. Chem Biol Interact 2024; 395:111000. [PMID: 38614318 DOI: 10.1016/j.cbi.2024.111000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/02/2024] [Accepted: 04/07/2024] [Indexed: 04/15/2024]
Abstract
Nucleic acid delivery by viral and non-viral methods has been a cornerstone for the contemporary gene therapy aimed at correcting the defective genes, replacing of the missing genes, or downregulating the expression of anomalous genes is highly desirable for the management of various diseases. Ostensibly, it becomes paramount for the delivery vectors to intersect the biological barriers for accessing their destined site within the cellular environment. However, the lipophilic nature of biological membranes and their potential to limit the entry of large sized, charged, hydrophilic molecules thus presenting a sizeable challenge for the cellular integration of negatively charged nucleic acids. Furthermore, the susceptibility of nucleic acids towards the degrading enzymes (nucleases) in the lysosomes present in cytoplasm is another matter of concern for their cellular and nuclear delivery. Hence, there is a pressing need for the identification and development of cationic delivery systems which encapsulate the cargo nucleic acids where the charge facilitates their cellular entry by evading the membrane barriers, and the encapsulation shields them from the enzymatic attack in cytoplasm. Cycloamylose bearing a closed loop conformation presents a robust candidature in this regard owing to its remarkable encapsulating tendency towards nucleic acids including siRNA, CpG DNA, and siRNA. The presence of numerous hydroxyl groups on the cycloamylose periphery provides sites for its chemical modification for the introduction of cationic groups, including spermine, (3-Chloro-2 hydroxypropyl) trimethylammonium chloride (Q188), and diethyl aminoethane (DEAE). The resulting cationic cycloamylose possesses a remarkable transfection efficiency and provides stability to cargo oligonucleotides against endonucleases, in addition to modulating the undesirable side effects such as unwanted immune stimulation. Cycloamylose is known to interact with the cell membranes where they release certain membrane components such as phospholipids and cholesterol thereby resulting in membrane destabilization and permeabilization. Furthermore, cycloamylose derivatives also serve as formulation excipients for improving the efficiency of other gene delivery systems. This review delves into the various vector and non-vector-based gene delivery systems, their advantages, and limitations, eventually leading to the identification of cycloamylose as an ideal candidate for nucleic acid delivery. The synthesis of cationic cycloamylose is briefly discussed in each section followed by its application for specific delivery/transfection of a particular nucleic acid.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, 248007, India.
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Dehradun, 248007, India
| | - Vipul Agarwal
- Cluster for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; School of Medical and Life Sciences, Sunway University, 47500 Sunway City, Malaysia
| | - Gaurav Gupta
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharishi Dayanand University, Rohtak, 124001, India
| | - Kamal Dua
- Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
3
|
Puvvula PK, Martinez-Medina L, Cinar M, Feng L, Pisarev A, Johnson A, Bernal-Mizrachi L. A retrotransposon-derived DNA zip code internalizes myeloma cells through Clathrin-Rab5a-mediated endocytosis. Front Oncol 2024; 14:1288724. [PMID: 38463228 PMCID: PMC10920344 DOI: 10.3389/fonc.2024.1288724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/29/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction We have demonstrated that transposons derived from ctDNA can be transferred between cancer cells. The present research aimed to investigate the cellular uptake and intracellular trafficking of Multiple Myeloma-zip code (MM-ZC), a cell-specific zip code, in myeloma cell lines. We demonstrated that MM-ZC uptake by myeloma cells was concentration-, time- and cell-type-dependent. Methods Flow cytometry and confocal microscopy methods were used to identify the level of internalization of the zip codes in MM cells. To screen for the mechanism of internalization, we used multiple inhibitors of endocytosis. These experiments were followed by biotin pulldown and confocal microscopy for validation. Single interference RNA (siRNA) targeting some of the proteins involved in endocytosis was used to validate the role of this pathway in ZC cell internalization. Results Endocytosis inhibitors identified that Monensin and Chlorpromazine hydrochloride significantly reduced MM-ZC internalization. These findings suggested that Clathrin-mediated endocytosis and endosomal maturation play a crucial role in the cellular uptake of MM-ZC. Biotin pulldown and confocal microscopic studies revealed the involvement of proteins such as Clathrin, Rab5a, Syntaxin-6, and RCAS1 in facilitating the internalization of MM-ZC. Knockdown of Rab5a and Clathrin proteins reduced cellular uptake of MM-ZC and conclusively demonstrated the involvement of Clathrin-Rab5a pathways in MM-ZC endocytosis. Furthermore, both Rab5a and Clathrin reciprocally affected their association with MM-ZC when we depleted their proteins by siRNAs. Additionally, the loss of Rab5a decreased the Syntaxin-6 association with MMZC but not vice versa. Conversely, MM-ZC treatment enhanced the association between Clathrin and Rab5a. Conclusion Overall, the current study provides valuable insights into the cellular uptake and intracellular trafficking of MM-ZC in myeloma cells. Identifying these mechanisms and molecular players involved in MM-ZC uptake contributes to a better understanding of the delivery and potential applications of cell-specific Zip-Codes in gene delivery and drug targeting in cancer research.
Collapse
Affiliation(s)
| | | | - Munevver Cinar
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Lei Feng
- Kodikaz Therapeutic Solutions, New York, NY, United States
| | - Andrey Pisarev
- Kodikaz Therapeutic Solutions, New York, NY, United States
| | | | - Leon Bernal-Mizrachi
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, United States
| |
Collapse
|
4
|
Casadidio C, Hartman JEM, Mesquita B, Haegebaert R, Remaut K, Neumann M, Hak J, Censi R, Di Martino P, Hennink WE, Vermonden T. Effect of Polyplex Size on Penetration into Tumor Spheroids. Mol Pharm 2023; 20:5515-5531. [PMID: 37811785 PMCID: PMC10630948 DOI: 10.1021/acs.molpharmaceut.3c00397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 10/10/2023]
Abstract
Ovarian cancer is one of the most lethal gynecological cancers in the world. In recent years, nucleic acid (NA)-based formulations have been shown to be promising treatments for ovarian cancer, including tumor nodules. However, gene therapy is not that far advanced in clinical reality due to unfavorable physicochemical properties of the NAs, such as high molecular weight, poor cellular uptake, rapid degradation by nucleases, etc. One of the strategies used to overcome these drawbacks is the complexation of anionic NAs via electrostatic interactions with cationic polymers, resulting in the formation of so-called polyplexes. In this work, the role of the size of pDNA and siRNA polyplexes on their penetration into ovarian-cancer-based tumor spheroids was investigated. For this, a methoxypoly(ethylene glycol) poly(2-(dimethylamino)ethyl methacrylate) (mPEG-pDMAEMA) diblock copolymer was synthesized as a polymeric carrier for NA binding and condensation with either plasmid DNA (pDNA) or short interfering RNA (siRNA). When prepared in HEPES buffer (10 mM, pH 7.4) at a nitrogen/phosphate (N/P) charge ratio of 5 and pDNA polyplexes were formed with a size of 162 ± 11 nm, while siRNA-based polyplexes displayed a size of 25 ± 2 nm. The polyplexes had a slightly positive zeta potential of +7-8 mV in the same buffer. SiRNA and pDNA polyplexes were tracked in vitro into tumor spheroids, resembling in vivo avascular ovarian tumor nodules. For this purpose, reproducible spheroids were obtained by coculturing ovarian carcinoma cells with primary mouse embryonic fibroblasts in different ratios (5:2, 1:1, and 2:5). Penetration studies revealed that after 24 h of incubation, siRNA polyplexes were able to penetrate deeper into the homospheroids (composed of only cancer cells) and heterospheroids (cancer cells cocultured with fibroblasts) compared to pDNA polyplexes which were mainly located in the rim. The penetration of the polyplexes was slowed when increasing the fraction of fibroblasts present in the spheroids. Furthermore, in the presence of serum siRNA polyplexes encoding for luciferase showed a high cellular uptake in 2D cells resulting in ∼50% silencing of luciferase expression. Taken together, these findings show that self-assembled small siRNA polyplexes have good potential as a platform to test ovarian tumor nodulus penetration..
Collapse
Affiliation(s)
- Cristina Casadidio
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
- School
of Pharmacy, Drug Delivery Division, University
of Camerino, CHiP Research Center, Via Madonna delle Carceri, 62032 Camerino, Macerata, Italy
| | - Jet E. M. Hartman
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Bárbara
S. Mesquita
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Ragna Haegebaert
- Laboratory
of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical
Sciences, Ghent University, 9000 Ghent, Belgium
| | - Katrien Remaut
- Laboratory
of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical
Sciences, Ghent University, 9000 Ghent, Belgium
| | - Myriam Neumann
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Jaimie Hak
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Roberta Censi
- School
of Pharmacy, Drug Delivery Division, University
of Camerino, CHiP Research Center, Via Madonna delle Carceri, 62032 Camerino, Macerata, Italy
- Recusol
Srl, Via del Bastione
16, 62032 Camerino, Macerata, Italy
| | - Piera Di Martino
- Department
of Pharmacy, “G. D’Annunzio”
University of Chieti and Pescara, Via dei Vestini 1, 66100 Chieti, Chieti, Italy
- Recusol
Srl, Via del Bastione
16, 62032 Camerino, Macerata, Italy
| | - Wim E. Hennink
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| | - Tina Vermonden
- Department
of Pharmaceutical Sciences, Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS),
Utrecht University 99, 3508 TB Utrecht, The Netherlands
| |
Collapse
|
5
|
Chastagnier L, Marquette C, Petiot E. In situ transient transfection of 3D cell cultures and tissues, a promising tool for tissue engineering and gene therapy. Biotechnol Adv 2023; 68:108211. [PMID: 37463610 DOI: 10.1016/j.biotechadv.2023.108211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/26/2023] [Accepted: 07/09/2023] [Indexed: 07/20/2023]
Abstract
Various research fields use the transfection of mammalian cells with genetic material to induce the expression of a target transgene or gene silencing. It is a tool widely used in biological research, bioproduction, and therapy. Current transfection protocols are usually performed on 2D adherent cells or suspension cultures. The important rise of new gene therapies and regenerative medicine in the last decade raises the need for new tools to empower the in situ transfection of tissues and 3D cell cultures. This review will present novel in situ transfection methods based on a chemical or physical non-viral transfection of cells in tissues and 3D cultures, discuss the advantages and remaining gaps, and propose future developments and applications.
Collapse
Affiliation(s)
- Laura Chastagnier
- 3D Innovation Lab - 3d.FAB - ICBMS, University Claude Bernard Lyon 1, Université Lyon 1, CNRS, INSA, CPE-Lyon, UMR 5246, bat. Lederer, 5 rue Gaston Berger, 69100 Villeurbanne, France
| | - Christophe Marquette
- 3D Innovation Lab - 3d.FAB - ICBMS, University Claude Bernard Lyon 1, Université Lyon 1, CNRS, INSA, CPE-Lyon, UMR 5246, bat. Lederer, 5 rue Gaston Berger, 69100 Villeurbanne, France
| | - Emma Petiot
- 3D Innovation Lab - 3d.FAB - ICBMS, University Claude Bernard Lyon 1, Université Lyon 1, CNRS, INSA, CPE-Lyon, UMR 5246, bat. Lederer, 5 rue Gaston Berger, 69100 Villeurbanne, France.
| |
Collapse
|
6
|
Mott L, Hancock M, Grulke EA, Pack DW. Polymer/Nanoceria Hybrid Polyplexes for Gene and Antioxidant Delivery. ACS APPLIED BIO MATERIALS 2023; 6:3166-3175. [PMID: 37493016 DOI: 10.1021/acsabm.3c00295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Various diseases, including cancers and inflammatory diseases, are characterized by a disruption of redox homeostasis, suggesting the need for synergistic treatments involving co-delivery of gene therapies and free radical scavengers. In this report, polyethylenimine (PEI), nanoceria (NC), and DNA were complexed to form nanoparticles providing simultaneous delivery of a gene and an antioxidant. NC was coated in citric acid to provide stable, 4 nm particles that electrostatically bound PEI/DNA polyplexes. The resulting ternary particles transfected HeLa cells with similar efficiency to that of ternary polyplexes comprising 15 kDa poly-l-α-glutamic acid/PEI/DNA while providing smaller particle sizes by more than 100 nm. NC/PEI/DNA polyplexes exhibited enhanced radical-scavenging activity compared to free NC, and oxidative stress from the superoxide-generating agent, menadione, could be completely reversed by the delivery of NC/PEI/DNA polyplexes. Transfection by NC/PEI/DNA polyplexes was demonstrated to occur efficiently through caveolin-mediated endocytosis and macropinocytosis. Co-delivery of genes encoding reactive oxygen species-scavenging proteins, transcription factors, growth factors, tumor suppressors, or anti-inflammatory genes with NC, therefore, may be a promising strategy in synergistic therapeutics.
Collapse
Affiliation(s)
- Landon Mott
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Matthew Hancock
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Eric A Grulke
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Daniel W Pack
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40536, United States
| |
Collapse
|
7
|
Anticancer polypyrrole-polyethylenimine drug-free nanozyme for precise B-cell lymphoma therapy. Biomed Pharmacother 2023; 160:114397. [PMID: 36796279 DOI: 10.1016/j.biopha.2023.114397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/08/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
As an alternative strategy for cancer treatment, the combination of cancer nanomedicine and immunotherapy is promising with regard to efficacy and safety; however, precise modulation of the activation of antitumor immunity remains challenging. Therefore, the aim of the present study was to describe an intelligent nanocomposite polymer immunomodulator, drug-free polypyrrole-polyethyleneimine nanozyme (PPY-PEI NZ), which responds to the B-cell lymphoma tumor microenvironment, for precision cancer immunotherapy. Earlier engulfment of PPY-PEI NZs in an endocytosis-dependent manner resulted in rapid binding in four different types of B-cell lymphoma cells. The PPY-PEI NZ effectively suppressed B cell colony-like growth in vitro accompanied by cytotoxicity via apoptosis induction. During PPY-PEI NZ-induced cell death, mitochondrial swelling, loss of mitochondrial transmembrane potential (MTP), downregulation of antiapoptotic proteins, and caspase-dependent apoptosis were observed. Deregulated AKT and ERK signaling contributed to glycogen synthase kinase-3-regulated cell apoptosis following deregulation of Mcl-1 and MTP loss. Additionally, PPY-PEI NZs induced lysosomal membrane permeabilization while inhibiting endosomal acidification, partly protecting cells from lysosomal apoptosis. PPY-PEI NZs selectively bound and eliminated exogenous malignant B cells in a mixed culture system with healthy leukocytes ex vivo. While PPY-PEI NZs showed no cytotoxicity in wild-type mice, they provided long-term and efficient inhibition of the growth of B-cell lymphoma-driven nodules in a subcutaneous xenograft model. This study explores a potential PPY-PEI NZ-based anticancer agent against B-cell lymphoma.
Collapse
|
8
|
Wang X, Wu DH, Senyo SE. mRNA therapy for myocardial infarction: A review of targets and delivery vehicles. Front Bioeng Biotechnol 2022; 10:1037051. [PMID: 36507276 PMCID: PMC9732118 DOI: 10.3389/fbioe.2022.1037051] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/11/2022] [Indexed: 11/27/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death in the world. This is partly due to the low regenerative capacity of adult hearts. mRNA therapy is a promising approach under development for cardiac diseases. In mRNA therapy, expression of the target protein is modulated by delivering synthetic mRNA. mRNA therapy benefits cardiac regeneration by increasing cardiomyocyte proliferation, reducing fibrosis, and promoting angiogenesis. Because mRNA is translated in the cytoplasm, the delivery efficiency of mRNA into the cytoplasm and nucleus significantly affects its therapeutic efficacy. To improve delivery efficiency, non-viral vehicles such as lipid nanoparticles have been developed. Non-viral vehicles can protect mRNA from enzymatic degradation and facilitate the cellular internalization of mRNA. In addition to non-viral vehicles, viral vectors have been designed to deliver mRNA templates into cardiac cells. This article reviews lipid nanoparticles, polymer nanoparticles, and viral vectors that have been utilized to deliver mRNA into the heart. Because of the growing interest in lipid nanoparticles, recent advances in lipid nanoparticles designed for cardiac mRNA delivery are discussed. Besides, potential targets of mRNA therapy for myocardial infarction are discussed. Gene therapies that have been investigated in patients with cardiac diseases are analyzed. Reviewing mRNA therapy from a clinically relevant perspective can reveal needs for future investigations.
Collapse
Affiliation(s)
- Xinming Wang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Douglas H. Wu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Samuel E. Senyo
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
9
|
Kaur J, Gulati M, Corrie L, Awasthi A, Jha NK, Chellappan DK, Gupta G, MacLoughlin R, Oliver BG, Dua K, Singh SK. Role of nucleic acid-based polymeric micelles in treating lung diseases. Nanomedicine (Lond) 2022; 17:1951-1960. [PMID: 36606499 DOI: 10.2217/nnm-2022-0260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The prevalence of lung diseases is increasing year by year and existing drug therapies only provide symptomatic relief rather than targeting the actual cause. Nucleic acids can be used as an alternative therapeutic approach owing to their potential to reform a homeostatic balance by upregulating protective genes or downregulating damaging genes. However, their inherent properties, such as poor stability, ineffective cellular uptake, negative charge and so on, hinder their clinical utility. Such limitations can be overcome by exploiting the functional chemistry of polymeric micelles (PMs) for site-specific delivery, transfection efficiency and improved stability. With this objective, the present work describes the advancements made in designing nucleic acid-based PMs for treating lung diseases followed by approaches requiring consideration for clinical applications.
Collapse
Affiliation(s)
- Jaskiran Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India.,Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Leander Corrie
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Ankit Awasthi
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Plot No.32-34, Knowledge Park III, Greater Noida, Uttar Pradesh, 201310, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, 57000, Malaysia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India.,Department of Pharmacology, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, India.,Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ronan MacLoughlin
- Research and Development, Science & Emerging Technologies, Aerogen Limited, Galway Business Park, Galway, H91 HE94, Ireland.,School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Leinster, D02 YN77, Ireland.,School of Pharmacy & Pharmaceutical Sciences, Trinity College, Dublin, Leinster, D02 PN40, Ireland
| | - Brian G Oliver
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia.,Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Kamal Dua
- Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| |
Collapse
|
10
|
Ponti F, Bono N, Russo L, Bigini P, Mantovani D, Candiani G. Vibropolyfection: coupling polymer-mediated gene delivery to mechanical stimulation to enhance transfection of adherent cells. J Nanobiotechnology 2022; 20:363. [PMID: 35933375 PMCID: PMC9356458 DOI: 10.1186/s12951-022-01571-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/22/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND With the success of recent non-viral gene delivery-based COVID-19 vaccines, nanovectors have gained some public acceptance and come to the forefront of advanced therapies. Unfortunately, the relatively low ability of the vectors to overcome cellular barriers adversely affects their effectiveness. Scientists have thus been striving to develop ever more effective gene delivery vectors, but the results are still far from satisfactory. Therefore, developing novel strategies is probably the only way forward to bring about genuine change. Herein, we devise a brand-new gene delivery strategy to boost dramatically the transfection efficiency of two gold standard nucleic acid (NA)/polymer nanoparticles (polyplexes) in vitro. RESULTS We conceived a device to generate milli-to-nanoscale vibrational cues as a function of the frequency set, and deliver vertical uniaxial displacements to adherent cells in culture. A short-lived high-frequency vibrational load (t = 5 min, f = 1,000 Hz) caused abrupt and extensive plasmalemma outgrowths but was safe for cells as neither cell proliferation rate nor viability was affected. Cells took about 1 hr to revert to quasi-naïve morphology through plasma membrane remodeling. In turn, this eventually triggered the mechano-activated clathrin-mediated endocytic pathway and made cells more apt to internalize polyplexes, resulting in transfection efficiencies increased from 10-to-100-fold. Noteworthy, these results were obtained transfecting three cell lines and hard-to-transfect primary cells. CONCLUSIONS In this work, we focus on a new technology to enhance the intracellular delivery of NAs and improve the transfection efficiency of non-viral vectors through priming adherent cells with a short vibrational stimulation. This study paves the way for capitalizing on physical cell stimulation(s) to significantly raise the effectiveness of gene delivery vectors in vitro and ex vivo.
Collapse
Affiliation(s)
- Federica Ponti
- genT_LΛB, Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy
- Laboratory for Biomaterials and Bioengineering, CRC Tier I, Department of Min-Met-Mat Engineering and CHU de Québec Research Center, Division of Regenerative Medicine, Laval University, Quebec, QC, Canada
| | - Nina Bono
- genT_LΛB, Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy
| | - Luca Russo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Milan, Italy
| | - Paolo Bigini
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Milan, Italy
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, CRC Tier I, Department of Min-Met-Mat Engineering and CHU de Québec Research Center, Division of Regenerative Medicine, Laval University, Quebec, QC, Canada
| | - Gabriele Candiani
- genT_LΛB, Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy.
| |
Collapse
|
11
|
Street STG, He Y, Harniman RL, Garcia-Hernandez JD, Manners I. Precision polymer nanofibers with a responsive polyelectrolyte corona designed as a modular, functionalizable nanomedicine platform. Polym Chem 2022. [DOI: 10.1039/d2py00152g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We describe the development of a modular, functionalizable platform for biocompatible core-shell block copolymer nanofibers of controlled length (22 nm – 1.3 μm) and low dispersity produced via living crystallization-driven...
Collapse
|
12
|
Marschall ALJ. Targeting the Inside of Cells with Biologicals: Chemicals as a Delivery Strategy. BioDrugs 2021; 35:643-671. [PMID: 34705260 PMCID: PMC8548996 DOI: 10.1007/s40259-021-00500-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Delivering macromolecules into the cytosol or nucleus is possible in vitro for DNA, RNA and proteins, but translation for clinical use has been limited. Therapeutic delivery of macromolecules into cells requires overcoming substantially higher barriers compared to the use of small molecule drugs or proteins in the extracellular space. Breakthroughs like DNA delivery for approved gene therapies and RNA delivery for silencing of genes (patisiran, ONPATTRO®, Alnylam Pharmaceuticals, Cambridge, MA, USA) or for vaccination such as the RNA-based coronavirus disease 2019 (COVID-19) vaccines demonstrated the feasibility of using macromolecules inside cells for therapy. Chemical carriers are part of the reason why these novel RNA-based therapeutics possess sufficient efficacy for their clinical application. A clear advantage of synthetic chemicals as carriers for macromolecule delivery is their favourable properties with respect to production and storage compared to more bioinspired vehicles like viral vectors or more complex drugs like cellular therapies. If biologicals can be applied to intracellular targets, the druggable space is substantially broadened by circumventing the limited utility of small molecules for blocking protein–protein interactions and the limitation of protein-based drugs to the extracellular space. An in depth understanding of the macromolecular cargo types, carrier types and the cell biology of delivery is crucial for optimal application and further development of biologicals inside cells. Basic mechanistic principles of the molecular and cell biological aspects of cytosolic/nuclear delivery of macromolecules, with particular consideration of protein delivery, are reviewed here. The efficiency of macromolecule delivery and applications in research and therapy are highlighted.
Collapse
Affiliation(s)
- Andrea L J Marschall
- Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Brunswick, Germany.
| |
Collapse
|
13
|
Monnery BD. Polycation-Mediated Transfection: Mechanisms of Internalization and Intracellular Trafficking. Biomacromolecules 2021; 22:4060-4083. [PMID: 34498457 DOI: 10.1021/acs.biomac.1c00697] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Polyplex-mediated gene transfection is now in its' fourth decade of serious research, but the promise of polyplex-mediated gene therapy has yet to fully materialize. Only approximately one in a million applied plasmids actually expresses. A large part of this is due to an incomplete understanding of the mechanism of polyplex transfection. There is an assumption that internalization must follow a canonical mechanism of receptor mediated endocytosis. Herein, we present arguments that untargeted (and most targeted) polyplexes do not utilize these routes. By incorporating knowledge of syndecan-polyplex interactions, we can show that syndecans are the "target" for polyplexes. Further, it is known that free polycations (which disrupt cell-membranes by acid-catalyzed hydrolysis of phospholipid esters) are necessary for (untargeted) endocytosis. This can be incorporated into the model to produce a novel mechanism of endocytosis, which fits the observed phenomenology. After membrane translocation, polyplex containing vesicles reach the endosome after diffusing through the actin mesh below the cell membrane. From there, they are acidified and trafficked toward the lysosome. Some polyplexes are capable of escaping the endosome and unpacking, while others are not. Herein, it is argued that for some polycations, as acidification proceeds the polyplexes excluding free polycations, which disrupt the endosomal membrane by acid-catalyzed hydrolysis, allowing the polyplex to escape. The polyplex's internal charge ratio is now insufficient for stability and it releases plasmids which diffuse to the nucleus. A small proportion of these plasmids diffuse through the nuclear pore complex (NPC), with aggregation being the major cause of loss. Those plasmids that have diffused through the NPC will also aggregate, and this appears to be the reason such a small proportion of nuclear plasmids express mRNA. Thus, the structural features which promote unpacking in the endosome and allow for endosomal escape can be determined, and better polycations can be designed.
Collapse
Affiliation(s)
- Bryn D Monnery
- Department of Organic and (Bio)Polymer Chemistry, Hasselt University, Building F, Agoralaan 1, B-3590 Diepenbeek, Belgium
| |
Collapse
|
14
|
Bera H, Abosheasha MA, Ito Y, Ueda M. Etherified pullulan-polyethylenimine based nanoscaffolds improved chemosensitivity of erlotinib on hypoxic cancer cells. Carbohydr Polym 2021; 271:118441. [PMID: 34364579 DOI: 10.1016/j.carbpol.2021.118441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/18/2021] [Accepted: 07/12/2021] [Indexed: 12/21/2022]
Abstract
The current research endeavor aimed to accomplish hypoxia-responsive polyethyleneimine-conjugated carboxymethyl pullulan-based co-polymer (CMP-HA-NI-PEI-NBA) bearing nitroaromatic subunits to efficiently deliver erlotinib (ERL) to reverse its hypoxia-induced resistance in cancer cells. As compared to a control co-polymer (CMP-HA-MI-PEI-BA) devoid of hypoxia-sensitive moieties, this scaffold demonstrated a hypochromic shift in the UV spectra and rapid dismantling of its self-assembled architecture upon exposure to simulated hypoxic condition. The hypoxia-responsive co-polymer encapsulated ERL with desirable loading capacity (DEE, 63.05 ± 2.59%), causing attenuated drug crystallinity. The drug release rate of the scaffold under reducing condition was faster relative to that of non-reducing environment. Their cellular uptake occurred through an energy-dependent endocytic process, which could exploit its caveolae/lipid raft-mediated internalization pathway. The ERL-loaded scaffolds more efficiently induced apoptosis and suppressed the proliferation of drug-resistant hypoxic HeLa cells than the pristine ERL. Hence, this study presented a promising drug delivery nanoplatform to overcome hypoxia-evoked ERL resistance.
Collapse
Affiliation(s)
- Hriday Bera
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| | - Mohammed A Abosheasha
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo 192-0397, Japan
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo 192-0397, Japan; Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science (CEMS), 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Motoki Ueda
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science (CEMS), 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| |
Collapse
|
15
|
Ward DM, Shodeinde AB, Peppas NA. Innovations in Biomaterial Design toward Successful RNA Interference Therapy for Cancer Treatment. Adv Healthc Mater 2021; 10:e2100350. [PMID: 33973393 PMCID: PMC8273125 DOI: 10.1002/adhm.202100350] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/27/2021] [Indexed: 12/11/2022]
Abstract
Gene regulation using RNA interference (RNAi) therapy has been developed as one of the frontiers in cancer treatment. The ability to tailor the expression of genes by delivering synthetic oligonucleotides to tumor cells has transformed the way scientists think about treating cancer. However, its clinical application has been limited due to the need to deliver synthetic RNAi oligonucleotides efficiently and effectively to target cells. Advances in nanotechnology and biomaterials have begun to address the limitations to RNAi therapeutic delivery, increasing the likelihood of RNAi therapeutics for cancer treatment in clinical settings. Herein, innovations in the design of nanocarriers for the delivery of oligonucleotides for successful RNAi therapy are discussed.
Collapse
Affiliation(s)
- Deidra M Ward
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
| | - Aaliyah B Shodeinde
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
| | - Nicholas A Peppas
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave. Stop A1900, Austin, TX, 78712, USA
- Department of Pediatrics and Department of Surgery and Perioperative Care, Dell Medical School, 1601 Trinity St., Bldg. B, Stop Z0800, Austin, TX, 78712, USA
| |
Collapse
|
16
|
Mastrobattista E. Formulation and delivery solutions for the next generation biotherapeutics. J Control Release 2021; 336:583-597. [PMID: 34174354 DOI: 10.1016/j.jconrel.2021.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 11/16/2022]
Abstract
In 2018 I was appointed full professor of Pharmaceutical Biotechnology & Delivery at the Pharmaceutics division of the department of Pharmaceutical Sciences at Utrecht University, The Netherlands. In this contribution to the Orations - New Horizons of the Journal of Controlled Release I will introduce my research group (see also www.uu.nl/pharmaceutics) and will highlight my current and future research projects. In coming years the focus of my research will be on the administration of biotherapeutics, aiming to control their fate from the site of injection to the site of action. I will discuss issues related to formulation of biotherapeutics into nanomedicines (NMs), intracellular delivery of nucleic acids as well as protein therapeutics, and targeted delivery of biotherapeutics beyond the liver. In addition, I will provide a forward view on how current developments in the drug delivery and gene therapy field may result in sustainable and cost-effective dosing regimens for biotherapeutics.
Collapse
Affiliation(s)
- Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
17
|
Franck CO, Fanslau L, Bistrovic Popov A, Tyagi P, Fruk L. Biopolymer-based Carriers for DNA Vaccine Design. Angew Chem Int Ed Engl 2021; 60:13225-13243. [PMID: 32893932 PMCID: PMC8247987 DOI: 10.1002/anie.202010282] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Indexed: 12/16/2022]
Abstract
Over the last 30 years, genetically engineered DNA has been tested as novel vaccination strategy against various diseases, including human immunodeficiency virus (HIV), hepatitis B, several parasites, and cancers. However, the clinical breakthrough of the technique is confined by the low transfection efficacy and immunogenicity of the employed vaccines. Therefore, carrier materials were designed to prevent the rapid degradation and systemic clearance of DNA in the body. In this context, biopolymers are a particularly promising DNA vaccine carrier platform due to their beneficial biochemical and physical characteristics, including biocompatibility, stability, and low toxicity. This article reviews the applications, fabrication, and modification of biopolymers as carrier medium for genetic vaccines.
Collapse
Affiliation(s)
- Christoph O. Franck
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| | - Luise Fanslau
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| | - Andrea Bistrovic Popov
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| | - Puneet Tyagi
- Dosage Form Design and DevelopmentBioPharmaceuticals DevelopmentR&DAstra ZenecaGaithersburgMD20878USA
| | - Ljiljana Fruk
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgePhillipa Fawcett DriveCambridgeCB3 0ASUK
| |
Collapse
|
18
|
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: 196] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
19
|
Nicolle L, Casper J, Willimann M, Journot CMA, Detampel P, Einfalt T, Grisch-Chan HM, Thöny B, Gerber-Lemaire S, Huwyler J. Development of Covalent Chitosan-Polyethylenimine Derivatives as Gene Delivery Vehicle: Synthesis, Characterization, and Evaluation. Int J Mol Sci 2021; 22:ijms22083828. [PMID: 33917124 PMCID: PMC8067803 DOI: 10.3390/ijms22083828] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 01/03/2023] Open
Abstract
There is an increasing interest in cationic polymers as important constituents of non-viral gene delivery vectors. In the present study, we developed a versatile synthetic route for the production of covalent polymeric conjugates consisting of water-soluble depolymerized chitosan (dCS; MW 6–9 kDa) and low molecular weight polyethylenimine (PEI; 2.5 kDa linear, 1.8 kDa branched). dCS-PEI derivatives were evaluated based on their physicochemical properties, including purity, covalent bonding, solubility in aqueous media, ability for DNA condensation, and colloidal stability of the resulting polyplexes. They were complexed with non-integrating DNA vectors coding for reporter genes by simple admixing and assessed in vitro using liver-derived HuH-7 cells for their transfection efficiency and cytotoxicity. Using a rational screening cascade, a lead compound was selected (dCS-Suc-LPEI-14) displaying the best balance of biocompatibility, cytotoxicity, and transfection efficiency. Scale-up and in vivo evaluation in wild-type mice allowed for a direct comparison with a commercially available non-viral delivery vector (in vivo-jetPEI). Hepatic expression of the reporter gene luciferase resulted in liver-specific bioluminescence, upon intrabiliary infusion of the chitosan-based polyplexes, which exceeded the signal of the in vivo jetPEI reference formulation by a factor of 10. We conclude that the novel chitosan-derivative dCS-Suc-LPEI-14 shows promise and potential as an efficient polymeric conjugate for non-viral in vivo gene therapy.
Collapse
Affiliation(s)
- Laura Nicolle
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering Ecole Polytechnique Fédérale de Lausanne, EPFL SB ISIC SCI-SB-SG, Station 6, CH-1015 Lausanne, Switzerland; (L.N.); (C.M.A.J.)
| | - Jens Casper
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland; (J.C.); (P.D.); (T.E.)
| | - Melanie Willimann
- Division of Metabolism and Children’s Research Center, University Children’s Hospital Zürich, CH-8032 Zürich, Switzerland; (M.W.); (H.M.G.-C.); (B.T.)
| | - Céline M. A. Journot
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering Ecole Polytechnique Fédérale de Lausanne, EPFL SB ISIC SCI-SB-SG, Station 6, CH-1015 Lausanne, Switzerland; (L.N.); (C.M.A.J.)
| | - Pascal Detampel
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland; (J.C.); (P.D.); (T.E.)
| | - Tomaž Einfalt
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland; (J.C.); (P.D.); (T.E.)
| | - Hiu Man Grisch-Chan
- Division of Metabolism and Children’s Research Center, University Children’s Hospital Zürich, CH-8032 Zürich, Switzerland; (M.W.); (H.M.G.-C.); (B.T.)
| | - Beat Thöny
- Division of Metabolism and Children’s Research Center, University Children’s Hospital Zürich, CH-8032 Zürich, Switzerland; (M.W.); (H.M.G.-C.); (B.T.)
| | - Sandrine Gerber-Lemaire
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering Ecole Polytechnique Fédérale de Lausanne, EPFL SB ISIC SCI-SB-SG, Station 6, CH-1015 Lausanne, Switzerland; (L.N.); (C.M.A.J.)
- Correspondence: (S.G.-L.); (J.H.); Tel.: +41-21-693-93-72 (S.G.-L.); +41-61-207-15-13 (J.H.)
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland; (J.C.); (P.D.); (T.E.)
- Correspondence: (S.G.-L.); (J.H.); Tel.: +41-21-693-93-72 (S.G.-L.); +41-61-207-15-13 (J.H.)
| |
Collapse
|
20
|
Zeng K, Ma L, Yang W, Lei S, Wang M, You Y, Zhao Y, Ge X. Biodegradable nano-organosilica gene carrier for high-efficiency gene transfection. J Mater Chem B 2021; 8:2483-2494. [PMID: 32110782 DOI: 10.1039/d0tb00108b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Finding and exploiting safe and high-efficiency gene carriers have always been critical tasks for gene therapy. In this work, novel GSH-triggered degradable organosilica nanoparticles grafted with guanidinated-fluorinated α-polylysine (o-SiNP-GF) are prepared to be studied as gene carriers. The organosilica matrix of o-SiNP-GF is synthesized through the hydrolysis and condensation of 1,2-bis(triethoxysilyl)ethane (BTSE) and bis[3-(triethoxysilyl)propyl]tetrasulfide (BTSPTS). The o-SiNP-GF nanoparticles have a size of about 20 nm. They possess a positive zeta potential of 42 mV in PBS (pH 7.4) and can be disintegrated in the presence of GSH. The cytotoxicity and DNA-binding ability of o-SiNP-GF, as well as in vitro gene transfection performance of DNA/o-SiNP-GF complexes, have been investigated using enhanced green fluorescent protein plasmid (pEGFP) as the DNA model. MTT assay shows that the cytotoxicity of o-SiNP-GF is very low even at a concentration up to 800 μg mL-1. The o-SiNP-GF nanoparticles can effectively bind to pEGFP through a complex coacervation method. The in vitro transfection efficiency of pEGFP/o-SiNP-GF complexes in 293T cells is up to 94.7% at the N/P ratio of 10, much higher than that of pEGFP/PEI complexes. Luciferase gene and fibroblast growth factor (FGF2) gene are also used as the DNA models to study the in vivo gene transfection performance of the o-SiNP-GF carrier by bioluminescence imaging and the evaluation of the healing rate of a mouse wound, respectively. Compared with naked DNA and DNA/PEI complexes, DNA/o-SiNP-GF complexes show much higher in vivo transfection efficiency. This work not only provides a way to prepare novel GSH-triggered degradable organosilica nanoparticles of size less than 50 nm, but also proves that the modification of guanidinated-fluorinated α-polylysine is an effective method to improve the efficiency of gene carriers.
Collapse
Affiliation(s)
- Kun Zeng
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Li Ma
- Department of Plastic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Wenxiu Yang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Shan Lei
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Mozhen Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Yezi You
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Yu Zhao
- Department of Plastic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Xuewu Ge
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China.
| |
Collapse
|
21
|
Franck CO, Fanslau L, Bistrovic Popov A, Tyagi P, Fruk L. Biopolymer‐based Carriers for DNA Vaccine Design. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202010282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Christoph O. Franck
- Department of Chemical Engineering and Biotechnology University of Cambridge Phillipa Fawcett Drive Cambridge CB3 0AS UK
| | - Luise Fanslau
- Department of Chemical Engineering and Biotechnology University of Cambridge Phillipa Fawcett Drive Cambridge CB3 0AS UK
| | - Andrea Bistrovic Popov
- Department of Chemical Engineering and Biotechnology University of Cambridge Phillipa Fawcett Drive Cambridge CB3 0AS UK
| | - Puneet Tyagi
- Dosage Form Design and Development BioPharmaceuticals Development R&D Astra Zeneca Gaithersburg MD 20878 USA
| | - Ljiljana Fruk
- Department of Chemical Engineering and Biotechnology University of Cambridge Phillipa Fawcett Drive Cambridge CB3 0AS UK
| |
Collapse
|
22
|
Tuning the Surface Charge of Self-Assembled Polydiacetylene Vesicles to Control Aggregation and Cell Binding. BIOSENSORS-BASEL 2020; 10:bios10100132. [PMID: 32987658 PMCID: PMC7598607 DOI: 10.3390/bios10100132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
Polydiacetylene vesicles of various compositions were assembled using a two-part mixture of 10,12-pentacosadiynoic acid (PCDA) and ethylenedioxy-bis-ethylamine (EDEA)-labeled PCDA in order to control surface charge and stability within a desired pH range. Investigation of the interaction of the vesicles with mammalian cells as a function of surface charge was carried out and identified a clear correlation in cell–vesicle association and corresponding cell death for vesicles with positive surface charge. The binding behavior of the vesicles was found to be tunable by regulating the proportion of anionic PCDA relative to cationic PCDA–EDEA content within vesicles as to control the surface charge as a function of pH. Association of vesicles with cells thus depended on the corresponding charge of the vesicles and cell surface. The prospect of this work may serve as a step toward future vesicle designs to allow triggered uptake of vesicles locally within low pH tumor microenvironments.
Collapse
|
23
|
Hu LZ, Wang W, Xu JL, Jia YY, Huan ML, Li C, Zhou SY, Zhang BL. Polyethylenimine-based nanovector grafted with mannitol moieties to achieve effective gene delivery and transfection. NANOTECHNOLOGY 2020; 31:325101. [PMID: 32325436 DOI: 10.1088/1361-6528/ab8c76] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Polyethylenimine (PEI), a kind of cationic non-viral gene delivery vector, is capable of stable and efficient transgene expression for gene delivery. However, low transfection efficiency in vivo along with high toxicity limited the further application of gene therapy in the clinic. To enhance gene transfection performance and reduce cytotoxicity of polyethylenimine, branched polyethylenimine-derived cationic polymers BPEI25 k-man-S/L/M/H with different grafting degree with mannitol moieties were prepared and the transfection efficiency was evaluated. Among them, BPEI25 k-man-L showed the best transfection efficiency, lower toxicity, and significantly enhanced long-term systemic transgene expression for 96 h in vivo even at a single-dose administration. The results of cellular uptake mechanism and western-blot experiments revealed that the mannitol modification of BPEI25 k induced and up-regulated the phosphorylation of caveolin-1 and thus enhanced the caveolae-mediated cellular uptake. This class of gene delivery system highlights a paradigmatic approach for the development of novel and safe non-viral vectors for gene therapy.
Collapse
Affiliation(s)
- Li-Zhong Hu
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, People's Republic of China. Department of Pharmacy, Luoyang Polytechnic, Luoyang 471000, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Khalifehzadeh R, Arami H. Biodegradable calcium phosphate nanoparticles for cancer therapy. Adv Colloid Interface Sci 2020; 279:102157. [PMID: 32330734 PMCID: PMC7261203 DOI: 10.1016/j.cis.2020.102157] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 12/16/2022]
Abstract
Calcium phosphate is the inorganic mineral of hard tissues such as bone and teeth. Due to their similarities to the natural bone, calcium phosphates are highly biocompatible and biodegradable materials that have found numerous applications in dental and orthopedic implants and bone tissue engineering. In the form of nanoparticles, calcium phosphate nanoparticles (CaP's) can also be used as effective delivery vehicles to transfer therapeutic agents such as nucleic acids, drugs, proteins and enzymes into tumor cells. In addition, facile preparation and functionalization of CaP's, together with their inherent properties such as pH-dependent solubility provide advantages in delivery and release of these bioactive agents using CaP's as nanocarriers. In this review, the challenges and achievements in the intracellular delivery of these agents to tumor cells are discussed. Also, the most important issues in the design and potential applications of CaP-based biominerals are addressed with more focus on their biodegradability in tumor microenvironment.
Collapse
Affiliation(s)
- Razieh Khalifehzadeh
- Department of Chemical Engineering, Stanford University, Shriram Center, 443 Via Ortega, Stanford, California 94305, United States; Department of Radiology, Stanford University School of Medicine, James H. Clark Center, 318 Campus Drive, E-153, Stanford, California 94305, United States
| | - Hamed Arami
- Department of Radiology, Stanford University School of Medicine, James H. Clark Center, 318 Campus Drive, E-153, Stanford, California 94305, United States; Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, James H. Clark Center, 318 Campus Drive, E-153, Stanford, California 94305, United States.
| |
Collapse
|
25
|
Chien CS, Wang CY, Yang YP, Chou SJ, Ko YL, Tsai FT, Yu WC, Chang CC, Cherng JY, Yang MY. Using cationic polyurethane-short branch PEI as microRNA-driven nano-delivery system for stem cell differentiation. J Chin Med Assoc 2020; 83:367-370. [PMID: 32101899 DOI: 10.1097/jcma.0000000000000272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Non-viral gene delivery, such as using biodegradable polyurethane short-branch polyethylenimine (PU-PEI), has been considered a potentially safer gene delivery system in comparison to conventional virus systems. METHODS The polycationization of DNA complexes protects DNA from nuclease degradation, and these DNA complexes are nanoscale in size to enter the cell through endocytosis. RESULTS Due to the net positive surface charge of the cell, these polyplexes efficiently bind to the cell through electrostatic interactions with negatively charged membrane components. Cationic PU-PEI has been shown to be non-cytotoxic and has a high transfection efficiency, making it a practical gene delivery material in diseases. CONCLUSION We developed a PU-PEI nanomedicine-based platform to efficiently deliver microRNA in promoting differentiation capacity of stem cells, especially on induced pluripotent stem cells.
Collapse
Affiliation(s)
- Chian-Shiu Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Chien-Ying Wang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Trauma, Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Shih-Jie Chou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Yu-Ling Ko
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Fu-Ting Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Wen-Chung Yu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Chia-Ching Chang
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao Tung University, Hsinchu, Taiwan, ROC
- Department of Materials Science and Engineering, National Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Jong-Yuh Cherng
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan, ROC
| | - Meng-Yin Yang
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- Department of Neurosurgery, Da-Li Jan-Ai Hospital, Taichung, Taiwan, ROC
- Department of Surgery/Neurosurgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
- College of Nursing, Central Taiwan University of Science and Technology, Taichung, Taiwan, ROC
| |
Collapse
|
26
|
Desai AS, Hunter MR, Kapustin AN. Using macropinocytosis for intracellular delivery of therapeutic nucleic acids to tumour cells. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180156. [PMID: 30967005 DOI: 10.1098/rstb.2018.0156] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Nucleic acids are a rapidly emerging therapeutic modality with the potential to become the third major drug modality alongside antibodies and small molecules. Owing to the unfavourable physico-chemical characteristics of nucleic acids, such as large size and negative charge, intracellular delivery remains a fundamental challenge to realizing this potential. Delivery technologies such as lipids, polymers and peptides have been used to facilitate delivery, with many of the most successful technologies using macropinocytosis to gain cellular entry; mostly by default rather than design. Fundamental knowledge of macropinocytosis is rapidly growing, presenting opportunities to better tailor design strategies to target this pathway. Furthermore, certain types of tumour cells have been observed to have high levels of macropinocytic activity and traffic cargo to favourable destinations within the cell for endosomal release, providing unique opportunities to further use this entry route for drug delivery. In this article, we review the delivery systems reported to be taken up by macropinocytosis and what is known about the mechanisms for regulating macropinocytosis in tumour cells. From this analysis, we identify new opportunities for exploiting this pathway for the intracellular delivery of nucleic acids to tumour cells. This article is part of the Theo Murphy meeting issue 'Macropinocytosis'.
Collapse
Affiliation(s)
- Arpan S Desai
- AstraZeneca, IMED Biotech Unit, Pharmaceutical Sciences , Aaron Klug Building, Granta Park, Cambridge CB21 6GH , UK
| | - Morag R Hunter
- AstraZeneca, IMED Biotech Unit, Pharmaceutical Sciences , Aaron Klug Building, Granta Park, Cambridge CB21 6GH , UK
| | - Alexander N Kapustin
- AstraZeneca, IMED Biotech Unit, Pharmaceutical Sciences , Aaron Klug Building, Granta Park, Cambridge CB21 6GH , UK
| |
Collapse
|
27
|
Wang CY, Yang YP, Liu CY, Lu KH, Liu YY, Wu WW, Chen KH, Chang YL, Lee SD, Lin HC. Nanoparticle-delivery system enhanced the improvement and recovery in toxicity-induced acute hepatic failure. J Chin Med Assoc 2019; 82:363-367. [PMID: 31058711 DOI: 10.1097/jcma.0000000000000066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND The major curative remedy for advanced liver failure is hepatic transplantation. However, the conventional medicine still shows the limitations and obstacles for liver regeneration. Importantly, it is unclear whether we can get a rapid and high efficacy platform to facilitate to reprogram hepatic capability. The main work of this study was to develop a platform for a nanomedicine-based gene-delivery platform of novel nanoparticles (NNPs) to efficiently facilitate the liver function recovery. METHODS In this study, we studied the feasibility and efficiency of NNP and produced the multiple abilities of NNPs for a potential platform of gene transduction. We showed that NNPs played an important role in hepatic protection. The cytoprotective effects of NNPs in toxic-hepatic cells were investigated and evaluated by cell viability, reactive oxygen species production, in vitro cell abilities, and in vivo animal studies. RESULTS We demonstrated that NNPs possess the abilities to protect the cell after toxic-stress both in vitro and in vivo. Under the stress condition, our result showed that cell viabilities can be improved by NNP-carried hepatocyte nuclear factor 3 (HNF3) gene (NNP-HNF3), which is a famous hepatic transcriptional factor and regenerative marker to modulate essential molecular pathways activating various hepatic-specific markers. Importantly, compared to control and NNP-control, NNP-HNF3 exhibited the cytoprotective effects that prevented toxic-induced oxidative stress and cell damage in vitro as well as in vivo. Notably, our data showed that NNP-HNF3 treatment may improve toxic-induced hepatic encephalopathy. CONCLUSION Herein, we demonstrated that novel nanoparticle, such as NNP-HNF3, serves as a key regulator for protecting the damaged hepatic cell and the bioproduct-based source for the new therapeutics of hepatic failure.
Collapse
Affiliation(s)
- Chien-Ying Wang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Trauma, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Ping Yang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Neurological Surgery, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chao-Yu Liu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Thoracic Surgery, Far-Eastern Memorial Hospital, New Taipei City, Taiwan, ROC
- Department of Surgery, Far-Eastern Memorial Hospital, New Taipei City, Taiwan, ROC
| | - Kai-Hsi Lu
- Department of Medical Research and Education, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Yung-Yang Liu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Chest, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Wai-Wah Wu
- Division of Gastroenterology, Department of Internal Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Kuan-Hsuan Chen
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yuh-Lih Chang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Shou-Dong Lee
- Division of Gastroenterology, Department of Internal Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
- Department of Medical Research and Education, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Hsin-Chi Lin
- Division of Gastroenterology, Department of Internal Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
- Department of Medical Research and Education, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
28
|
Chiang CW, Chuang EY. Biofunctional core-shell polypyrrole-polyethylenimine nanocomplex for a locally sustained photothermal with reactive oxygen species enhanced therapeutic effect against lung cancer. Int J Nanomedicine 2019; 14:1575-1585. [PMID: 30880966 PMCID: PMC6400129 DOI: 10.2147/ijn.s163299] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Polymeric delivery systems have been elucidated over the last few years as an approach of achieving high therapeutic effect to the local site of malignant disease patients who have cancer. Polypyrrole (Ppy) is a potential organic conducting polymer which has long been recognized as a versatile material due to its excellent stability, conductive properties, and great absorbance in the range of near-infrared (NIR). It is tremendously versatile for use in various biomedical fields such as cancer therapy. NIR irradiation-activated treatment platform technologies are now being considered to be novel and exciting options in potential nanomedicine. However, the realistic photothermal use of Ppy-applied nanomaterials is yet in its early phase, and there are a few disadvantages of Ppy, such as its water insolubility. In the clinic, the common approach for treatment of lung cancer is the delivery of therapeutic active substances through intratumoral administration. Nevertheless, the tumor uptake, regional retention, mechanism of treatment, and tissue organ penetration regarding the developed strategy of this nanomaterial with photothermal hyperthermia are important issues for exerting effective cancer therapy. MATERIALS AND METHODS In this study, we developed a cationic Ppy-polyethylenimine nanocomplex (NC) with photothermal hyperthermia to study its physicochemical characteristics, including size distribution, zeta potential, and transmission electron microscopy, scanning electron microscopy, and Fourier transform infrared morphology. We also examined the cellular uptake effect on lung cancer cells, the photothermal properties, intracellularly generated reactive oxygen species (ROS), and cytotoxicity. RESULTS The results suggested that this nanocarrier system was able to effectively attach onto lung cancer cells for subsequent endocytosis. The NCs taken up were able to absorb NIR and then converted the NIR light into local hyperthermia with its intracellular photothermal performance to provide local hyperthermic treatment. This regionally generated hyperthermia also induced ROS formation and improved the killing of lung cancer cells as a promising local photothermal therapy. CONCLUSION This development of a nanocarrier would bring a novel therapeutic strategy for lung cancer in the future.
Collapse
Affiliation(s)
- Chih-Wei Chiang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
- Bone and Joint Research Center, Department of Orthopedics, Taipei Medical University Hospital, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University and International Ph.D. Program in Biomedical Engineering College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan,
| |
Collapse
|
29
|
Yang Y, Wang X, Hu X, Kawazoe N, Yang Y, Chen G. Influence of Cell Morphology on Mesenchymal Stem Cell Transfection. ACS APPLIED MATERIALS & INTERFACES 2019; 11:1932-1941. [PMID: 30571082 DOI: 10.1021/acsami.8b20490] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Gene transfection has broad applications in bioengineering and biomedical fields. Although many gene carrier materials and transfection methods have been developed, it remains unclear how cell morphology including cell spreading and elongation affects gene transfection. In this study, human bone marrow-derived mesenchymal stem cells (hMSCs) were cultured on micropatterns and transfected with cationic pAcGFP1-N1 plasmid complexes. The relationship between the cell morphology of hMSCs and gene transfection was investigated using micropatterning techniques. Spreading and elongation of hMSCs were precisely controlled by micropatterned surfaces. The results showed that well-spread and elongated hMSCs had high transfection efficiency. Analysis of the uptake of exogenous genes and DNA synthesis activity indicated that the well-spread and elongated cell morphology promoted gene transfection through enhanced uptake of the cationic complexes and accelerated DNA synthesis. The results should provide useful information for understanding of cell morphology on gene transfection and development of efficient gene transfection methods.
Collapse
Affiliation(s)
- Yingjun Yang
- Research Center for Functional Materials , National Institute for Materials Science , 1-1 Namiki , Tsukuba , Ibaraki 305-0044 , Japan
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences , University of Tsukuba , 1-1-1 Tennodai , Tsukuba , Ibaraki 305-8577 , Japan
| | - Xinlong Wang
- Research Center for Functional Materials , National Institute for Materials Science , 1-1 Namiki , Tsukuba , Ibaraki 305-0044 , Japan
| | - Xiaohong Hu
- Graduate School of Life and Environmental Science , University of Tsukuba , 1-1-1 Tennodai , Tsukuba , Ibaraki 305-8571 , Japan
| | - Naoki Kawazoe
- Research Center for Functional Materials , National Institute for Materials Science , 1-1 Namiki , Tsukuba , Ibaraki 305-0044 , Japan
| | - Yingnan Yang
- Graduate School of Life and Environmental Science , University of Tsukuba , 1-1-1 Tennodai , Tsukuba , Ibaraki 305-8571 , Japan
| | - Guoping Chen
- Research Center for Functional Materials , National Institute for Materials Science , 1-1 Namiki , Tsukuba , Ibaraki 305-0044 , Japan
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences , University of Tsukuba , 1-1-1 Tennodai , Tsukuba , Ibaraki 305-8577 , Japan
| |
Collapse
|
30
|
Tiwari S, Bahadur P. Modified hyaluronic acid based materials for biomedical applications. Int J Biol Macromol 2019; 121:556-571. [DOI: 10.1016/j.ijbiomac.2018.10.049] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/02/2018] [Accepted: 10/11/2018] [Indexed: 12/22/2022]
|
31
|
Liechty WB, Scheuerle RL, Vela Ramirez JE, Peppas NA. Uptake and function of membrane-destabilizing cationic nanogels for intracellular drug delivery. Bioeng Transl Med 2019; 4:17-29. [PMID: 30680315 PMCID: PMC6336667 DOI: 10.1002/btm2.10120] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/12/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023] Open
Abstract
The design of intracellular drug delivery vehicles demands an in-depth understanding of their internalization and function upon entering the cell to tailor the physicochemical characteristics of these platforms and achieve efficacious treatments. Polymeric cationic systems have been broadly accepted to be membrane disruptive thus being beneficial for drug delivery inside the cell. However, if excessive destabilization takes place, it can lead to adverse effects. One of the strategies used to modulate the cationic charge is the incorporation of hydrophobic moieties, thus increasing the hydrophobic content. We have demonstrated the successful synthesis of nanogels based on diethylaminoethyl methacrylate and poly(ethylene glycol) methyl ether methacrylate. Addition of the hydrophobic monomers tert-butyl methacrylate or 2-(tert-butylamino)ethyl methacrylate shows improved polymer hydrophobicity and modulation of the critical swelling pH. Here, we evaluate the cytocompatibility, uptake, and function of these membrane-destabilizing cationic methacrylated nanogels using in vitro models. The obtained results suggest that the incorporation of hydrophobic monomers decreases the cytotoxicity of the nanogels to epithelial colorectal adenocarcinoma cells. Furthermore, analysis of the internalization pathways of these vehicles using inhibitors and imaging flow cytometry showed a significant decrease in uptake when macropinocytosis/phagocytosis inhibitors were present. The membrane-disruptive abilities of the cationic polymeric nanogels were confirmed using three different models. They demonstrated to cause hemolysis in sheep erythrocytes, lactate dehydrogenase leakage from a model cell line, and disrupt giant unilamellar vesicles. These findings provide new insights of the potential of polymeric nanoformulations for intracellular delivery.
Collapse
Affiliation(s)
- William B. Liechty
- McKetta Dept. of Chemical EngineeringThe University of Texas at AustinAustinTX 78712
| | - Rebekah L. Scheuerle
- McKetta Dept. of Chemical EngineeringThe University of Texas at AustinAustinTX 78712
| | - Julia E. Vela Ramirez
- McKetta Dept. of Chemical EngineeringThe University of Texas at AustinAustinTX 78712
- Dept. of Biomedical EngineeringThe University of Texas at AustinAustinTX 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative MedicineThe University of Texas at AustinAustinTX 78712
| | - Nicholas A. Peppas
- McKetta Dept. of Chemical EngineeringThe University of Texas at AustinAustinTX 78712
- Dept. of Biomedical EngineeringThe University of Texas at AustinAustinTX 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative MedicineThe University of Texas at AustinAustinTX 78712
- Depts. of Surgery and Perioperative CareDell Medical School, The University of Texas at AustinAustinTX 78712
- Division of Molecular Pharmaceutics and Drug DeliveryCollege of Pharmacy, The University of Texas at AustinAustinTX 78712
| |
Collapse
|
32
|
Zhou D, Zeng M, Gao Y, Sigen A, Lyu J, Wang W. Advanced Polymers for Nonviral Gene Delivery. NUCLEIC ACID NANOTHERANOSTICS 2019:311-364. [DOI: 10.1016/b978-0-12-814470-1.00010-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
33
|
Protein moiety in oligochitosan modified vector regulates internalization mechanism and gene delivery: Polyplex characterization, intracellular trafficking and transfection. Carbohydr Polym 2018; 202:143-156. [PMID: 30286987 DOI: 10.1016/j.carbpol.2018.08.131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/13/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022]
Abstract
Oligochitosan-modified proteins have gained attention as efficient non-viral vectors for gene delivery. However, little information exists if protein moieties can serve as an important role for internalization and endosome escape ability of the genetic material. To explore this issue, we designed two cationic oligochitosan-modified vectors that consist of different proteins, namely a hydrophobic plant protein (zein) and a hydrophilic animal protein (ovalbumin (OVA)) to deliver pDNA to epithelial cell line CHO-K1 and HEK 293 T. These cationic vectors were systematically characterized by molecular weight, infrared (IR) structural analysis, transmission electron microscopy (TEM) morphology, and surface charge. A remarkable impact of protein moieties was observed on physiochemical properties of the developed vectors. Oligochitosan-modified zein containing hydrophobic protein exhibited high buffering capacity and excellent DNA binding ability compared to the oligochitosan-modified OVA. The data on transfection in the presence of endocytic inhibitors indicated that the caveolae-mediated pathway (CvME) played a key role in the internalization of the zein-based polyplex. However, the OVA-based polyplex was internalized in CHO-K1 cells via CvME and in HEK 293 T cells via the lipid-mediated pathway. Moreover, oligochitosan-modified zein exhibited lower cytotoxicity, greater lysosomal escape ability, better plasmid stability, and better transfection efficiency than the oligochitosan-modified OVA. This study offers a facile procedure for the synthesis of cationic vectors and elucidates the relationship that exists between protein moieties and transfection activity, thus providing an alternative, non-viral platform for the gene delivery.
Collapse
|
34
|
Bitoque DB, Rosa da Costa AM, Silva GA. Insights on the intracellular trafficking of PDMAEMA gene therapy vectors. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 93:277-288. [PMID: 30274059 DOI: 10.1016/j.msec.2018.07.071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 11/29/2022]
Abstract
It is known that an efficient gene therapy vector must overcome several steps to be able to express the gene of interest: (I) enter the cell by crossing the cell membrane; (II) escape the endo-lysosomal degradation pathway; (III) release the genetic material; (IV) traffic through the cytoplasm and enter the nucleus; and last (V), enable gene expression to synthetize the protein of interest. In recent years, we and others have demonstrated the potential of poly(2‑(N,N'‑dimethylamino)ethylmethacrylate) (PDMAEMA) as a gene therapy vehicle. Further optimization of gene transfer efficiency requires the understanding of the intracellular pathway of PDMAEMA. Therefore the goal of this study was to determine the cellular entry and intracellular trafficking mechanisms of our PDMAEMA vectors and determine the gene transfer bottleneck. For this, we have produced rhodamine-labeled PDMAEMA polyplexes that were used to transfect retinal cells and the cellular localization determined by co-localization with cellular markers. Our vectors quickly and efficiently cross the cell membrane, and escape the endo-lysosomal system by 24 h. We have observed the PDMAEMA vectors to concentrate around the nucleus, and the DNA load to be released in the first 24 h after transfection. These results allow us to conclude that although the endo-lysosomal system is an important obstacle, PDMAEMA gene vectors can overcome it. The nuclear membrane, however, constitutes the bottleneck to PDMAEMA gene transfer ability.
Collapse
Affiliation(s)
- Diogo B Bitoque
- ProRegeM PhD Program, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal; Algarve Chemistry Research Centre (CIQA), University of Algarve, 8005-139 Faro, Portugal; CEDOC - Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal
| | - Ana M Rosa da Costa
- Department of Chemistry and Pharmacy, University of Algarve, Faro, Portugal; Algarve Chemistry Research Centre (CIQA), University of Algarve, 8005-139 Faro, Portugal
| | - Gabriela A Silva
- CEDOC - Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal.
| |
Collapse
|
35
|
Galbiati E, Avvakumova S, La Rocca A, Pozzi M, Messali S, Magnaghi P, Colombo M, Prosperi D, Tortora P. A fast and straightforward procedure for vault nanoparticle purification and the characterization of its endocytic uptake. Biochim Biophys Acta Gen Subj 2018; 1862:2254-2260. [PMID: 30036602 DOI: 10.1016/j.bbagen.2018.07.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/12/2018] [Accepted: 07/17/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUND Vaults are eukaryotic ribonucleoprotein particles composed of up 78 copies of the 97 kDa major vault protein that assembles into a barrel-like, "nanocapsule" enclosing poly(ADP-ribose) polymerase, telomerase-associated protein-1 and small untranslated RNAs. Overall, the molecular mass of vault particles amounts to about 13 MDa. Although it has been implicated in several cellular functions, its physiological roles remain poorly understood. Also, the possibility to exploit it as a nanovector for drug delivery is currently being explored in several laboratories. METHODS Using the baculovirus expression system, vaults were expressed and purified by a dialysis step using a 1 MDa molecular weight cutoff membrane and a subsequent size exclusion chromatography. Purity was assessed by SDS-PAGE, transmission electron microscopy and dynamic light scattering. Particle's endocytic uptake was monitored by flow cytometry and confocal microscopy. RESULTS The purification protocol here reported is far simpler and faster than those currently available and lead to the production of authentic vault. We then demonstrated its clathrin-mediated endocytic uptake by normal fibroblast and glioblastoma, but not carcinoma cell lines. In contrast, no significant caveolin-mediated endocytosis was detected. CONCLUSIONS These results provide the first evidence for an intrinsic propensity of the vault complex to undergo endocytic uptake cultured eukaryotic cells. GENERAL SIGNIFICANCE The newly developed purification procedure will greatly facilitate any investigation based on the use of the vault particle as a natural nanocarrier. Its clathrin-mediated endocytic uptake observed in normal and in some tumor cell lines sheds light on its physiological role.
Collapse
Affiliation(s)
- Elisabetta Galbiati
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy
| | - Svetlana Avvakumova
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy
| | - Alessandra La Rocca
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy
| | - Maria Pozzi
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy
| | - Silvia Messali
- Oncology, Nerviano Medical Sciences, Viale Pasteur 10, Milano, 20014, Nerviano, Italy
| | - Paola Magnaghi
- Oncology, Nerviano Medical Sciences, Viale Pasteur 10, Milano, 20014, Nerviano, Italy
| | - Miriam Colombo
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy
| | - Davide Prosperi
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy
| | - Paolo Tortora
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy.
| |
Collapse
|
36
|
Mielańczyk A, Kupczak M, Burek M, Mielańczyk Ł, Klymenko O, Wandzik I, Neugebauer D. Functional (mikto)stars and star-comb copolymers from d-gluconolactone derivative: An efficient route for tuning the architecture and responsiveness to stimuli. POLYMER 2018. [DOI: 10.1016/j.polymer.2018.05.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
37
|
Walsh DP, Murphy RD, Panarella A, Raftery RM, Cavanagh B, Simpson JC, O'Brien FJ, Heise A, Cryan SA. Bioinspired Star-Shaped Poly(l-lysine) Polypeptides: Efficient Polymeric Nanocarriers for the Delivery of DNA to Mesenchymal Stem Cells. Mol Pharm 2018; 15:1878-1891. [PMID: 29590755 DOI: 10.1021/acs.molpharmaceut.8b00044] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The field of tissue engineering is increasingly recognizing that gene therapy can be employed for modulating in vivo cellular response thereby guiding tissue regeneration. However, the field lacks a versatile and biocompatible gene delivery platform capable of efficiently delivering transgenes to mesenchymal stem cells (MSCs), a cell type often refractory to transfection. Herein, we describe the extensive and systematic exploration of three architectural variations of star-shaped poly(l-lysine) polypeptide (star-PLL) with varying number and length of poly(l-lysine) arms as potential nonviral gene delivery vectors for MSCs. We demonstrate that star-PLL vectors are capable of self-assembling with pDNA to form stable, cationic nanomedicines. Utilizing high content screening, live cell imaging, and mechanistic uptake studies we confirm the intracellular delivery of pDNA by star-PLLs to MSCs is a rapid process, which likely proceeds via a clathrin-independent mechanism. We identify a star-PLL composition with 64 poly(l-lysine) arms and five l-lysine subunits per arm as a particularly efficient vector that is capable of delivering both reporter genes and the therapeutic transgenes bone morphogenetic protein-2 and vascular endothelial growth factor to MSCs. This composition facilitated a 1000-fold increase in transgene expression in MSCs compared to its linear analogue, linear poly(l-lysine). Furthermore, it demonstrated comparable transgene expression to the widely used vector polyethylenimine using a lower pDNA dose with significantly less cytotoxicity. Overall, this study illustrates the ability of the star-PLL vectors to facilitate efficient, nontoxic nucleic acid delivery to MSCs thereby functioning as an innovative nanomedicine platform for tissue engineering applications.
Collapse
Affiliation(s)
- David P Walsh
- Drug Delivery & Advanced Materials Team, School of Pharmacy , RCSI , Dublin , Ireland.,Tissue Engineering Research Group, Department of Anatomy , RCSI , Dublin , Ireland.,Trinity Centre for Bioengineering , Trinity College Dublin (TCD) , Dublin , Ireland
| | - Robert D Murphy
- Pharmaceutical and Medicinal Chemistry , RCSI , Dublin , Ireland
| | - Angela Panarella
- School of Biology and Environmental Science , University College Dublin , Dublin , Ireland
| | - Rosanne M Raftery
- Tissue Engineering Research Group, Department of Anatomy , RCSI , Dublin , Ireland.,Trinity Centre for Bioengineering , Trinity College Dublin (TCD) , Dublin , Ireland
| | | | - Jeremy C Simpson
- School of Biology and Environmental Science , University College Dublin , Dublin , Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy , RCSI , Dublin , Ireland.,Trinity Centre for Bioengineering , Trinity College Dublin (TCD) , Dublin , Ireland
| | - Andreas Heise
- Pharmaceutical and Medicinal Chemistry , RCSI , Dublin , Ireland
| | - Sally-Ann Cryan
- Drug Delivery & Advanced Materials Team, School of Pharmacy , RCSI , Dublin , Ireland.,Tissue Engineering Research Group, Department of Anatomy , RCSI , Dublin , Ireland.,Trinity Centre for Bioengineering , Trinity College Dublin (TCD) , Dublin , Ireland
| |
Collapse
|
38
|
Vermeulen LMP, Brans T, Samal SK, Dubruel P, Demeester J, De Smedt SC, Remaut K, Braeckmans K. Endosomal Size and Membrane Leakiness Influence Proton Sponge-Based Rupture of Endosomal Vesicles. ACS NANO 2018; 12:2332-2345. [PMID: 29505236 DOI: 10.1021/acsnano.7b07583] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
In gene therapy, endosomal escape represents a major bottleneck since nanoparticles often remain entrapped inside endosomes and are trafficked toward the lysosomes for degradation. A detailed understanding of the endosomal barrier would be beneficial for developing rational strategies to improve transfection and endosomal escape. By visualizing individual endosomal escape events in live cells, we obtain insight into mechanistic factors that influence proton sponge-based endosomal escape. In a comparative study, we found that HeLa cells treated with JetPEI/pDNA polyplexes have a 3.5-fold increased endosomal escape frequency compared to ARPE-19 cells. We found that endosomal size has a major impact on the escape capacity. The smaller HeLa endosomes are more easily ruptured by the proton sponge effect than the larger ARPE-19 endosomes, a finding supported by a mathematical model based on the underlying physical principles. Still, it remains intriguing that even in the small HeLa endosomes, <10% of the polyplex-containing endosomes show endosomal escape. Further experiments revealed that the membrane of polyplex-containing endosomes becomes leaky to small compounds, preventing effective buildup of osmotic pressure, which in turn prevents endosomal rupture. Analysis of H1299 and A549 cells revealed that endosomal size determines endosomal escape efficiency when cells have comparable membrane leakiness. However, at high levels of membrane leakiness, buildup of osmotic pressure is no longer possible, regardless of endosomal size. Based on our findings that both endosomal size and membrane leakiness have a high impact on proton sponge-based endosomal rupture, we provide important clues toward further improvement of this escape strategy.
Collapse
Affiliation(s)
- Lotte M P Vermeulen
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| | - Toon Brans
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| | - Sangram K Samal
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| | | | - Jo Demeester
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| | - Katrien Remaut
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| |
Collapse
|
39
|
Ma XX, Gao H, Zhang YX, Jia YY, Li C, Zhou SY, Zhang BL. Construction and evaluation of BSA-CaP nanomaterials with enhanced transgene performance via biocorona-inspired caveolae-mediated endocytosis. NANOTECHNOLOGY 2018; 29:085101. [PMID: 29256442 DOI: 10.1088/1361-6528/aaa2b2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Non-viral nanovectors have attracted much attention owing to their ability to condense genetic materials and their ease of modification. However, their poor stability, low biocompatibility and gene degradation in endosomes or lysosomes has significantly hampered their application in vivo and in the clinic. In an attempt to overcome these difficulties a series of bovine serum albumin (BSA)-calcium phosphate (CaP) nanoparticles were constructed. The CaP condenses with DNA to form nanocomplexes coated with a biomimetic corona of BSA. Such complexes may retain the inherent endocytosis profile of BSA, with improved biocompatibility. In particular the transgene performance may be enhanced by stimulating the cellular uptake pathway via caveolae-mediated endocytosis. Two methods were employed to construct and optimize the formulation of BSA-CaP nanomaterials. The optimized BSA-CaP-50-M2 nanoparticles prepared by our second method exhibited good stability, negligible cytotoxicity and enhanced transgene performance with long-term expression for 72 h in vivo even with a single dose. Determination of the cellular uptake pathway and Western blot revealed that cellular uptake of the designed BSA-CaP-50-M2 nanoparticles was mainly via caveolae-mediated endocytosis in a non-degradative pathway in which the biomimetic uptake profile of BSA was retained.
Collapse
Affiliation(s)
- Xi-Xi Ma
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
40
|
Che J, Xue Y, Feng J, Bai G, Yuan W. Comparison of Biological Responses of Polymers Based on Imine and Disulfide Backbones for siRNA Delivery. ACS APPLIED MATERIALS & INTERFACES 2018; 10:5196-5202. [PMID: 29384351 DOI: 10.1021/acsami.7b16101] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
To achieve a successful delivery of siRNA by carriers in vivo, the degradation of polymers in response to tiny intracellular changes should be seriously considered. In addition, the balance between degradation and stability of polymers is another key point for high performance of carriers. In this study, imine and disulfide linkages, which are sensitive to pH changes and redox environment, respectively, were constructed as the main backbone of polymers to deliver siRNA at the intracellular and animal level. Comparisons were made between performances of these two different polymers. Both of the polymers synthesized here have good ability to condense siRNA. However, polyplexes formed by the imine backbone-based polymer (TPSP) showed a larger particle size and a higher zeta potential than that of the disulfide backbone-based polymer (DTDPS). Although both TPSP and DTDPS could deliver the target siRNA into 7721 cells, polyplexes formed by TPSP showed a higher silence efficiency in vitro and accomplished more accumulation in tumors. In conclusion, we believe TPSP is superior to be used for siRNA delivery and promises a potential for widespread use.
Collapse
Affiliation(s)
- Junyi Che
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University , No. 800, Dongchuan Road, Shanghai 200240, China
| | - Yonger Xue
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University , No. 800, Dongchuan Road, Shanghai 200240, China
| | - Jia Feng
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University , No. 800, Dongchuan Road, Shanghai 200240, China
| | - Guang Bai
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University , No. 800, Dongchuan Road, Shanghai 200240, China
| | - Weien Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University , No. 800, Dongchuan Road, Shanghai 200240, China
| |
Collapse
|
41
|
Maiti B, Kamra M, Karande AA, Bhattacharya S. Transfection efficiencies of α-tocopherylated cationic gemini lipids with hydroxyethyl bearing headgroups under high serum conditions. Org Biomol Chem 2018; 16:1983-1993. [DOI: 10.1039/c7ob02835k] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Liposomal gene transfection under high serum conditions.
Collapse
Affiliation(s)
- Bappa Maiti
- Department of Organic Chemistry
- Indian Institute of Science
- Bangalore 560012
- India
- Director's Research Unit
| | - Mohini Kamra
- Department of Organic Chemistry
- Indian Institute of Science
- Bangalore 560012
- India
| | - Anjali A. Karande
- Department of Biochemistry
- Indian Institute of Science
- Bangalore 560012
- India
| | - Santanu Bhattacharya
- Department of Organic Chemistry
- Indian Institute of Science
- Bangalore 560012
- India
- Director's Research Unit
| |
Collapse
|
42
|
Bus T, Traeger A, Schubert US. The great escape: how cationic polyplexes overcome the endosomal barrier. J Mater Chem B 2018; 6:6904-6918. [DOI: 10.1039/c8tb00967h] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Endo-lysosomal escape strategies of cationic polymer-mediated gene delivery at a glance.
Collapse
Affiliation(s)
- Tanja Bus
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC)
- Friedrich Schiller University Jena
- 07743 Jena
- Germany
- Jena Center for Soft Matter (JCSM)
| | - Anja Traeger
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC)
- Friedrich Schiller University Jena
- 07743 Jena
- Germany
- Jena Center for Soft Matter (JCSM)
| | - Ulrich S. Schubert
- Laboratory of Organic Chemistry and Macromolecular Chemistry (IOMC)
- Friedrich Schiller University Jena
- 07743 Jena
- Germany
- Jena Center for Soft Matter (JCSM)
| |
Collapse
|
43
|
Selection and Identification of Skeletal-Muscle-Targeted RNA Aptamers. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 10:199-214. [PMID: 29499933 PMCID: PMC5862129 DOI: 10.1016/j.omtn.2017.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/06/2017] [Accepted: 12/06/2017] [Indexed: 01/16/2023]
Abstract
Oligonucleotide gene therapy has shown great promise for the treatment of muscular dystrophies. Nevertheless, the selective delivery to affected muscles has shown to be challenging because of their high representation in the body and the high complexity of their cell membranes. Current trials show loss of therapeutic molecules to non-target tissues leading to lower target efficacy. Therefore, strategies that increase uptake efficiency would be particularly compelling. To address this need, we applied a cell-internalization SELEX (Systematic Evolution of Ligands by Exponential Enrichment) approach and identified a skeletal muscle-specific RNA aptamer. A01B RNA aptamer preferentially internalizes in skeletal muscle cells and exhibits decreased affinity for off-target cells. Moreover, this in vitro selected aptamer retained its functionality in vivo, suggesting a potential new approach for targeting skeletal muscles. Ultimately, this will aid in the development of targeted oligonucleotide therapies against muscular dystrophies.
Collapse
|
44
|
Santo D, Cordeiro RA, Sousa A, Serra A, Coelho JF, Faneca H. Combination of Poly[(2-dimethylamino)ethyl methacrylate] and Poly(β-amino ester) Results in a Strong and Synergistic Transfection Activity. Biomacromolecules 2017; 18:3331-3342. [DOI: 10.1021/acs.biomac.7b00983] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Daniela Santo
- Center
for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department
of Life Sciences, Faculty of Science and Technology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Rosemeyre A. Cordeiro
- Center
for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ana Sousa
- Center
for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department
of Life Sciences, Faculty of Science and Technology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Arménio Serra
- CEMMPRE,
Department of Chemical Engineering, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Jorge F.J. Coelho
- CEMMPRE,
Department of Chemical Engineering, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Henrique Faneca
- Center
for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department
of Life Sciences, Faculty of Science and Technology, University of Coimbra, 3004-517 Coimbra, Portugal
| |
Collapse
|
45
|
Homann S, Hofmann C, Gorin AM, Nguyen HCX, Huynh D, Hamid P, Maithel N, Yacoubian V, Mu W, Kossyvakis A, Sen Roy S, Yang OO, Kelesidis T. A novel rapid and reproducible flow cytometric method for optimization of transfection efficiency in cells. PLoS One 2017; 12:e0182941. [PMID: 28863132 PMCID: PMC5580984 DOI: 10.1371/journal.pone.0182941] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 07/19/2017] [Indexed: 01/10/2023] Open
Abstract
Transfection is one of the most frequently used techniques in molecular biology that is also applicable for gene therapy studies in humans. One of the biggest challenges to investigate the protein function and interaction in gene therapy studies is to have reliable monospecific detection reagents, particularly antibodies, for all human gene products. Thus, a reliable method that can optimize transfection efficiency based on not only expression of the target protein of interest but also the uptake of the nucleic acid plasmid, can be an important tool in molecular biology. Here, we present a simple, rapid and robust flow cytometric method that can be used as a tool to optimize transfection efficiency at the single cell level while overcoming limitations of prior established methods that quantify transfection efficiency. By using optimized ratios of transfection reagent and a nucleic acid (DNA or RNA) vector directly labeled with a fluorochrome, this method can be used as a tool to simultaneously quantify cellular toxicity of different transfection reagents, the amount of nucleic acid plasmid that cells have taken up during transfection as well as the amount of the encoded expressed protein. Finally, we demonstrate that this method is reproducible, can be standardized and can reliably and rapidly quantify transfection efficiency, reducing assay costs and increasing throughput while increasing data robustness.
Collapse
Affiliation(s)
- Stefanie Homann
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, United States of America
| | - Christian Hofmann
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, United States of America
| | - Aleksandr M. Gorin
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, United States of America
| | - Huy Cong Xuan Nguyen
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, United States of America
| | - Diana Huynh
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, United States of America
| | - Phillip Hamid
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, United States of America
| | - Neil Maithel
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, United States of America
| | - Vahe Yacoubian
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, United States of America
| | - Wenli Mu
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, United States of America
| | - Athanasios Kossyvakis
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, United States of America
| | - Shubhendu Sen Roy
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, United States of America
| | - Otto Orlean Yang
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, United States of America
| | - Theodoros Kelesidis
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, United States of America
| |
Collapse
|
46
|
Liu G, Fang Z, Yuan M, Li W, Yang Y, Jiang M, Ouyang Y, Yuan W. Biodegradable Carriers for Delivery of VEGF Plasmid DNA for the Treatment of Critical Limb Ischemia. Front Pharmacol 2017; 8:528. [PMID: 28848442 PMCID: PMC5552722 DOI: 10.3389/fphar.2017.00528] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 07/26/2017] [Indexed: 01/06/2023] Open
Abstract
The safe and efficient delivery of therapeutic nucleic acid is a prerequisite for an effective DNA therapy. In this study, we condensed the low molecular weight polyethylenimine (PEI, 1.8k Da) with 2,6-pyridinedicarboxaldehyde (PDA), both of which are degradable in vivo, to synthesize a biodegradable polycationic material (PDAPEI) to deliver vascular endothelial growth factor (VEGF) plasmid DNA (pDNA). Particle size and zeta potential of this novel degradable PEI derivatives-pDNA nanoparticle were investigated and in vitro cytotoxicity was estimated on human umbilical vein endothelial cells (HUVECs). Using pDNA-encoding VEGF-A and green fluorescence protein (GFP), we also checked transfection efficiency of the vector (PDAPEI) and found its excellent performance at 40 w/w ratio. We successfully established peripheral ischemia animal model on C57/BL6J mice to evaluate the therapeutic effect of PDAPEI/pVEGF-A polyplex system on ischemic disease and a conclusion was made that PDAPEI is a promising gene vector in the treatment of peripheral ischemic artery disease (PAD).
Collapse
Affiliation(s)
- Guang Liu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Zhiwei Fang
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghai, China
| | - Minglu Yuan
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghai, China
| | - Weimin Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Yunqi Yang
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghai, China
| | - Mier Jiang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Yuanming Ouyang
- Shanghai Sixth People's Hospital, Shanghai University of Medicine and HealthShanghai, China
| | - Weien Yuan
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghai, China
| |
Collapse
|
47
|
Gu J, Chen X, Fang X, Sha X. Retro-inverso d-peptide-modified hyaluronic acid/bioreducible hyperbranched poly(amido amine)/pDNA core-shell ternary nanoparticles for the dual-targeted delivery of short hairpin RNA-encoding plasmids. Acta Biomater 2017; 57:156-169. [PMID: 28442415 DOI: 10.1016/j.actbio.2017.04.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 03/29/2017] [Accepted: 04/20/2017] [Indexed: 01/08/2023]
Abstract
The active targeting of gene carriers is a powerful strategy for improving tumour-specific delivery and therapy. Although numerous l-peptide ligands play significant roles in the active targeting of nanomedicine, retro-inverso d-peptides have been explored as targeting ligands due to their superior stability and bioactivity in vivo. In this study, retro-inverso d-peptide (RIF7)-modified hyaluronic acid (HA)/bioreducible hyperbranched poly(amido amine) (RHB)/plasmid DNA (pDNA) ternary nanoparticles were successfully developed using the layer-by-layer method for the CD44-positive tumour-specific delivery of short hairpin RNA (shRNA)-encoding pDNA through the combination of the Anxa1 (tumour vasculature) and CD44 (tumour cell-surface) receptors, which mediated the dual targeting. The potential of these newly designed nanoparticles was evaluated by examining the efficacy of their cellular uptake and transfection in cell monolayers, tumour spheroids, and malignant xenograft animal models. With negligible cytotoxicity, the spherical-shaped RIF7-HA/RHB/pDNA nanoparticles were the direct result of an electrostatic complex that had efficiently targeted CD44-positive tumour delivery, penetration, and cellular uptake in vitro. The nanoparticles showed excellent target-specific gene transfection even in the presence of serum. The in vivo therapeutic effect of RIF7-HA/RHB/pDNA-shRNA nanoparticle-mediated shRNA targeting of the Cyclin gene (shCyclin) was evaluated in tumour-bearing mice. The RIF7-HA/RHB/pDNA-shCyclin nanoparticles significantly increased the survival time of tumour-bearing mice and substantially reduced tumour growth due to their extremely specific tumour-targeting activity. These results suggested that the combination of HA and retro-inverso peptide RIF7 significantly increased the therapeutic effect of pDNA-shCyclin-loaded nanoparticles for CD44-positive tumours. Thus, RIF7-HA-mediated multi-target ternary gene vectors are an efficient and promising strategy for the delivery of pDNA-shRNA in the targeted treatment of malignant and metastatic cancers. STATEMENT OF SIGNIFICANCE Although l-peptide ligands play significant roles in the active targeting of nanomedicine, retro-inverso d-peptides have been explored as targeting ligands due to their superior stability and bioactivity in vivo. Retro-inverso peptide RIF7 was designed as a ligand of Anxa1 receptor. The resultant peptide, RIF7, displayed high binding efficiency within Anxa1 receptor, which is highly expressed tumour vasculature cells and some tumour cells such as B16F10 and U87MG cells. The most important feature of RIF7 is its high stability in the blood, which is suitable and promising for application in vivo. Multifunctional RIF7-HA was then synthesized by conjugating the RIF7 peptide to HA, which was used to modify the surface of RHB/pDNA nanoparticles to prepare RIF7-HA/RHB/pDNA core-shell ternary nanoparticles for the dual-targeted delivery of shRNA-encoding plasmids in vitro and in vivo.
Collapse
Affiliation(s)
- Jijin Gu
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China; Laboratory for Drug Delivery and Biomaterials, Faculty of Pharmacy, University of Manitoba, 750 McDermot Ave, Winnipeg, Manitoba R3E 0T5, Canada
| | - Xinyi Chen
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Xiaoling Fang
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Xianyi Sha
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China.
| |
Collapse
|
48
|
Sun Y, Lu J, Yan D, Shen L, Hu H, Chen D. Cellular uptake mechanism and clearance kinetics of fluorescence-labeled glycyrrhetinic acid and glycyrrhetinic acid-modified liposome in hepatocellular carcinoma cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 53:46-56. [PMID: 28501784 DOI: 10.1016/j.etap.2017.05.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 04/12/2017] [Accepted: 05/04/2017] [Indexed: 06/07/2023]
Abstract
Glycyrrhetinic acid (GA) is a natural pentacyclic triterpene derivative that exerts significant effects in the suppression of liver cancer. The receptors of GA on liver cells and hepatocellular carcinoma (HCC) cells have drawn broad attention. The effects of GA might depend on its transport into and out of cells. However, the question has not been previously addressed despite its obvious and fundamental importance. In this paper, GA and GA-modified liposome (GA-Lip) were labeled with fluorescein isothiocyanate (FITC) or coumarin 6 (Cou6) using chemical or pharmaceutical techniques. The transport courses of FITC-GA and GA-Cou6-Lip were studied in HepG2 cells in vitro. We found that the fluorescence labeled GA and GA-Lip uptake and clearance were time-dependent. FITC-GA uptake involved passive diffusion and active transport, and the receptors were in the cytomembrane proteins. GA-Cou6-Lip uptake was mediated by caveolae-dependent endocytosis. In addition, FITC-GA and GA-Cou6-Lip clearance of the HCC cells fitted exponential decay and second-order processes, respectively. These findings provide new insights into the anti-HCC actions of GA.
Collapse
Affiliation(s)
- Yuqi Sun
- School of Pharmacy, Jinzhou Medical University, Jinzhou, PR China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Jinghua Lu
- School of Pharmacy, Jinzhou Medical University, Jinzhou, PR China
| | - Dongxue Yan
- School of Pharmacy, Jinzhou Medical University, Jinzhou, PR China
| | - Liping Shen
- School of Pharmacy, Jinzhou Medical University, Jinzhou, PR China
| | - Haiyang Hu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Dawei Chen
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China.
| |
Collapse
|
49
|
Liu JN, Bu W, Shi J. Chemical Design and Synthesis of Functionalized Probes for Imaging and Treating Tumor Hypoxia. Chem Rev 2017; 117:6160-6224. [DOI: 10.1021/acs.chemrev.6b00525] [Citation(s) in RCA: 556] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jia-nan Liu
- State
Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, P.R. China
| | - Wenbo Bu
- State
Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, P.R. China
- Shanghai
Key Laboratory of Green Chemistry and Chemical Processes, School of
Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, P.R. China
| | - Jianlin Shi
- State
Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, P.R. China
| |
Collapse
|
50
|
Hill EK, Li J. Current and future prospects for nanotechnology in animal production. J Anim Sci Biotechnol 2017; 8:26. [PMID: 28316783 PMCID: PMC5351054 DOI: 10.1186/s40104-017-0157-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 03/02/2017] [Indexed: 11/10/2022] Open
Abstract
Nanoparticles have been used as diagnostic and therapeutic agents in the human medical field for quite some time, though their application in veterinary medicine and animal production is still relatively new. Recently, production demands on the livestock industry have been centered around the use of antibiotics as growth promoters due to growing concern over microbial antibiotic resistance. With many countries reporting increased incidences of antibiotic-resistant bacteria, laws and regulations are being updated to end in-feed antibiotic use in the animal production industry. This sets the need for suitable alternatives to be established for inclusion in feed. Many reports have shown evidence that nanoparticles may be good candidates for animal growth promotion and antimicrobials. The current status and advancements of nanotechnological applications in animal production will be the focus of this review and the emerging roles of nanoparticles for nutrient delivery, biocidal agents, and tools in veterinary medicine and reproduction will be discussed. Additionally, influences on meat, egg, and milk quality will be reviewed.
Collapse
Affiliation(s)
- Emily K Hill
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong China.,Department of Animal Biosciences, University of Guelph, 50 Stone Road East, Building #70, Guelph, ON N1G 2 W1 Canada
| | - Julang Li
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong China.,Department of Animal Biosciences, University of Guelph, 50 Stone Road East, Building #70, Guelph, ON N1G 2 W1 Canada
| |
Collapse
|