1
|
Greitens C, Leroux JC, Burger M. The intracellular visualization of exogenous DNA in fluorescence microscopy. Drug Deliv Transl Res 2024; 14:2242-2261. [PMID: 38526634 PMCID: PMC11208204 DOI: 10.1007/s13346-024-01563-4] [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] [Accepted: 02/28/2024] [Indexed: 03/27/2024]
Abstract
In the development of non-viral gene delivery vectors, it is essential to reliably localize and quantify transfected DNA inside the cell. To track DNA, fluorescence microscopy methods are commonly applied. These mostly rely on fluorescently labeled DNA, DNA binding proteins fused to a fluorescent protein, or fluorescence in situ hybridization (FISH). In addition, co-stainings are often used to determine the colocalization of the DNA in specific cellular compartments, such as the endolysosomes or the nucleus. We provide an overview of these DNA tracking methods, advice on how they should be combined, and indicate which co-stainings or additional methods are required to draw precise conclusions from a DNA tracking experiment. Some emphasis is given to the localization of exogenous DNA inside the nucleus, which is the last step of DNA delivery. We argue that suitable tools which allow for the nuclear detection of faint signals are still missing, hampering the rational development of more efficient non-viral transfection systems.
Collapse
Affiliation(s)
- Christina Greitens
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland.
| | - Michael Burger
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093, Zurich, Switzerland.
| |
Collapse
|
2
|
Sokolova V, Loza K, Ebel JF, Buer J, Westendorf AM, Epple M. Barium sulphate microparticles are taken up by three different cell types: HeLa, THP-1, and hMSC. Acta Biomater 2023; 164:577-587. [PMID: 37019167 DOI: 10.1016/j.actbio.2023.03.043] [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: 12/11/2022] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023]
Abstract
Cytotoxicity and cellular uptake of spherical barium sulphate microparticles (diameter 1 µm) were studied with three different cell lines, i.e. THP-1 cells (monocytes; model for a phagocytosing cell line), HeLa cells (epithelial cells; model for a non-phagocytosing cell line), and human mesenchymal stem cells (hMSCs; model for non-phagocytosing primary cells). Barium sulphate is a chemically and biologically inert solid which allows to distinguish two different processes, e.g. the particle uptake and potential adverse biological reactions. Barium sulphate microparticles were surface-coated by carboxymethylcellulose (CMC) which gave the particles a negative charge. Fluorescence was added by conjugating 6-aminofluorescein to CMC. The cytotoxicity of these microparticles was studied by the MTT test and a live/dead assay. The uptake was visualized by confocal laser scanning microscopy (CLSM) and scanning electron microscopy (SEM). The particle uptake mechanism was quantified by flow cytometry with different endocytosis inhibitors in THP-1 and HeLa cells. The microparticles were easily taken up by all cell types, mostly by phagocytosis and micropinocytosis, within a few hours. STATEMENT OF SIGNIFICANCE: The interaction of particles and cells is of primary importance in nanomedicine, drug delivery, and nanotoxicology. It is commonly assumed that cells take up only nanoparticles unless they are able to phagocytosis. Here, we demonstrate with chemically and biologically inert microparticles of barium sulphate that even non-phagocytosing cells like HeLa and hMSCs take up microparticles to a considerable degree. This has considerable implication in biomaterials science, e.g. in case of abrasive debris and particulate degradation products from implants like endoprostheses.
Collapse
Affiliation(s)
- V Sokolova
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitätsstr. 5-7, 45117 Essen, Germany.
| | - K Loza
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitätsstr. 5-7, 45117 Essen, Germany.
| | - J F Ebel
- Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany.
| | - J Buer
- Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany.
| | - A M Westendorf
- Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany.
| | - M Epple
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitätsstr. 5-7, 45117 Essen, Germany.
| |
Collapse
|
3
|
Li Q, Chen X, Lin W, Guo X, Ma Y. Application of a Novel Multicomponent Nanoemulsion to Tumor Therapy Based on the Theory of “Unification of Drugs and Excipients”. Pharm Dev Technol 2023; 28:351-362. [PMID: 36971746 DOI: 10.1080/10837450.2023.2196330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Toad skin has many pharmacological activities and bufadienolides are regarded as its main anti-tumor components. The poor water solubility, high toxicity, rapid elimination and less selectivity in vivo of bufadienolides limit the application of toad skin. Based on the "unification of drugs and excipients" theory, the toad skin extracts (TSE) and Brucea javanica oil (BJO) nanoemulsions (NEs) were designed to solve the aforementioned problems. BJO as the main oil phase was not only used to prepare the NEs, but played a synergistic therapeutic role combined with TSE. TSE-BJO NEs showed 155 nm particle size, entrapment efficiency of >95% and good stability. TSE-BJO NEs demonstrated superior anti-tumor activity compared with the TSE or BJO NEs alone. The mechanism of TSE-BJO NEs to enhance the antineoplastic efficacy involved several pathways, such as inhibiting cell proliferation, inducing tumor cell apoptosis >40% and arresting cell cycle at G2/M. TSE-BJO NEs could co-deliver drugs into the target cells efficiently and exhibit satisfying synergism. Besides, TSE-BJO NEs facilitated the long circulation of bufadienolides contributing to the high accumulation of drugs at tumor sites and the improvement of anti-tumor efficacy. The study achieves the combinative administration of the toxic TSE and BJO with high efficacy and safety.
Collapse
|
4
|
Chang SM, Yu CY, Chen YF. Mechanism of endosomal escape by pH-responsive nucleic-acid vectors. Phys Rev E 2022; 106:034408. [PMID: 36266809 DOI: 10.1103/physreve.106.034408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/08/2022] [Indexed: 06/16/2023]
Abstract
Successful intracellular delivery of nucleic acids (NAs) hinges on many factors, one of them being NAs' efficacious escape from endosomes. As competent NA vectors, pH-responsive gemini surfactants (GSs) might achieve high efficacy by facilitating endosomal escape. However, how the GSs assist the escape remains debated as many proposed mechanisms still lack experimental support, which hinders replication and further improvement of the efficient delivery. Here, via UV, fluorescence spectroscopy, and small-angle neutron scattering (SANS), we examined a pH-responsive GS's and a pH-unresponsive GS's capabilities to compact DNA and withstand binding competition, and their interactions with model endosomal and lysosomal membranes, at varied pHs. Acidification-driven enhancement of DNA-compaction capability and of stability against binding competition were found specific to the pH-responsive GS. Alongside the pH-responsive GS's structural perturbation to the membranes as observed with SANS, the features suggest that pH-responsive GSs facilitate endosomal escape by releasing excess GS molecules from DNA-GS complexes upon acidification in endosome maturation, with the released GS molecules disrupting endosomal and lysosomal membranes and thereby assisting the escape. A general design principle for NA vectors is proposed on the basis of this experimental finding.
Collapse
Affiliation(s)
- Shih-Min Chang
- Department of Chemical and Materials Engineering, National Central University, Taoyuan 32001, Taiwan
| | - Chia Ying Yu
- Department of Chemical and Materials Engineering, National Central University, Taoyuan 32001, Taiwan
| | - Yi-Fan Chen
- Department of Chemical and Materials Engineering, National Central University, Taoyuan 32001, Taiwan
| |
Collapse
|
5
|
Light triggered nanoscale biolistics for efficient intracellular delivery of functional macromolecules in mammalian cells. Nat Commun 2022; 13:1996. [PMID: 35422038 PMCID: PMC9010410 DOI: 10.1038/s41467-022-29713-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 03/22/2022] [Indexed: 11/17/2022] Open
Abstract
Biolistic intracellular delivery of functional macromolecules makes use of dense microparticles which are ballistically fired onto cells with a pressurized gun. While it has been used to transfect plant cells, its application to mammalian cells has met with limited success mainly due to high toxicity. Here we present a more refined nanotechnological approach to biolistic delivery with light-triggered self-assembled nanobombs (NBs) that consist of a photothermal core particle surrounded by smaller nanoprojectiles. Upon irradiation with pulsed laser light, fast heating of the core particle results in vapor bubble formation, which propels the nanoprojectiles through the cell membrane of nearby cells. We show successful transfection of both adherent and non-adherent cells with mRNA and pDNA, outperforming electroporation as the most used physical transfection technology by a factor of 5.5–7.6 in transfection yield. With a throughput of 104-105 cells per second, biolistic delivery with NBs offers scalable and highly efficient transfections of mammalian cells. Ballistic delivery with micro/nano-particles has been successfully used to transfect plant cells, however, has failed in mammalian cells due to toxic effects. Here, the authors report on a self-assembled nano-ballistic delivery system for the delivery of functional macromolecules and demonstrate efficient transfection of mammalian cells.
Collapse
|
6
|
Seisel Q, Lakumpa I, Josse E, Vivès E, Varilh J, Taulan-Cadars M, Boisguérin P. Highway to Cell: Selection of the Best Cell-Penetrating Peptide to Internalize the CFTR-Stabilizing iCAL36 Peptide. Pharmaceutics 2022; 14:pharmaceutics14040808. [PMID: 35456644 PMCID: PMC9032934 DOI: 10.3390/pharmaceutics14040808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 11/23/2022] Open
Abstract
Therapeutic peptides have regained interest as they can address unmet medical needs and can be an excellent complement to pharmaceutic small molecules and other macromolecular therapeutics. Over the past decades, correctors and potentiators of the cystic fibrosis transmembrane conductance regulator (CFTR), a chloride ion channel causing cystic fibrosis (CF) when mutated, were developed to reduce the symptoms of the patients. In this context, we have previously designed a CFTR-stabilizing iCAL36 peptide able to further increase the CFTR amount in epithelial cells, thereby resulting in a higher CFTR activity. In the present study, optimization of the peptidyl inhibitor was performed by coupling five different cell-penetrating peptides (CPP), which are Tat, dTat, TatRI (retro-inverso), MPG, and Penetratin. Screening of the internalization properties of these CPP-iCAL36 peptides under different conditions (with or without serum or endocytosis inhibitors, etc.) was performed to select TatRI as the optimal CPP for iCAL36 delivery. More importantly, using this TatRI-iCAL36 peptide, we were able to reveal for the first time an additive increase in the CFTR amount in the presence of VX-445/VX-809 compared to VX-445/VX-809 treatment alone. This finding is a significant contribution to the development of CFTR-stabilizing peptides in addition to currently used treatments (small-molecule correctors or potentiators) for CF patients.
Collapse
Affiliation(s)
- Quentin Seisel
- CRBM, University of Montpellier, CNRS UMR 5237, 34000 Montpellier, France
| | - Israpong Lakumpa
- CRBM, University of Montpellier, CNRS UMR 5237, 34000 Montpellier, France
| | - Emilie Josse
- PhyMedExp, Bâtiment Crastes de Paulet, University of Montpellier, INSERM U1046, CNRS UMR 9214, 34000 Montpellier, France
| | - Eric Vivès
- PhyMedExp, Bâtiment Crastes de Paulet, University of Montpellier, INSERM U1046, CNRS UMR 9214, 34000 Montpellier, France
| | - Jessica Varilh
- PhyMedExp, Institut Universitaire de Recherche Clinique, University of Montpellier, INSERM U1046, CNRS UMR 9214, 34000 Montpellier, France
| | - Magali Taulan-Cadars
- PhyMedExp, Institut Universitaire de Recherche Clinique, University of Montpellier, INSERM U1046, CNRS UMR 9214, 34000 Montpellier, France
| | - Prisca Boisguérin
- PhyMedExp, Bâtiment Crastes de Paulet, University of Montpellier, INSERM U1046, CNRS UMR 9214, 34000 Montpellier, France
| |
Collapse
|
7
|
Faria R, Paul M, Biswas S, Vivès E, Boisguérin P, Sousa Â, Costa D. Peptides vs. Polymers: Searching for the Most Efficient Delivery System for Mitochondrial Gene Therapy. Pharmaceutics 2022; 14:757. [PMID: 35456591 PMCID: PMC9026848 DOI: 10.3390/pharmaceutics14040757] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Together with the nucleus, the mitochondrion has its own genome. Mutations in mitochondrial DNA are responsible for a variety of disorders, including neurodegenerative diseases and cancer. Current therapeutic approaches are not effective. In this sense, mitochondrial gene therapy emerges as a valuable and promising therapeutic tool. To accomplish this goal, the design/development of a mitochondrial-specific gene delivery system is imperative. In this work, we explored the ability of novel polymer- and peptide-based systems for mitochondrial targeting, gene delivery, and protein expression, performing a comparison between them to reveal the most adequate system for mitochondrial gene therapy. Therefore, we synthesized a novel mitochondria-targeting polymer (polyethylenimine-dequalinium) to load and complex a mitochondrial-gene-based plasmid. The polymeric complexes exhibited physicochemical properties and cytotoxic profiles dependent on the nitrogen-to-phosphate-group ratio (N/P). A fluorescence confocal microscopy study revealed the mitochondrial targeting specificity of polymeric complexes. Moreover, transfection mediated by polymer and peptide delivery systems led to gene expression in mitochondria. Additionally, the mitochondrial protein was produced. A comparative study between polymeric and peptide/plasmid DNA complexes showed the great capacity of peptides to complex pDNA at lower N/P ratios, forming smaller particles bearing a positive charge, with repercussions on their capacity for cellular transfection, mitochondria targeting and, ultimately, gene delivery and protein expression. This report is a significant contribution to the implementation of mitochondrial gene therapy, instigating further research on the development of peptide-based delivery systems towards clinical translation.
Collapse
Affiliation(s)
- Rúben Faria
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilha, Portugal; (R.F.); (Â.S.)
| | - Milan Paul
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, India; (M.P.); (S.B.)
| | - Swati Biswas
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, India; (M.P.); (S.B.)
| | - Eric Vivès
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34295 Montpellier, France; (E.V.); (P.B.)
| | - Prisca Boisguérin
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34295 Montpellier, France; (E.V.); (P.B.)
| | - Ângela Sousa
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilha, Portugal; (R.F.); (Â.S.)
| | - Diana Costa
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilha, Portugal; (R.F.); (Â.S.)
| |
Collapse
|
8
|
Van de Vyver T, De Smedt SC, Raemdonck K. Modulating intracellular pathways to improve non-viral delivery of RNA therapeutics. Adv Drug Deliv Rev 2022; 181:114041. [PMID: 34763002 DOI: 10.1016/j.addr.2021.114041] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/12/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022]
Abstract
RNA therapeutics (e.g. siRNA, oligonucleotides, mRNA, etc.) show great potential for the treatment of a myriad of diseases. However, to reach their site of action in the cytosol or nucleus of target cells, multiple intra- and extracellular barriers have to be surmounted. Several non-viral delivery systems, such as nanoparticles and conjugates, have been successfully developed to meet this requirement. Unfortunately, despite these clear advances, state-of-the-art delivery agents still suffer from relatively low intracellular delivery efficiencies. Notably, our current understanding of the intracellular delivery process is largely oversimplified. Gaining mechanistic insight into how RNA formulations are processed by cells will fuel rational design of the next generation of delivery carriers. In addition, identifying which intracellular pathways contribute to productive RNA delivery could provide opportunities to boost the delivery performance of existing nanoformulations. In this review, we discuss both established as well as emerging techniques that can be used to assess the impact of different intracellular barriers on RNA transfection performance. Next, we highlight how several modulators, including small molecules but also genetic perturbation technologies, can boost RNA delivery by intervening at differing stages of the intracellular delivery process, such as cellular uptake, intracellular trafficking, endosomal escape, autophagy and exocytosis.
Collapse
Affiliation(s)
- Thijs Van de Vyver
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
9
|
A core-shell nanoplatform as a nonviral vector for targeted delivery of genes to the retina. Acta Biomater 2021; 134:605-620. [PMID: 34329781 DOI: 10.1016/j.actbio.2021.07.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/18/2021] [Accepted: 07/22/2021] [Indexed: 01/05/2023]
Abstract
Retinal diseases, including age-related macular degeneration (AMD), are a major cause of blindness. Efficient delivery of therapeutic genes to retinal cells to treat retinal disease is a formidable challenge. In this study, we developed a core-shell nanoplatform composed of a core and two external layers for targeted delivery of the gene to the retina. The inner core was composed of amino acid-functionalized dendrimers and a nuclear localization signal (NLS) for DNA complexation, nuclear transport and efficient transfection. The inner core was coated in a lipid bilayer that comprised pH-sensitive lipids as the inner shell layer. Hyaluronic acid (HA)-1,2-dioleoylphosphatidylethanolamine (DOPE) as the outermost shell layer was used for retinal cell targeting. This core-shell nanoplatform was developed so that the mobility in the vitreous body of these negatively charged carriers would not be affected by their surface charge, allowing diffusion into the retina, uptake into the retinal cells via CD44-mediated internalization, and finally transport into the nucleus by the NLS. The designed nanoparticles showed safety both in vitro and in vivo and inhibited the expression of VEGF under hypoxia-mimicking conditions. In vitro angiogenesis assays exhibited significant inhibitory effects on cell migration and tube formation. The in vivo assays indicated that this nanoplatform could be delivered to the retina. Taken together, this nanoplatform has the potential to transfer gene material into the retina for the treatment of retinal diseases, including AMD. STATEMENT OF SIGNIFICANCE: It remains a challenge to develop an efficient nonviral vector for gene therapy, especially retinal gene therapy. Various barriers exist in gene delivery and the unique ocular environment, making gene delivery to the retina difficult. In this study, we designed a negatively charged core-shell nanoplatform (HD-NPPND) for the targeted delivery of gene to the retina. The developed nanoplatform possessed excellent transfection efficiency and safety both in vitro and in vivo. It efficiently delivered a gene to the retina. The results of this study suggested that this core-shell nanoplatform has the potential to deliver genes to the retina to treat retinal diseases, including age-related macular degeneration (AMD).
Collapse
|
10
|
Zhang Y, Zhu J, Xu H, Yi Q, Yan L, Ye L, Zhang X, Xie M, Tan B. Time-Dependent Internalization of S100B by Mesenchymal Stem Cells via the Pathways of Clathrin- and Lipid Raft-Mediated Endocytosis. Front Cell Dev Biol 2021; 9:674995. [PMID: 34381770 PMCID: PMC8351554 DOI: 10.3389/fcell.2021.674995] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/06/2021] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are promising tools for cancer therapy, but there is a risk of malignant transformation in their clinical application. Our previous work revealed that the paracrine protein S100B in the glioma microenvironment induces malignant transformation of MSCs and upregulates intracellular S100B, which could affect cell homeostasis by interfering with p53. The purpose of this study was to investigate whether extracellular S100B can be internalized by MSCs and the specific endocytic pathway involved in S100B internalization. By using real-time confocal microscopy and structured illumination microscopy (SIM), we visualized the uptake of fluorescently labeled S100B protein (S100B-Alexa488) and monitored the intracellular trafficking of internalized vesicles. The results showed that S100B-Alexa488 was efficiently internalized into MSCs in a time-dependent manner and transported through endolysosomal pathways. After that, we used chemical inhibitors and RNA interference approaches to investigate possible mechanisms involved in S100B-Alexa488 uptake. The internalization of S100B-Alexa488 was inhibited by pitstop-2 or dyngo-4a treatment or RNA-mediated silencing of clathrin or dynamin, and the lipid raft-mediated endocytosis inhibitors nystatin and MβCD. In conclusion, our findings show that clathrin and lipid rafts contribute to the internalization of S100B-Alexa488, which provides promising interventions for the safe application of MSCs in glioma therapy.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jing Zhu
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Hao Xu
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
- Department of Clinical Laboratory, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Yi
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Liang Yan
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Liang Ye
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xinyuan Zhang
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Min Xie
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Bin Tan
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
11
|
Dutta K, Das R, Medeiros J, Kanjilal P, Thayumanavan S. Charge-Conversion Strategies for Nucleic Acid Delivery. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2011103. [PMID: 35832306 PMCID: PMC9275120 DOI: 10.1002/adfm.202011103] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Indexed: 05/05/2023]
Abstract
Nucleic acids are now considered as one of the most potent therapeutic modalities, as their roles go beyond storing genetic information and chemical energy or as signal transducer. Attenuation or expression of desired genes through nucleic acids have profound implications in gene therapy, gene editing and even in vaccine development for immunomodulation. Although nucleic acid therapeutics bring in overwhelming possibilities towards the development of molecular medicines, there are significant loopholes in designing and effective translation of these drugs into the clinic. One of the major pitfalls lies in the traditional design concepts for nucleic acid drug carriers, viz. cationic charge induced cytotoxicity in delivery pathway. Targeting this bottleneck, several pioneering research efforts have been devoted to design innovative carriers through charge-conversion approaches, whereby built-in functionalities convert from cationic to neutral or anionic, or even from anionic to cationic enabling the carrier to overcome several critical barriers for therapeutics delivery, such as serum deactivation, instability in circulation, low transfection and poor endosomal escape. This review will critically analyze various molecular designs of charge-converting nanocarriers in a classified approach for the successful delivery of nucleic acids. Accompanied by the narrative on recent clinical nucleic acid candidates, the review concludes with a discussion on the pitfalls and scope of these interesting approaches.
Collapse
Affiliation(s)
- Kingshuk Dutta
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Corteva Agriscience, 9330 Zionsville Road, Indianapolis 46268, United States
| | - Ritam Das
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Jewel Medeiros
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Pintu Kanjilal
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
- The Center for Bioactive Delivery- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
12
|
Åberg C. Kinetics of nanoparticle uptake into and distribution in human cells. NANOSCALE ADVANCES 2021; 3:2196-2212. [PMID: 36133761 PMCID: PMC9416924 DOI: 10.1039/d0na00716a] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 03/12/2021] [Indexed: 05/17/2023]
Abstract
Whether one wishes to optimise drug delivery using nano-sized carriers or avoid hazard posed by engineered nanomaterials, the kinetics of nanoparticle uptake into human cells and their subsequent intracellular distribution is key. Unique properties of the nanoscale implies that such nanoparticles are taken up and trafficked in a different fashion compared to molecular species. In this review, we discuss in detail how to describe the kinetics of nanoparticle uptake and intracellular distribution, using previous studies for illustration. We also cover the extracellular kinetics, particle degradation, endosomal escape and cell division, ending with an outlook on the future of kinetic studies.
Collapse
Affiliation(s)
- Christoffer Åberg
- Groningen Research Institute of Pharmacy, University of Groningen Antonius Deusinglaan 1 9713AV Groningen The Netherlands
| |
Collapse
|
13
|
Åberg C, Poolman B. Glass-like characteristics of intracellular motion in human cells. Biophys J 2021; 120:2355-2366. [PMID: 33887228 DOI: 10.1016/j.bpj.2021.04.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 03/29/2021] [Accepted: 04/09/2021] [Indexed: 10/21/2022] Open
Abstract
The motion in the cytosol of microorganisms such as bacteria and yeast has been observed to undergo a dramatic slowing down upon cell energy depletion. These observations have been interpreted as the motion being "glassy," but whether this notion is useful also for active, motor-protein-driven transport in eukaryotic cells is less clear. Here, we use fluorescence microscopy of beads in human (HeLa) cells to probe the motion of membrane-surrounded structures that are carried along the cytoskeleton by motor proteins. Evaluating several hallmarks of glassy dynamics, we show that at short length scales, the motion is heterogeneous, is nonergodic, is well described by a model for the displacement distribution in glassy systems, and exhibits a decoupling of the exchange and persistence times. Overall, these results suggest that the short length scale behavior of objects that can be transported actively by motor proteins in human cells shares features with the motion in glassy systems.
Collapse
Affiliation(s)
- Christoffer Åberg
- Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands; Department of Biochemistry, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands.
| | - Bert Poolman
- Department of Biochemistry, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
14
|
Polycaprolactone Nanoparticles as Promising Candidates for Nanocarriers in Novel Nanomedicines. Pharmaceutics 2021; 13:pharmaceutics13020191. [PMID: 33535563 PMCID: PMC7912766 DOI: 10.3390/pharmaceutics13020191] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 01/04/2023] Open
Abstract
An investigation of the interactions between bio-polymeric nanoparticles (NPs) and the RAW 264.7 mouse murine macrophage cell line has been presented. The cell viability, immunological response, and endocytosis efficiency of NPs were studied. Biopolymeric NPs were synthesized from a nanoemulsion using the phase inversion composition (PIC) technique. The two types of biopolymeric NPs that were obtained consisted of a biocompatible polymer, polycaprolactone (PCL), either with or without its copolymer with poly(ethylene glycol) (PCL-b-PEG). Both types of synthesized PCL NPs passed the first in vitro quality assessments as potential drug nanocarriers. Non-pegylated PCL NPs were internalized more effectively and the clathrin-mediated pathway was involved in that process. The investigated NPs did not affect the viability of the cells and did not elicit an immune response in the RAW 264.7 cells (neither a significant increase in the expression of genes encoding pro-inflammatory cytokines nor NO (nitric oxide) production were observed). It may be concluded that the synthesized NPs are promising candidates as nanocarriers of therapeutic compounds.
Collapse
|
15
|
Gu W, Meng F, Haag R, Zhong Z. Actively targeted nanomedicines for precision cancer therapy: Concept, construction, challenges and clinical translation. J Control Release 2021; 329:676-695. [DOI: 10.1016/j.jconrel.2020.10.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/13/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023]
|
16
|
Tao J, Diao L, Chen F, Shen A, Wang S, Jin H, Cai D, Hu Y. pH-Sensitive Nanoparticles Codelivering Docetaxel and Dihydroartemisinin Effectively Treat Breast Cancer by Enhancing Reactive Oxidative Species-Mediated Mitochondrial Apoptosis. Mol Pharm 2020; 18:74-86. [PMID: 33084332 DOI: 10.1021/acs.molpharmaceut.0c00432] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tumor growth and metastasis are the major causes of high mortality in breast cancer. We previously constructed pH-sensitive nanoparticles (D/D NPs) for the codelivery of docetaxel (DTX) and dihydroartemisinin (DHA) and demonstrated that D/D NPs showed anticancer activity in breast cancer cells in vitro. The present study further investigated the therapeutic effect of D/D NPs on orthotopic breast cancer in vivo and examined the antitumor mechanism of D/D NPs. D/D NPs significantly increased the apoptosis of 4T1 cells with a synergistic effect of DTX and DHA. D/D NPs increased reactive oxygen species, reduced mitochondrial membrane potential, increased the expression of p53, and induced cytochrome c release into the cytoplasm to activate caspase-3. In an orthotopic metastatic breast cancer mouse model derived from 4T1 cells, D/D NPs inhibited tumor growth and prevented lung metastasis due to the synergistic effect of DTX and DHA. No distinct changes were observed in the histology of major organs. These results indicate that pH-sensitive D/D NP-based combination therapy may be a promising strategy for the treatment of metastatic breast cancers via the ROS-mediated mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Jin Tao
- Zhejiang Pharmaceutical College, Ningbo, Zhejiang 315100, China.,School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Lu Diao
- Zhejiang Pharmaceutical College, Ningbo, Zhejiang 315100, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Fangcheng Chen
- Zhejiang Pharmaceutical College, Ningbo, Zhejiang 315100, China
| | - Ao Shen
- The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Shutian Wang
- Zhejiang Pharmaceutical College, Ningbo, Zhejiang 315100, China
| | - Hongyan Jin
- Zhejiang Pharmaceutical College, Ningbo, Zhejiang 315100, China
| | - Danwei Cai
- Zhejiang Pharmaceutical College, Ningbo, Zhejiang 315100, China
| | - Ying Hu
- Zhejiang Pharmaceutical College, Ningbo, Zhejiang 315100, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| |
Collapse
|
17
|
A molecular sensor to quantify the localization of proteins, DNA and nanoparticles in cells. Nat Commun 2020; 11:4482. [PMID: 32901011 PMCID: PMC7479595 DOI: 10.1038/s41467-020-18082-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 07/28/2020] [Indexed: 12/22/2022] Open
Abstract
Intracellular trafficking governs receptor signaling, pathogenesis, immune responses and fate of nanomedicines. These processes are typically tracked by observing colocalization of fluorescent markers using confocal microscopy. However, this method is low throughput, limited by the resolution of microscopy, and can miss fleeting interactions. To address this, we developed a localization sensor composed of a quenched SNAP-tag substrate (SNAPSwitch) that can be conjugated to biomolecules using click chemistry. SNAPSwitch enables quantitative detection of trafficking to locations of interest within live cells using flow cytometry. Using SNAPSwitch, we followed the trafficking of DNA complexes from endosomes into the cytosol and nucleus. We show that antibodies against the transferrin or hyaluronan receptor are initially sorted into different compartments following endocytosis. In addition, we can resolve which side of the cellular membrane material was located. These results demonstrate SNAPSwitch is a high-throughput and broadly applicable tool to quantitatively track localization of materials in cells. Determining the trafficking of intracellular material is commonly done by colocalisation analysis using microscopy. Here the authors monitor trafficking of select cargo by measuring the conversion of quenched SNAP-tag substrates by subcellularly-localised SNAP-tag and detection by flow cytometry.
Collapse
|
18
|
Liu J, Fraire JC, De Smedt SC, Xiong R, Braeckmans K. Intracellular Labeling with Extrinsic Probes: Delivery Strategies and Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2000146. [PMID: 32351015 DOI: 10.1002/smll.202000146] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/29/2020] [Accepted: 03/16/2020] [Indexed: 06/11/2023]
Abstract
Extrinsic probes have outstanding properties for intracellular labeling to visualize dynamic processes in and of living cells, both in vitro and in vivo. Since extrinsic probes are in many cases cell-impermeable, different biochemical, and physical approaches have been used to break the cell membrane barrier for direct delivery into the cytoplasm. In this Review, these intracellular delivery strategies are discussed, briefly explaining the mechanisms and how they are used for live-cell labeling applications. Methods that are discussed include three biochemical agents that are used for this purpose-purpose-different nanocarriers, cell penetrating peptides and the pore-foraming bacterial toxin streptolysin O. Most successful intracellular label delivery methods are, however, based on physical principles to permeabilize the membrane and include electroporation, laser-induced photoporation, micro- and nanoinjection, nanoneedles or nanostraws, microfluidics, and nanomachines. The strengths and weaknesses of each strategy are discussed with a systematic comparison provided. Finally, the extrinsic probes that are reported for intracellular labeling so-far are summarized, together with the delivery strategies that are used and their performance. This combined information should provide for a useful guide for choosing the most suitable delivery method for the desired probes.
Collapse
Affiliation(s)
- Jing Liu
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
| | - Juan C Fraire
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
- Centre for Advanced Light Microscopy, Ghent University, Ghent, B-9000, Belgium
- Joint Laboratory of Advanced Biomedical Technology (NFU-UGent), College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, 210037, P. R. China
| | - Ranhua Xiong
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
- Centre for Advanced Light Microscopy, Ghent University, Ghent, B-9000, Belgium
| |
Collapse
|
19
|
Zhang HT, Yu M, Niu YJ, Liu WZ, Pang WH, Ding J, Wang JC. Polyarginine-Mediated siRNA Delivery: A Mechanistic Study of Intracellular Trafficking of PCL-R15/siRNA Nanoplexes. Mol Pharm 2020; 17:1685-1696. [PMID: 32191042 DOI: 10.1021/acs.molpharmaceut.0c00120] [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] [Indexed: 01/06/2023]
Abstract
As a cell-penetrating peptide, polyarginine is widely used in drug delivery systems based on its membrane permeation ability. Previously, we developed the mPEG-PLA-b-polyarginine(R15) triblock copolymer, which exhibited a high siRNA delivery efficiency both in vitro and in vivo. As a continued effort, here the amphiphilic diblock polymer PCL-R15 was synthesized as a simplified model to further elucidate the structure-activity relationship of arginine-based amphiphilic polymers as siRNA delivery systems, and the cellular trafficking mechanisms of the PCL-R15/siRNA nanoplexes were investigated to understand the interaction patterns between the nanoplexes and cells. Compared to the R15/siRNA complexes, the introduction of PCL moiety was found to result in the stronger interactions with cells and the enhanced transfection efficiency after the formation of condensed nanoplexes. Caveolae-mediated endocytosis and clathrin-mediated endocytosis were major routes for the internalization of PCL-R15/siRNA nanoplexes. The intracellular release of siRNA from nanoplexes was confirmed by fluorescence resonance energy transfer assay. It was also noticed that the internalized PCL-R15/siRNA nanoplexes were transported through digestive routes and trapped in lysosomes, which may be the bottleneck for efficient siRNA delivery of PCL-R15/siRNA nanoplexes. This study investigated the relationship between the polymer structure of PCL-R15 and the cellular interaction patterns, which may render implications on the rational design of polyarginine-based siRNA delivery systems.
Collapse
Affiliation(s)
- Hai-Tao Zhang
- Xiangya School of Pharmaceutical Sciences, Central South University, 172 Tongzipo Road, Changsha, Hunan 410013, P. R. China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, P. R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, 28 Western Changshen Road, Hengyang, Hunan 421001, P. R. China
| | - Minzhi Yu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, P. R. China
| | - Yu-Jie Niu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, P. R. China
| | - Wei-Zhong Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, P. R. China
| | - Wen-Hao Pang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, P. R. China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, 172 Tongzipo Road, Changsha, Hunan 410013, P. R. China
| | - Jian-Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, P. R. China
| |
Collapse
|
20
|
Deshayes S, Konate K, Dussot M, Chavey B, Vaissière A, Van TNN, Aldrian G, Padari K, Pooga M, Vivès E, Boisguérin P. Deciphering the internalization mechanism of WRAP:siRNA nanoparticles. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183252. [PMID: 32135145 DOI: 10.1016/j.bbamem.2020.183252] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/17/2020] [Accepted: 02/27/2020] [Indexed: 01/08/2023]
Abstract
Gene silencing mediated by double-stranded small interfering RNA (siRNA) has been widely investigated as a potential therapeutic approach for a variety of diseases and, indeed, the first therapeutic siRNA was approved by the FDA in 2018. As an alternative to the traditional delivery systems for nucleic acids, peptide-based nanoparticles (PBNs) have been applied successfully for siRNA delivery. Recently, we have developed amphipathic cell-penetrating peptides (CPPs), called WRAP allowing a rapid and efficient siRNA delivery into several cell lines at low doses (20 to 50 nM). In this study, using a highly specific gene silencing system, we aimed to elucidate the cellular uptake mechanism of WRAP:siRNA nanoparticles by combining biophysical, biological, confocal and electron microscopy approaches. We demonstrated that WRAP:siRNA complexes remain fully active in the presence of chemical inhibitors of different endosomal pathways suggesting a direct cell membrane translocation mechanism. Leakage studies on lipid vesicles indicated membrane destabilization properties of the nanoparticles and this was supported by the measurement of WRAP:siRNA internalization in dynamin triple-KO cells. However, we also observed some evidences for an endocytosis-dependent cellular internalization. Indeed, nanoparticles co-localized with transferrin, siRNA silencing was inhibited by the scavenger receptor A inhibitor Poly I and nanoparticles encapsulated in vesicles were observed by electron microscopy in U87 cells. In conclusion, we demonstrate here that the efficiency of WRAP:siRNA nanoparticles is mainly based on the use of multiple internalization mechanisms including direct translocation as well as endocytosis-dependent pathways.
Collapse
Affiliation(s)
- Sébastien Deshayes
- Centre de Recherche de Biologie cellulaire de Montpellier, CNRS UMR 5237, 1919 Route de Mende, 34293, Montpellier Cedex 5, France
| | - Karidia Konate
- Centre de Recherche de Biologie cellulaire de Montpellier, CNRS UMR 5237, 1919 Route de Mende, 34293, Montpellier Cedex 5, France
| | - Marion Dussot
- Centre de Recherche de Biologie cellulaire de Montpellier, CNRS UMR 5237, 1919 Route de Mende, 34293, Montpellier Cedex 5, France
| | - Bérengère Chavey
- Centre de Recherche de Biologie cellulaire de Montpellier, CNRS UMR 5237, 1919 Route de Mende, 34293, Montpellier Cedex 5, France; Sys2Diag, UMR 9005-CNRS/ALCEDIAG, 1682 Rue de la Valsière, 34184, Montpellier, CEDEX 4, France
| | - Anaïs Vaissière
- Centre de Recherche de Biologie cellulaire de Montpellier, CNRS UMR 5237, 1919 Route de Mende, 34293, Montpellier Cedex 5, France
| | - Thi Nhu Ngoc Van
- Sys2Diag, UMR 9005-CNRS/ALCEDIAG, 1682 Rue de la Valsière, 34184, Montpellier, CEDEX 4, France
| | - Gudrun Aldrian
- Sys2Diag, UMR 9005-CNRS/ALCEDIAG, 1682 Rue de la Valsière, 34184, Montpellier, CEDEX 4, France
| | - Kärt Padari
- Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
| | - Margus Pooga
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | - Eric Vivès
- Centre de Recherche de Biologie cellulaire de Montpellier, CNRS UMR 5237, 1919 Route de Mende, 34293, Montpellier Cedex 5, France
| | - Prisca Boisguérin
- Centre de Recherche de Biologie cellulaire de Montpellier, CNRS UMR 5237, 1919 Route de Mende, 34293, Montpellier Cedex 5, France.
| |
Collapse
|
21
|
Muhammad K, Zhao J, Ullah I, Guo J, Ren XK, Feng Y. Ligand targeting and peptide functionalized polymers as non-viral carriers for gene therapy. Biomater Sci 2020; 8:64-83. [DOI: 10.1039/c9bm01112a] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ligand targeting and peptide functionalized polymers serve as gene carriers for efficient gene delivery.
Collapse
Affiliation(s)
- Khan Muhammad
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- P. R. China
| | - Jing Zhao
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- P. R. China
| | - Ihsan Ullah
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- P. R. China
| | - Jintang Guo
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- P. R. China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| | - Xiang-kui Ren
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- P. R. China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| | - Yakai Feng
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- P. R. China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| |
Collapse
|
22
|
Pinnapireddy SR, Giselbrecht J, Strehlow B, Janich C, Husteden C, Meister A, Loppnow H, Sedding D, Erdmann F, Hause G, Brezesinski G, Groth T, Langner A, Bakowsky U, Wölk C. A triple chain polycationic peptide-mimicking amphiphile - efficient DNA-transfer without co-lipids. Biomater Sci 2019; 8:232-249. [PMID: 31681923 DOI: 10.1039/c9bm01093a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Non-viral gene delivery in its current form is largely dependent upon the ability of a delivery vehicle to protect its cargo in the extracellular environment and release it efficiently inside the target cell. Also a simple delivery system is required to simplify a GMP conform production if a marketing authorization is striven for. This work addresses these problems. We have developed a synthetic polycationic peptide-mimicking amphiphile, namely DiTT4, which shows efficient transfection rates and good biocompatibility without the use of a co-lipid in the formulation. The lipid-nucleic acid complex (lipoplex) was characterized at the structural (electron microscopy), physical (laser Doppler velocimetry and atomic force microscopy) and molecular levels (X-ray scattering). Stability studies of the lipoplexes in the presence of serum and heparin indicated a stable formation capable of protecting the cargo against the extracellular milieu. Hemocompatibility studies (hemolysis, complement activation and erythrocyte aggregation) demonstrated the biocompatibility of the formulation for systemic administration. The transfection efficiency was assessed in vitro using the GFP assay and confocal laser scanning microscopy studies. With the chorioallantoic membrane model, an animal replacement model according to the 3R strategy (replacement, refinement, and reduction), initial in vivo experiments were performed which demonstrate fast and efficient transfection in complex tissues and excellent biocompatibility.
Collapse
Affiliation(s)
- Shashank Reddy Pinnapireddy
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037, Marburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Pengnam S, Plainwong S, Patrojanasophon P, Rojanarata T, Ngawhirunpat T, Radchatawedchakoon W, Niyomtham N, Yingyongnarongkul BE, Opanasopit P. Effect of hydrophobic tails of plier-like cationic lipids on nucleic acid delivery and intracellular trafficking. Int J Pharm 2019; 573:118798. [PMID: 31759106 DOI: 10.1016/j.ijpharm.2019.118798] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/02/2019] [Accepted: 10/13/2019] [Indexed: 11/26/2022]
Abstract
In the optimization of transfection efficacy, one of the crucial barriers to effective gene delivery is in fact the intracellular trafficking of nucleic acids, besides the first and the last steps of gene transfer, i.e., delivery to the cell and transcription. Modifications of cationic lipid structure have been reported to have a significant effect on gene delivery. Therefore, the plier-like cationic lipids (PCLs) have been synthesized and the effect of the different types of hydrophobic tails (chain length and unsaturated hydrocarbon) on physicochemical properties, cellular uptake, trafficking process, transfection, and silencing efficiency has been investigated. In this study, the plier-like cationic niosomes (PCNs) containing PCL (A, B, and C) were evaluated their performance to deliver pDNA and siRNA to HeLa cells. Among the PCNs, PCN-B with saturated asymmetric hydrocarbon tails (C18 and C12) provided the highest efficiency for pDNA and siRNA delivery. Furthermore, the results revealed that the structure of the cationic lipids affected the internalization pathway and the intracellular trafficking. PCL-B and PCL-C with asymmetric tails preferred clathrin- and caveolae-mediated endocytosis as the predominant internalization pathways and were also involved in the polymerization process for transfection. However, PCL-A with symmetry hydrocarbon tails (C12) was predominantly taken up via macropinocytosis. All PCNs were able to escape from endosomal-lysosomal systems through facilitation of acidification. Results obtained from the cytotoxicity test revealed that the PCNs were safe in vitro. Therefore, PCNs provide a great prospect as an alternative effective gene delivery system.
Collapse
Affiliation(s)
- Supusson Pengnam
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | | | - Prasopchai Patrojanasophon
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Theerasak Rojanarata
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Tanasait Ngawhirunpat
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Widchaya Radchatawedchakoon
- Creative Chemistry and Innovation Research Unit, Department of Chemistry and Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahasarakham University, MahaSarakham 44150, Thailand
| | - Nattisa Niyomtham
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok 10240, Thailand
| | - Boon-Ek Yingyongnarongkul
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok 10240, Thailand
| | - Praneet Opanasopit
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
| |
Collapse
|
24
|
Zhong L, Liu Y, Xu L, Li Q, Zhao D, Li Z, Zhang H, Zhang H, Kan Q, Sun J, He Z. Exploring the relationship of hyaluronic acid molecular weight and active targeting efficiency for designing hyaluronic acid-modified nanoparticles. Asian J Pharm Sci 2019; 14:521-530. [PMID: 32104479 PMCID: PMC7032078 DOI: 10.1016/j.ajps.2018.11.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/12/2018] [Accepted: 11/04/2018] [Indexed: 01/08/2023] Open
Abstract
Although it is reported that the targeting ability of hyaluronic acid (HA)-based nanoparticles (NPs) is molecular weight (MW) dependent, the influence of HA MW on targeting efficiency of HA-functionalized NPs and the underlying mechanism remain elusive. In this study, we constituted three HA-functionalized Dox-loaded NPs (Dox/HCVs) different HA MWs (7, 63, and 102 kDa) and attempted to illustrate the effects of HA MW on the targeting efficiency. The three Dox/HCVs had similar physiochemical and pharmaceutical characteristics, but showed different affinity to CD44 receptor. Furthermore, Dox/HCV-63 exerted the best targeting effect and the highest cytotoxicity compared with Dox/HCV-7 and Dox/HCV-102. It was interesting to found that both the HA-CD44 binding affinity and induced CD44 clustering by HA-based NPs were HA MW-dependent, the two of which determine the apparent targeting efficacy of Dox/HCV NPs in the conflicting directions. Those results laid a good foundation for rationally designing HA-based NPs in cancer therapy.
Collapse
Affiliation(s)
- Lu Zhong
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yanying Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lu Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qingsong Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dongyang Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhenbao Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Huicong Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haotian Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qiming Kan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
25
|
Oligo-guanidyl targeted bioconjugates forming rod shaped polyplexes as a new nanoplatform for oligonucleotide delivery. J Control Release 2019; 310:58-73. [PMID: 31400381 DOI: 10.1016/j.jconrel.2019.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/04/2019] [Accepted: 08/06/2019] [Indexed: 12/15/2022]
Abstract
Novel bioconjugates (Agm6-M-PEG-FA) for active oligonucleotide (ON) delivery have been developed by conjugating a cationic oligo-guanidyl star-like shaped "head" (Agm6-M) to a polymeric "tail" (PEG) terminating with folic acid (FA) as targeting agent or methoxy group (Agm6-M-PEG-FA and Agm6-M-PEG-OCH3, respectively). Gel electrophoresis showed that the bioconjugates completely associated with ONs at 3 nitrogen/phosphate (N/P) ratio. Studies performed with folate receptor (FR)-overexpressing HeLa cells, showed that optimal cell up-take was obtained with the 75:25 w/w Agm6-M-PEG-OCH3:Agm6-M-PEG-FA mixture. Dynamic light scattering and transmission electron microscopy showed that the polyplexes had size <80 nm with narrow polydispersity and rod-shaped morphology. The polyplexes were stable for several hours in plasma while ON was released in the presence of heparin concentration 16-times higher than the physiological one. The polyplexes displayed negligible cytotoxicity, hemolysis and low pro-inflammatory TNF-α release. Studies performed with FR-overexpressing HeLa and MDA-MB-231 cells using siRac1 revealed that the folated polyplexes caused significantly higher gene silencing (86.1 ± 9.6%) and inhibition of cell migration (40%) than the non-folated polyplexes obtained with Agm6-M-PEG-OCH3 only. Although cytofluorimetric analyses showed similar cell uptake for both folated and non-folated polyplexes, confocal, TEM and competition studies showed that the folated polyplexes were taken-up by lysosome escaping caveolin-mediated pathway with final polyplex localization within cytosol, while non-folated polyplexes were preferentially taken-up via clathrin-mediated pathway to localize in the lysosomes. Finally, preliminary in vivo studies carried out in mice revealed that the folated polyplexes dispose in the tumor mass.
Collapse
|
26
|
Pengnam S, Patrojanasophon P, Rojanarata T, Ngawhirunpat T, Yingyongnarongkul BE, Radchatawedchakoon W, Opanasopit P. A novel plier-like gemini cationic niosome for nucleic acid delivery. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.04.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Nguyen MA, Wyatt H, Susser L, Geoffrion M, Rasheed A, Duchez AC, Cottee ML, Afolayan E, Farah E, Kahiel Z, Côté M, Gadde S, Rayner KJ. Delivery of MicroRNAs by Chitosan Nanoparticles to Functionally Alter Macrophage Cholesterol Efflux in Vitro and in Vivo. ACS NANO 2019; 13:6491-6505. [PMID: 31125197 DOI: 10.1021/acsnano.8b09679] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The prevention and treatment of cardiovascular diseases (CVD) has largely focused on lowering circulating LDL cholesterol, yet a significant burden of atherosclerotic disease remains even when LDL is low. Recently, microRNAs (miRNAs) have emerged as exciting therapeutic targets for cardiovascular disease. miRNAs are small noncoding RNAs that post-transcriptionally regulate gene expression by degradation or translational inhibition of target mRNAs. A number of miRNAs have been found to modulate all stages of atherosclerosis, particularly those that promote the efflux of excess cholesterol from lipid-laden macrophages in the vessel wall to the liver. However, one of the major challenges of miRNA-based therapy is to achieve tissue-specific, efficient, and safe delivery of miRNAs in vivo. We sought to develop chitosan nanoparticles (chNPs) that can deliver functional miRNA mimics to macrophages and to determine if these nanoparticles can alter cholesterol efflux and reverse cholesterol transport in vivo. We developed chNPs with a size range of 150-200 nm via the ionic gelation method using tripolyphosphate (TPP) as a cross-linker. In this method, negatively charged miRNAs were encapsulated in the nanoparticles by ionic interactions with polymeric components. We then optimized the efficiency of intracellular delivery of different formulations of chitosan/TPP/miRNA to mouse macrophages. Using a well-defined miRNA with roles in macrophage cholesterol metabolism, we tested whether chNPs could deliver functional miRNAs to macrophages. We find chNPs can transfer exogenous miR-33 to naïve macrophages and reduce the expression of ABCA1, a potent miR-33 target gene, both in vitro and in vivo, confirming that miRNAs delivered via nanoparticles can escape the endosomal system and function in the RISC complex. Because miR-33 and ABCA1 play a key role in regulating the efflux of cholesterol from macrophages, we also confirmed that macrophages treated with miR-33-loaded chNPs exhibited reduced cholesterol efflux to apolipoprotein A1, further confirming functional delivery of the miRNA. In vivo, mice treated with miR33-chNPs showed decreased reverse cholesterol transport (RCT) to the plasma, liver, and feces. In contrast, when efflux-promoting miRNAs were delivered via chNPs, ABCA1 expression and cholesterol efflux into the RCT pathway were improved. Over all, miRNAs can be efficiently delivered to macrophages via nanoparticles, where they can function to regulate ABCA1 expression and cholesterol efflux, suggesting that these miRNA nanoparticles can be used in vivo to target atherosclerotic lesions.
Collapse
Affiliation(s)
- My-Anh Nguyen
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Hailey Wyatt
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Leah Susser
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Michele Geoffrion
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Adil Rasheed
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Anne-Claire Duchez
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Mary Lynn Cottee
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
| | - Esther Afolayan
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Eliya Farah
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Zaina Kahiel
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Suresh Gadde
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Katey J Rayner
- University of Ottawa Heart Institute , Ottawa , Ontario K1Y 4W7 , Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| |
Collapse
|
28
|
Yu G, Ning Q, Mo Z, Tang S. Intelligent polymeric micelles for multidrug co-delivery and cancer therapy. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1476-1487. [DOI: 10.1080/21691401.2019.1601104] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Guangping Yu
- Learning Key Laboratory for Pharmacoproteomics of Hunan Province, Institute of Pharmacy and Pharmacology, University of South China, Henyang, China
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, Hunan University of Medicine, Huaihua, China
| | - Qian Ning
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, Hunan University of Medicine, Huaihua, China
| | - Zhongcheng Mo
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Hengyang Medical school, University of South China, Henyang, China
| | - Shengsong Tang
- Learning Key Laboratory for Pharmacoproteomics of Hunan Province, Institute of Pharmacy and Pharmacology, University of South China, Henyang, China
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, Hunan University of Medicine, Huaihua, China
| |
Collapse
|
29
|
Lipid gene nanocarriers for the treatment of skin diseases: Current state-of-the-art. Eur J Pharm Biopharm 2019; 137:95-111. [DOI: 10.1016/j.ejpb.2019.02.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/21/2019] [Accepted: 02/15/2019] [Indexed: 12/19/2022]
|
30
|
Patel S, Kim J, Herrera M, Mukherjee A, Kabanov AV, Sahay G. Brief update on endocytosis of nanomedicines. Adv Drug Deliv Rev 2019; 144:90-111. [PMID: 31419450 PMCID: PMC6986687 DOI: 10.1016/j.addr.2019.08.004] [Citation(s) in RCA: 237] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/06/2019] [Accepted: 08/10/2019] [Indexed: 12/14/2022]
Abstract
The complexity of nanoscale interactions between biomaterials and cells has limited the realization of the ultimate vision of nanotechnology in diagnostics and therapeutics. As such, significant effort has been devoted to advancing our understanding of the biophysical interactions of the myriad nanoparticles. Endocytosis of nanomedicine has drawn tremendous interest in the last decade. Here, we highlight the ever-present barriers to efficient intracellular delivery of nanoparticles as well as the current advances and strategies deployed to breach these barriers. We also introduce new barriers that have been largely overlooked such as the glycocalyx and macromolecular crowding. Additionally, we draw attention to the potential complications arising from the disruption of the newly discovered functions of the lysosomes. Novel strategies of exploiting the inherent intracellular defects in disease states to enhance delivery and the use of exosomes for bioanalytics and drug delivery are explored. Furthermore, we discuss the advances in imaging techniques like electron microscopy, super resolution fluorescence microscopy, and single particle tracking which have been instrumental in our growing understanding of intracellular pathways and nanoparticle trafficking. Finally, we advocate for the push towards more intravital analysis of nanoparticle transport phenomena using the multitude of techniques available to us. Unraveling the underlying mechanisms governing the cellular barriers to delivery and biological interactions of nanoparticles will guide the innovations capable of breaching these barriers.
Collapse
Affiliation(s)
- Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Jeonghwan Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Marco Herrera
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Anindit Mukherjee
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA
| | - Alexander V Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow 119992, Russia.
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA; Department of Biomedical Engineering, Oregon Health and Science University, Robertson Life Science Building, 2730 SW Moody Avenue, Portland, OR 97201, USA.
| |
Collapse
|
31
|
Givens BE, Naguib YW, Geary SM, Devor EJ, Salem AK. Nanoparticle-Based Delivery of CRISPR/Cas9 Genome-Editing Therapeutics. AAPS J 2018; 20:108. [PMID: 30306365 PMCID: PMC6398936 DOI: 10.1208/s12248-018-0267-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/18/2018] [Indexed: 12/17/2022] Open
Abstract
The recent progress in harnessing the efficient and precise method of DNA editing provided by CRISPR/Cas9 is one of the most promising major advances in the field of gene therapy. However, the development of safe and optimally efficient delivery systems for CRISPR/Cas9 elements capable of achieving specific targeting of gene therapy to the location of interest without off-target effects is a primary challenge for clinical therapeutics. Nanoparticles (NPs) provide a promising means to meet such challenges. In this review, we present the most recent advances in developing innovative NP-based delivery systems that efficiently deliver CRISPR/Cas9 constructs and maximize their effectiveness.
Collapse
Affiliation(s)
- Brittany E Givens
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
- Department of Chemical and Biochemical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Youssef W Naguib
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Sean M Geary
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Eric J Devor
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA.
- Department of Chemical and Biochemical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa, 52242, USA.
| |
Collapse
|
32
|
Xu B, Zhu YJ, Wang CH, Qiu C, Sun J, Yan Y, Chen X, Wang JC, Zhang Q. Improved Cell Transfection of siRNA by pH-Responsive Nanomicelles Self-Assembled with mPEG- b-PHis- b-PEI Copolymers. ACS APPLIED MATERIALS & INTERFACES 2018; 10:21847-21860. [PMID: 29882640 DOI: 10.1021/acsami.8b04301] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Here, the novel pH-responsive nanomicelles self-assembled with amphipathic meo-poly(ethylene glycol)- b-poly(l-histidine)- b-polyethylenimine (mPEG- b-PHis- b-PEI, EHE) copolymers based on hydrophobic interaction of PHis with deprotonation of imidazoles were developed for siRNA transfection. The cationic nanomicelles could electrostatically compact siRNA into stable EHE/siRNA nanoplexes with a hydrodynamic diameter of ∼190 nm and present a low toxicity in normal physiological condition (pH ∼ 7.4). Different from pH-irresponsive ECE/siRNA nanoplexes based on mPEG- b-poly(ε-caprolactone)- b-PEI (ECE), the EHE/siRNA nanoplexes exhibited a higher cellular uptake along with an increased ζ-potential (from +18 to +32 mV) when the pH changed from 7.4 to 6.8 (extracellular acidic microenvironments). After cell internalization, the EHE/siRNA nanoplexes also exhibited an enhanced nanostructural disassembling and release of siRNA from lysosomal acidic microenvironments (pH ∼ 5.5). Furthermore, it was demonstrated that the EHE/siEGFR nanoplexes downregulated the expression levels of the corresponding mRNA and protein more efficiently than ECE/siEGFR in HeLa cells. The improved siRNA silencing effects of EHE/siEGFR nanoplexes resulted from the higher cellular uptake and enhanced endosomal/lysosomal escape, which is associated with the pH-responsive disassembly of nanostructure as well as the synergistic "proton sponge" effects of PHis and PEI in EHE copolymers. Therefore, the pH-responsive EHE nanomicelles would be promising and potential carriers for cell transfection of siRNA.
Collapse
Affiliation(s)
- Bin Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xueyuan Road 38 , Beijing 100191 , China
| | - Yuan-Jun Zhu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xueyuan Road 38 , Beijing 100191 , China
| | - Cheng-Han Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xueyuan Road 38 , Beijing 100191 , China
| | - Chong Qiu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xueyuan Road 38 , Beijing 100191 , China
| | - Jing Sun
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xueyuan Road 38 , Beijing 100191 , China
| | - Yi Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xueyuan Road 38 , Beijing 100191 , China
| | - Xin Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xueyuan Road 38 , Beijing 100191 , China
| | - Jian-Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xueyuan Road 38 , Beijing 100191 , China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Xueyuan Road 38 , Beijing 100191 , China
| |
Collapse
|
33
|
Trützschler AK, Bus T, Reifarth M, Brendel JC, Hoeppener S, Traeger A, Schubert US. Beyond Gene Transfection with Methacrylate-Based Polyplexes-The Influence of the Amino Substitution Pattern. Bioconjug Chem 2018; 29:2181-2194. [PMID: 29712427 DOI: 10.1021/acs.bioconjchem.8b00074] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Methacrylate-based polymers represent promising nonviral gene delivery vectors, since they offer a large variety of polymer architectures and functionalities, which are beneficial for specific demands in gene delivery. In combination with controlled radical polymerization techniques, such as the reversible addition-fragmentation chain transfer polymerization, the synthesis of well-defined polymers is possible. In this study we prepared a library of defined linear polymers based on (2-aminoethyl)-methacrylate (AEMA), N-methyl-(2-aminoethyl)-methacrylate (MAEMA), and N,N-dimethyl-(2-aminoethyl)-methacrylate (DMAEMA) monomers, bearing pendant primary, secondary, and tertiary amino groups, and investigated the influence of the substitution pattern on their gene delivery capability. The polymers and the corresponding plasmid DNA complexes were investigated regarding their physicochemical characteristics, cytocompatibility, and transfection performance. The nonviral transfection by methacrylate-based polyplexes differs significantly from poly(ethylene imine)-based polyplexes, as a successful transfection is not affected by the buffer capacity. We observed that polyplexes containing a high content of primary amino groups (AEMA) offered the highest transfection efficiency, whereas polyplexes bearing tertiary amino groups (DMAEMA) exhibited the lowest transfection efficiency. Further insights into the uptake and release mechanisms could be identified by fluorescence and transmission electron microscopy, emphasizing the theory of membrane-pore formation for the time-efficient endosomal release of methacrylate-based vectors.
Collapse
Affiliation(s)
- Anne-Kristin Trützschler
- Institue for Organic Chemistry and Macromolecular Chemistry , Friedrich Schiller University Jena , Humboldtstrasse 10 , 07743 Jena , Germany.,Jena Center for Soft Matter (JCSM) , Friedrich Schiller University Jena , Philosophenweg 7 , 07743 Jena , Germany
| | - Tanja Bus
- Institue for Organic Chemistry and Macromolecular Chemistry , Friedrich Schiller University Jena , Humboldtstrasse 10 , 07743 Jena , Germany.,Jena Center for Soft Matter (JCSM) , Friedrich Schiller University Jena , Philosophenweg 7 , 07743 Jena , Germany
| | - Martin Reifarth
- Institue for Organic Chemistry and Macromolecular Chemistry , Friedrich Schiller University Jena , Humboldtstrasse 10 , 07743 Jena , Germany.,Jena Center for Soft Matter (JCSM) , Friedrich Schiller University Jena , Philosophenweg 7 , 07743 Jena , Germany.,Institute of Physical Chemistry and Abbe Center of Photonics , Friedrich Schiller University Jena , Helmholtzweg 4 , 07743 Jena , Germany.,Leibniz Institute of Photonic Technology , Albert-Einstein-Strasse 9 , 07745 Jena , Germany
| | - Johannes C Brendel
- Institue for Organic Chemistry and Macromolecular Chemistry , Friedrich Schiller University Jena , Humboldtstrasse 10 , 07743 Jena , Germany.,Jena Center for Soft Matter (JCSM) , Friedrich Schiller University Jena , Philosophenweg 7 , 07743 Jena , Germany
| | - Stephanie Hoeppener
- Institue for Organic Chemistry and Macromolecular Chemistry , Friedrich Schiller University Jena , Humboldtstrasse 10 , 07743 Jena , Germany.,Jena Center for Soft Matter (JCSM) , Friedrich Schiller University Jena , Philosophenweg 7 , 07743 Jena , Germany
| | - Anja Traeger
- Institue for Organic Chemistry and Macromolecular Chemistry , Friedrich Schiller University Jena , Humboldtstrasse 10 , 07743 Jena , Germany.,Jena Center for Soft Matter (JCSM) , Friedrich Schiller University Jena , Philosophenweg 7 , 07743 Jena , Germany
| | - Ulrich S Schubert
- Institue for Organic Chemistry and Macromolecular Chemistry , Friedrich Schiller University Jena , Humboldtstrasse 10 , 07743 Jena , Germany.,Jena Center for Soft Matter (JCSM) , Friedrich Schiller University Jena , Philosophenweg 7 , 07743 Jena , Germany
| |
Collapse
|
34
|
Yang S, Meng Z, Kang Z, Sun C, Wang T, Feng S, Meng Q, Liu K. The structure and configuration changes of multifunctional peptide vectors enhance gene delivery efficiency. RSC Adv 2018; 8:28356-28366. [PMID: 35542475 PMCID: PMC9084241 DOI: 10.1039/c8ra04101f] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/23/2018] [Indexed: 11/25/2022] Open
Abstract
We designed a series of peptide vectors that contain functional fragments with the goal of enhancing cellular internalization and gene transfection efficiency. The functional fragments included a cell-penetrating peptide (R9), a cationic amphiphilic α-helical peptide [(LLKK)3-H6 or (LLHH)3], a stearyl moiety, and cysteine residues. Vectors were also synthesized with D-type amino acids to improve their proteolytic stability. The conformations, particle sizes, and zeta potentials for complexes of these peptides with pGL3 plasmid DNA were characterized by circular dichroism and dynamic light scattering. In addition, cellular uptake of the peptide/DNA complexes and gene transfection efficiency were investigated with fluorescence-activated cell sorting and confocal laser-scanning microscopy. Greater transfection efficiency was achieved with the vectors containing the R9 segment, and the efficiency was greater than Lipo2000. In addition, the D-type C18-c(llkk)3ch6-r9 had about 7 times and 5.5 times the transfection efficiency of Lipo2000 in 293T cells and NIH-3T3 cells at the N/P ratio of 6, respectively. Overall, the multifunctional peptide gene vectors containing the R9 segment exhibited enhanced cellular internalization, a high gene transfection efficiency, and low cytotoxicity. The R9 containing peptide vectors can improve the gene transfection efficiency.![]()
Collapse
Affiliation(s)
- Sen Yang
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| | - Zhao Meng
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| | - Ziyao Kang
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| | - Chao Sun
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| | - Taoran Wang
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| | - Siliang Feng
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| | - Qingbin Meng
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| | - Keliang Liu
- State Key Laboratory of Toxicology and Medical Countermeasures
- Beijing Institute of Pharmacology and Toxicology
- Beijing
- China
| |
Collapse
|
35
|
Bire S, Ishac N, Rouleux-Bonnin F. In Vitro Synthesis, Delivery, and Bioavailability of Exogenous mRNA in Gene Transfer Mediated by PiggyBac Transposition. Methods Mol Biol 2017; 1428:187-217. [PMID: 27236801 DOI: 10.1007/978-1-4939-3625-0_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nowadays, nonviral gene transfer is currently of great importance for introducing exogenous genes into genomes and for ensuring that transgene expression is suitable for therapeutic and bioproduction purposes. The piggyBac transposon-based system is particularly interesting since it is easy to engineer and has a large cargo capacity, up to 100 kb. In its setup, the system requires only the piggyBac transposase protein and the transgene delineated by the two piggyBac-specific inverted terminal repeats. Usually the source of transposase is carried by a DNA plasmid. However, the principal drawback of this method is the lasting presence of the transposase, due to episomal persistence or possible integration of the transposase gene vector into the cell's genome. This can lead to genotoxic effects such as multiple genomic integration events and remobilization of the transposon vector once it has been integrated. One alternative to improve the safety of the system is to deliver the transposase as in vitro-synthesized messenger RNA in order to define a very narrow expression window during which a one-shot transposition process would occur. Issues that can be encountered when working on mRNA cell transfer are related to the quality of the synthetic mRNA, the system used to introduce mRNA into the cells and the bioavailability of the mRNA molecules. Here we describe a method to produce mRNA, verify its quality, determine which transfecting reagents can be used and how this mRNA is available to promote the transposition process in HeLa cells. Additionally, we illustrate this method in stromal mesenchymal cell lines in order to support hematopoiesis.
Collapse
Affiliation(s)
- Solenne Bire
- LBTM, Institute of Biotechnology, UNIL-EPFL, Station 6, Lausanne, 1015, Switzerland
| | - Nicole Ishac
- LNOX, GICC UMR CNRS 7292, UFR de Médecine, Bâtiment Dutrochet, 10 Boulevard Tonnellé, Tours, 37032, France
| | - Florence Rouleux-Bonnin
- LNOX, GICC UMR CNRS 7292, UFR de Médecine, Bâtiment Dutrochet, 10 Boulevard Tonnellé, Tours, 37032, France.
| |
Collapse
|
36
|
Sokolova V, Shi Z, Huang S, Du Y, Kopp M, Frede A, Knuschke T, Buer J, Yang D, Wu J, Westendorf AM, Epple M. Delivery of the TLR ligand poly(I:C) to liver cells in vitro and in vivo by calcium phosphate nanoparticles leads to a pronounced immunostimulation. Acta Biomater 2017; 64:401-410. [PMID: 28963016 DOI: 10.1016/j.actbio.2017.09.037] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 09/24/2017] [Accepted: 09/25/2017] [Indexed: 01/01/2023]
Abstract
The selective activation of the immune system is a concurrent problem in the treatment of persistent diseases like viral infections (e.g. hepatitis). For the delivery of the toll-like receptor ligand poly(I:C), an immunostimulatory action was discovered earlier by hydrodynamic injection. However, this technique is not clinically transferable to human patients. A modular system where the immunoactive toll-like-receptor ligand 3 (TLR-3) poly(I:C) was incorporated into calcium phosphate nanoparticles was developed. The nanoparticles had a hydrodynamic diameter of 275nm and a zeta potential of +20mV, measured by dynamic light scattering. The diameter of the solid core was 120nm by scanning electron microscopy. In vitro, the nanoparticle uptake was investigated after 1 and 24h of incubation of THP-1 cells (macrophages) with nanoparticles by fluorescence microscopy. After intravenous injection into BALB/c and C57BL/6J mice, respectively, the in vivo uptake was especially prominent in lung and liver, 1 and 3h after the injection. Pronounced immunostimulatory effects of the nanoparticles were found in vitro with primary liver cells, i.e. Kupffer cells (KC) and liver sinusoidal endothelial cells (LSEC) from wild-type C57BL/6J mice. Thus, they represent a suitable alternative to hydrodynamic injection treatments for future vaccination concepts. STATEMENT OF SIGNIFICANCE The selective activation of the immune system is a concurrent problem in the treatment of persistent diseases like viral infections (e.g. hepatitis). For the delivery of the toll-like receptor ligand poly(I:C), an immunostimulatory action has been discovered earlier by hydrodynamic injection. However, this technique is not clinically transferable to human patients. We have developed a modular system where poly(I:C) was incorporated into calcium phosphate nanoparticles. The uptake into relevant liver cells was studied both in vitro and in vivo. After intravenous injection into mice, the in vivo uptake was especially prominent in lung and liver, 1 and 3h after the injection. The corresponding strong immune reaction proves their high potential to turn up the immune system, e.g. against viral infections, without adverse side reactions.
Collapse
Affiliation(s)
- Viktoriya Sokolova
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117 Essen, Germany
| | - Zou Shi
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, 430030 Wuhan, PR China
| | - Shunmei Huang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, 430030 Wuhan, PR China
| | - Yanqin Du
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, 430030 Wuhan, PR China
| | - Mathis Kopp
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117 Essen, Germany
| | - Annika Frede
- Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Torben Knuschke
- Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Jan Buer
- Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Dongliang Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, 430030 Wuhan, PR China
| | - Jun Wu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, 430030 Wuhan, PR China
| | - Astrid Maria Westendorf
- Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Matthias Epple
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117 Essen, Germany.
| |
Collapse
|
37
|
Fast therapeutic DNA internalization – A high potential transfection system based on a peptide mimicking cationic lipid. Eur J Pharm Biopharm 2017; 118:38-47. [DOI: 10.1016/j.ejpb.2016.12.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 11/21/2016] [Accepted: 12/14/2016] [Indexed: 02/08/2023]
|
38
|
Costa D, Costa C, Caldeira M, Cortes L, Queiroz JA, Cruz C. Targeting of Cellular Organelles by Fluorescent Plasmid DNA Nanoparticles. Biomacromolecules 2017; 18:2928-2936. [DOI: 10.1021/acs.biomac.7b00877] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Diana Costa
- CICS-UBI
− Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Carolina Costa
- CICS-UBI
− Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Margarida Caldeira
- Microscopy
Unit-CNC − Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - Luísa Cortes
- Microscopy
Unit-CNC − Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - João A. Queiroz
- CICS-UBI
− Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Carla Cruz
- CICS-UBI
− Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
39
|
Lu M, Xing H, Xun Z, Yang T, Ding P, Cai C, Wang D, Zhao X. Exosome-based small RNA delivery: Progress and prospects. Asian J Pharm Sci 2017; 13:1-11. [PMID: 32104373 PMCID: PMC7032220 DOI: 10.1016/j.ajps.2017.07.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/14/2017] [Accepted: 07/28/2017] [Indexed: 12/29/2022] Open
Abstract
RNA interfering (RNAi), mediated by small interfering RNAs and microRNAs, is currently one of the most promising tools of gene therapy. Small RNAs are capable of inducing specific post-transcriptional gene silencing, providing a potentially effective platform for the treatment of a wide array of diseases. However, similar to other nucleic acid-based drugs, the major hurdle of RNAi therapy is lack of efficient and non-immunogenic delivery vehicles. Currently, viruses, synthetic polymers, and lipid-based carriers are among the most widely studied vehicles for small RNA delivery. However, many drawbacks are reported to be associated with these delivery vehicles. There is a pressing need to replace them with more efficient and better-tolerated approaches. Exosomes secreted from the endocytic compartment of live cells, are a subtype of endogenous extracellular vesicles that transfer genetic and biochemical information among different cells, thus playing an important role in cell-cell communication. Recently, accumulating attention has been focused on harnessing exosomes as nanaocarriers for small RNAs delivery. Due to their natural role in shuttling endogenous nucleic acid in our body, exosomes may exhibit higher delivery efficiency, lower immunogenicity, and better compatibility than existing foreign RNA carriers. Importantly, exosomes own intrinsic homing capacity that can guide small RNAs across natural membranous barriers. Moreover, such a capacity can be further improved by adding appropriate targeting moieties. In this manuscript, we briefly review the progress and challenges of RNAi therapy, and discuss the potential of exosomes' applications in small RNA delivery with focus on the most recent advances in exosome-based small RNA delivery for disease therapy.
Collapse
Affiliation(s)
- Mei Lu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Haonan Xing
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhe Xun
- Institute of Metabolic Disease Research and Drug Development, China Medical University, Shenyang, China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME, USA
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Cuifang Cai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Dongkai Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaoyun Zhao
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
40
|
Aoyama M, Yoshioka Y, Arai Y, Hirai H, Ishimoto R, Nagano K, Higashisaka K, Nagai T, Tsutsumi Y. Intracellular trafficking of particles inside endosomal vesicles is regulated by particle size. J Control Release 2017; 260:183-193. [DOI: 10.1016/j.jconrel.2017.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 05/10/2017] [Accepted: 06/11/2017] [Indexed: 02/04/2023]
|
41
|
Dasari BC, Cashman SM, Kumar-Singh R. Reducible PEG-POD/DNA Nanoparticles for Gene Transfer In Vitro and In Vivo: Application in a Mouse Model of Age-Related Macular Degeneration. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:77-89. [PMID: 28918058 PMCID: PMC5491761 DOI: 10.1016/j.omtn.2017.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/08/2017] [Accepted: 06/08/2017] [Indexed: 02/03/2023]
Abstract
Non-viral gene delivery systems are being developed to address limitations of viral gene delivery. Many of these non-viral systems are modeled on the properties of viruses including cell surface binding, endocytosis, endosomal escape, and nuclear targeting. Most non-viral gene transfer systems exhibit little correlation between in vitro and in vivo efficiency, hampering a systematic approach to their development. Previously, we have described a 3.5 kDa peptide (peptide for ocular delivery [POD]) that targets cell surface sialic acid. When functionalized with polyethylene glycol (PEG) via a sulfhydryl group on the N-terminal cysteine of POD, PEG-POD could compact plasmid DNA, forming 120- to 180-nm homogeneous nanoparticles. PEG-POD enabled modest gene transfer and rescue of retinal degeneration in vivo. Systematic investigation of different stages of gene transfer by PEG-POD nanoparticles was hampered by their inability to deliver genes in vitro. Herein, we describe functionalization of POD with PEG using a reducible orthopyridyl disulfide bond. These reducible nanoparticles enabled gene transfer in vitro while retaining their in vivo gene transfer properties. These reducible PEG-POD nanoparticles were utilized to deliver human FLT1 to the retina in vivo, achieving a 50% reduction in choroidal neovascularization in a murine model of age-related macular degeneration.
Collapse
Affiliation(s)
- Bhanu Chandar Dasari
- Department of Developmental, Molecular, and Chemical Biology, Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Siobhan M Cashman
- Department of Developmental, Molecular, and Chemical Biology, Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Rajendra Kumar-Singh
- Department of Developmental, Molecular, and Chemical Biology, Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA.
| |
Collapse
|
42
|
Kopp M, Rotan O, Papadopoulos C, Schulze N, Meyer H, Epple M. Delivery of the autofluorescent protein R-phycoerythrin by calcium phosphate nanoparticles into four different eukaryotic cell lines (HeLa, HEK293T, MG-63, MC3T3): Highly efficient, but leading to endolysosomal proteolysis in HeLa and MC3T3 cells. PLoS One 2017; 12:e0178260. [PMID: 28586345 PMCID: PMC5460861 DOI: 10.1371/journal.pone.0178260] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023] Open
Abstract
Nanoparticles can be used as carriers to transport biomolecules like proteins and synthetic molecules across the cell membrane because many molecules are not able to cross the cell membrane on their own. The uptake of nanoparticles together with their cargo typically occurs via endocytosis, raising concerns about the possible degradation of the cargo in the endolysosomal system. As the tracking of a dye-labelled protein during cellular uptake and processing is not indicative of the presence of the protein itself but only for the fluorescent label, a label-free tracking was performed with the red-fluorescing model protein R-phycoerythrin (R-PE). Four different eukaryotic cell lines were investigated: HeLa, HEK293T, MG-63, and MC3T3. Alone, the protein was not taken up by any cell line; only with the help of calcium phosphate nanoparticles, an efficient uptake occurred. After the uptake into HeLa cells, the protein was found in early endosomes (shown by the marker EEA1) and lysosomes (shown by the marker Lamp1). There, it was still intact and functional (i.e. properly folded) as its red fluorescence was detected. However, a few hours after the uptake, proteolysis started as indicated by the decreasing red fluorescence intensity in the case of HeLa and MC3T3 cells. 12 h after the uptake, the protein was almost completely degraded in HeLa cells and MC3T3 cells. In HEK293T cells and MG-63 cells, no degradation of the protein was observed. In the presence of Bafilomycin A1, an inhibitor of acidification and protein degradation in lysosomes, the fluorescence of R-PE remained intact over the whole observation period in the four cell lines. These results indicate that despite an efficient nanoparticle-mediated uptake of proteins by cells, a rapid endolysosomal degradation may prevent the desired (e.g. therapeutic) effect of a protein inside a cell.
Collapse
Affiliation(s)
- Mathis Kopp
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Essen, Germany
| | - Olga Rotan
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Essen, Germany
| | | | - Nina Schulze
- Imaging Centre Campus Essen (ICCE), University of Duisburg-Essen, Essen, Germany
| | - Hemmo Meyer
- Centre for Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Matthias Epple
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
43
|
Albuquerque LJC, de Castro CE, Riske KA, da Silva MCC, Muraro PIR, Schmidt V, Giacomelli C, Giacomelli FC. Gene Transfection Mediated by Catiomers Requires Free Highly Charged Polymer Chains To Overcome Intracellular Barriers. Biomacromolecules 2017; 18:1918-1927. [PMID: 28453254 DOI: 10.1021/acs.biomac.7b00344] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The prospective use of the block copolymers poly(ethylene oxide)113-b-poly[2-(diethylamino)ethyl methacrylate]50 (PEO113-b-PDEA50) and poly[oligo(ethylene glycol)methyl ether methacrylate]70-b-poly[oligo(ethylene glycol)methyl ether methacrylate10-co-2-(diethylamino)ethyl methacrylate47-co-2-(diisopropylamino)ethyl methacrylate47] (POEGMA70-b-P(OEGMA10-co-DEA47-co-DPA47)) as nonviral gene vectors was evaluated. The polymers are able to properly condense DNA into nanosized particles (RH ≈ 75 nm), which are marginally cytotoxic and can be uptaken by cells. However, the green fluorescent protein (GFP) expression assays evidenced that DNA delivery is essentially negligible meaning that intracellular trafficking hampers efficient gene release. Subsequently, we demonstrate that cellular uptake and particularly the quantity of GFP-positive cells are substantially enhanced when the block copolymer polyplexes are produced and further supplemented by BPEI chains (branched polyethylenimine). The dynamic light scattering/electrophoretic light scattering/isothermal titration calorimetry data suggest that such a strategy allows the adsorption of BPEI onto the surface of the polyplexes, and this phenomenon is responsible for increasing the size and surface charge of the assemblies. Nevertheless, most of the BPEI chains remain freely diffusing in the systems. The biological assays confirmed that cellular uptake is enhanced in the presence of BPEI and principally, the free highly charged polymer chains play the central role in intracellular trafficking and gene transfection. These investigations pointed out that the transfection efficiency versus cytotoxicity issue can be balanced by a mixture of BPEI and less cytotoxic agents such as for instance the proposed block copolymers.
Collapse
Affiliation(s)
- Lindomar J C Albuquerque
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC , 09210-580 Santo André, Brazil
| | - Carlos E de Castro
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC , 09210-580 Santo André, Brazil
| | - Karin A Riske
- Departamento de Biofísica, Universidade Federal de São Paulo , 04023-062 São Paulo, Brazil
| | - Maria C Carlan da Silva
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC , 09210-580 Santo André, Brazil
| | - Paulo I R Muraro
- Departamento de Química, Universidade Federal de Santa Maria , 97105-900 Santa Maria, Brazil
| | - Vanessa Schmidt
- Departamento de Química, Universidade Federal de Santa Maria , 97105-900 Santa Maria, Brazil
| | - Cristiano Giacomelli
- Departamento de Química, Universidade Federal de Santa Maria , 97105-900 Santa Maria, Brazil
| | - Fernando C Giacomelli
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC , 09210-580 Santo André, Brazil
| |
Collapse
|
44
|
A Triple-Fluorophore-Labeled Nucleic Acid pH Nanosensor to Investigate Non-viral Gene Delivery. Mol Ther 2017; 25:1697-1709. [PMID: 28479046 DOI: 10.1016/j.ymthe.2017.04.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/06/2017] [Accepted: 04/06/2017] [Indexed: 01/08/2023] Open
Abstract
There is a need for new tools to better quantify intracellular delivery barriers in high-throughput and high-content ways. Here, we synthesized a triple-fluorophore-labeled nucleic acid pH nanosensor for measuring intracellular pH of exogenous DNA at specific time points in a high-throughput manner by flow cytometry following non-viral transfection. By including two pH-sensitive fluorophores and one pH-insensitive fluorophore in the nanosensor, detection of pH was possible over the full physiological range. We further assessed possible correlation between intracellular pH of delivered DNA, cellular uptake of DNA, and DNA reporter gene expression at 24 hr post-transfection for poly-L-lysine and branched polyethylenimine polyplex nanoparticles. While successful transfection was shown to clearly depend on median cellular pH of delivered DNA at the cell population level, surprisingly, on an individual cell basis, there was no significant correlation between intracellular pH and transfection efficacy. To our knowledge, this is the first reported instance of high-throughput single-cell analysis between cellular uptake of DNA, intracellular pH of delivered DNA, and gene expression of the delivered DNA. Using the nanosensor, we demonstrate that the ability of polymeric nanoparticles to avoid an acidic environment is necessary, but not sufficient, for successful transfection.
Collapse
|
45
|
A preliminary study on the interaction between Asn-Gly-Arg (NGR)-modified multifunctional nanoparticles and vascular epithelial cells. Acta Pharm Sin B 2017; 7:361-372. [PMID: 28540174 PMCID: PMC5430811 DOI: 10.1016/j.apsb.2017.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 11/29/2016] [Accepted: 12/29/2016] [Indexed: 01/21/2023] Open
Abstract
Previously developed Asn-Gly-Arg (NGR) peptide-modified multifunctional poly(ethyleneimine)–poly(ethylene glycol) (PEI–PEG)-based nanoparticles (TPIC) have been considered to be promising carriers for the co-delivery of DNA and doxorubicin (DOX). As a continued effort, the aim of the present study was to further evaluate the interaction between TPIC and human umbilical vein endothelial cells (HUVEC) to better understand the cellular entry mechanism. In the present investigation, experiments relevant to co-localization, endocytosis inhibitors and factors influencing the internalization were performed. Without any treatment, there was no co-localization between aminopeptidase N/CD13 (APN/CD13) and caveolin 1 (CAV1). However, co-localization between CD13 and CAV1 was observed when cells were incubated with an anti-CD13 antibody or TPIC. As compared with antibody treatment, TPIC accelerated the speed and enhanced the degree of co-localization. TPIC entered HUVEC not only together with CD13 but also together with CAV1. However, this internalization was not dependent on the enzyme activity of CD13 but could be inhibited by methyl-β-eyclodextfin (MβCD), further identifying the involvement of caveolae-mediated endocytosis (CvME). This conclusion was also verified by endocytosis inhibitor experiments.
Collapse
|
46
|
Martens TF, Peynshaert K, Nascimento TL, Fattal E, Karlstetter M, Langmann T, Picaud S, Demeester J, De Smedt SC, Remaut K, Braeckmans K. Effect of hyaluronic acid-binding to lipoplexes on intravitreal drug delivery for retinal gene therapy. Eur J Pharm Sci 2017; 103:27-35. [DOI: 10.1016/j.ejps.2017.02.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/14/2017] [Accepted: 02/16/2017] [Indexed: 11/17/2022]
|
47
|
Borgheti-Cardoso LN, Kooijmans SAA, Fens MHAM, van der Meel R, Vicentini FTMC, Fantini MCA, Bentley MVLB, Schiffelers RM. In Situ Gelling Liquid Crystalline System as Local siRNA Delivery System. Mol Pharm 2017; 14:1681-1690. [DOI: 10.1021/acs.molpharmaceut.6b01141] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Livia N. Borgheti-Cardoso
- School of Pharmaceutical
Sciences of Ribeirao Preto, University of São Paulo, Avenida do Café, s/n, 14040-903 Ribeirão Preto, São Paulo, Brazil
| | - Sander A. A. Kooijmans
- Laboratory
of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan, 100, 3584 CX Utrecht, The Netherlands
- Department of
Medical Sciences, The Camussi Laboratory, University of Torino, Via Nizza, 52, 10126 Torino, Italy
| | - Marcel H. A. M. Fens
- Laboratory
of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan, 100, 3584 CX Utrecht, The Netherlands
| | - Roy van der Meel
- Laboratory
of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan, 100, 3584 CX Utrecht, The Netherlands
- Department of Biochemistry
and Molecular Biology, University of British Columbia, 2350 Health
Sciences Mall, Vancouver, British Columbia, Canada, V6T 1Z3
| | - Fabiana T. M. C. Vicentini
- School of Pharmaceutical
Sciences of Ribeirao Preto, University of São Paulo, Avenida do Café, s/n, 14040-903 Ribeirão Preto, São Paulo, Brazil
| | - Marcia C. A. Fantini
- Instituto de Física, University of São Paulo, Rua do Matão, 1371, Butantã, 05508-090 São
Paulo, São Paulo, Brazil
| | - Maria Vitória L. B. Bentley
- School of Pharmaceutical
Sciences of Ribeirao Preto, University of São Paulo, Avenida do Café, s/n, 14040-903 Ribeirão Preto, São Paulo, Brazil
| | - Raymond M. Schiffelers
- Laboratory
of Clinical Chemistry and Haematology, University Medical Center Utrecht, Heidelberglaan, 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
48
|
Lazebnik M, Pack DW. Rapid and facile quantitation of polyplex endocytic trafficking. J Control Release 2016; 247:19-27. [PMID: 28043862 DOI: 10.1016/j.jconrel.2016.12.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/17/2016] [Accepted: 12/29/2016] [Indexed: 11/25/2022]
Abstract
Design of safe and effective synthetic nucleic acid delivery vectors such as polycation/DNA or polycation/siRNA complexes (polyplexes) will be facilitated by quantitative understanding of the mechanisms by which such materials escort cargo from the cell surface to the nucleus. In particular, the mechanisms of cellular internalization by various endocytosis pathways and subsequent endocytic vesicle trafficking have been shown to strongly affect nucleic acid delivery efficiency. Fluorescence microscopy and subcellular fractionation methods are commonly employed to follow intracellular trafficking of biomolecules and nanoparticulate delivery systems such as polyplexes. However, it is difficult to obtain quantitative data from microscopy and subcellular fractionation is experimentally difficult and low throughput. We have developed a method for quantifying the transport of polyplexes through important endocytic vesicles. The method is based on polymerization of 3,3'-diaminobenzidine by endocytosed horseradish peroxidase, causing an increase in the vesicle density, resistance to being solubilized by detergent and quenching of fluorophores within the vesicles, which makes them easy to separate and quantify. Using this method in HeLa cells, we have observed polyethylenimine/siRNA polyplexes initially appearing in early endosomes and rapidly moving to other compartments within 30min post-transfection. At the same time, we observed the kinetics of accumulation of the polyplexes in lysosomes at a similar rate. The results from the new method are consistent with similar measurements by confocal fluorescence microscopy and subcellular fractionation of endocytic vesicles on a Percoll gradient. The relative ease of this new method will aid investigation of gene delivery mechanisms by providing the means to rapidly quantify endocytic trafficking of polyplexes and other vectors.
Collapse
Affiliation(s)
- Mihael Lazebnik
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana, IL 61801, USA
| | - Daniel W Pack
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY 40506, USA; Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
49
|
Askarian S, Abnous K, Ayatollahi S, Farzad SA, Oskuee RK, Ramezani M. PAMAM-pullulan conjugates as targeted gene carriers for liver cell. Carbohydr Polym 2016; 157:929-937. [PMID: 27988010 DOI: 10.1016/j.carbpol.2016.10.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/09/2016] [Accepted: 10/10/2016] [Indexed: 01/07/2023]
Abstract
Targeted nano-carriers are highly needed to promote nucleic acid delivery into the specific cell for therapeutic approaches. Pullulan as a linear carbohydrate has an intrinsic liver targeting property interacting with asialoglycoprotein receptor (ASGPR) found on liver cells. In the present study, we developed polyamidoamine (PAMAM)-pullulan conjugates and investigated their targeting activity in delivering gene into liver cells. The particle size, zeta potential, buffering capacity and ethidium bromide exclusion assays of the conjugates were evaluated. The cytotoxicity and transfection efficiency of new derivatives were assessed following in vitro transfection of HepG2 (receptor positive) and N2A (receptor negative) cell lines. Size of conjugated polymers ranged between 118 and 184 nanometers and their cytotoxicity were similar to PAMAM. Among six produced nanocarriers, G4PU4 and G5PU4 enhanced transfection efficiency in HepG2 cells compared to unmodified PAMAM. Therefore, the PAMAM-pullulan derivatives seem to improve delivery of nucleic acids into the liver cells expressing asialoglycoprotein receptor with minimal transfection in non-targeted cells.
Collapse
Affiliation(s)
- Saeedeh Askarian
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Khalil Abnous
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Sara Ayatollahi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Sara Amel Farzad
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Reza Kazemi Oskuee
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
50
|
Delivery of RNAi Therapeutics to the Airways-From Bench to Bedside. Molecules 2016; 21:molecules21091249. [PMID: 27657028 PMCID: PMC6272875 DOI: 10.3390/molecules21091249] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 09/05/2016] [Accepted: 09/13/2016] [Indexed: 12/12/2022] Open
Abstract
RNA interference (RNAi) is a potent and specific post-transcriptional gene silencing process. Since its discovery, tremendous efforts have been made to translate RNAi technology into therapeutic applications for the treatment of different human diseases including respiratory diseases, by manipulating the expression of disease-associated gene(s). Similar to other nucleic acid-based therapeutics, the major hurdle of RNAi therapy is delivery. Pulmonary delivery is a promising approach of delivering RNAi therapeutics directly to the airways for treating local conditions and minimizing systemic side effects. It is a non-invasive route of administration that is generally well accepted by patients. However, pulmonary drug delivery is a challenge as the lungs pose a series of anatomical, physiological and immunological barriers to drug delivery. Understanding these barriers is essential for the development an effective RNA delivery system. In this review, the different barriers to pulmonary drug delivery are introduced. The potential of RNAi molecules as new class of therapeutics, and the latest preclinical and clinical studies of using RNAi therapeutics in different respiratory conditions are discussed in details. We hope this review can provide some useful insights for moving inhaled RNAi therapeutics from bench to bedside.
Collapse
|