1
|
Kont A, Mendonça MCP, Malanga M, Felegyi K, Lindsay A, Cronin MF, Cahill MR, O'Driscoll CM. Structure-activity relationship of modified amphiphilic cationic cyclodextrins for enhanced siRNA delivery. Int J Pharm 2024; 670:125107. [PMID: 39708848 DOI: 10.1016/j.ijpharm.2024.125107] [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: 11/01/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
The presence of multiple hydroxyl groups at positions C2, C3 and C6 on the cyclodextrin (CD) ring structure allows for extensive functionalisation, enabling the development of biomaterials with significant potential for therapeutic siRNA delivery. To identify structural modifications that enhance activity, a range of cationic amphiphilic CDs, including both β- and γ-CDs, were synthesised, compared and evaluated. Each CDs incorporated a C12 lipid chain on the primary face of the CD. On the secondary rim, at positions C2 and C3, either primary or tertiary amine groups with different pKa values were introduced via either a triazole or a thiopropyl linker. Nanoparticles (NPs) encapsulating small interfering RNA (siRNA) were formulated at mass ratios 7.5:1 and 10:1 (CD:siRNA) and their physicochemical properties evaluated. A comparative in vitro study was conducted to assess the gene silencing efficacy of these NPs using the luciferase reporter gene in A549-luc cells. Gene silencing levels for both β- and γ-CDs increased when modified with a primary amine compared to a tertiary amine group at position C2. Gene expression inhibition was further improved when the CDs were functionalized with amine functionalities at positions C2 and C3. Modification of the secondary side of γ-CDs with two sets of primary amine functionalities via a thiopropyl linker, as compared to a triazole linker, achieved up to 80% gene knockdown, regardless of dose. In conclusion, the structure-activity relationship study successfully identified CD modifications that enhance gene silencing efficacy.
Collapse
Affiliation(s)
- Ayse Kont
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork T12 YN60, Ireland
| | - Monique C P Mendonça
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork T12 YN60, Ireland
| | - Milo Malanga
- CarboHyde Zrt., Berlini str., 47-49, Budapest 1045, Hungary
| | | | - Andrew Lindsay
- School of Biochemistry and Cell Biology, Biosciences Institute, University College Cork, Cork T12 YN60, Ireland
| | - Michael F Cronin
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork T12 YN60, Ireland
| | - Mary R Cahill
- Department of Haematology and Cancer Research@UCC, Cork University Hospital, University College Cork, Cork T12 XF62, Ireland
| | - Caitriona M O'Driscoll
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork T12 YN60, Ireland.
| |
Collapse
|
2
|
De Martini LB, Sulmona C, Brambilla L, Rossi D. Cell-Penetrating Peptides as Valuable Tools for Nose-to-Brain Delivery of Biological Drugs. Cells 2023; 12:1643. [PMID: 37371113 DOI: 10.3390/cells12121643] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/13/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Due to their high specificity toward the target and their low toxicity, biological drugs have been successfully employed in a wide range of therapeutic areas. It is yet to be mentioned that biologics exhibit unfavorable pharmacokinetic properties, are susceptible to degradation by endogenous enzymes, and cannot penetrate biological barriers such as the blood-brain barrier (i.e., the major impediment to reaching the central nervous system (CNS)). Attempts to overcome these issues have been made by exploiting the intracerebroventricular and intrathecal routes of administration. The invasiveness and impracticality of these procedures has, however, prompted the development of novel drug delivery strategies including the intranasal route of administration. This represents a non-invasive way to achieve the CNS, reducing systemic exposure. Nonetheless, biotherapeutics strive to penetrate the nasal epithelium, raising the possibility that direct delivery to the nervous system may not be straightforward. To maximize the advantages of the intranasal route, new approaches have been proposed including the use of cell-penetrating peptides (CPPs) and CPP-functionalized nanosystems. This review aims at describing the most impactful attempts in using CPPs as carriers for the nose-to-brain delivery of biologics by analyzing their positive and negative aspects.
Collapse
Affiliation(s)
- Lisa Benedetta De Martini
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri-IRCCS, 27100 Pavia, Italy
| | - Claudia Sulmona
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri-IRCCS, 27100 Pavia, Italy
| | - Liliana Brambilla
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri-IRCCS, 27100 Pavia, Italy
| | - Daniela Rossi
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri-IRCCS, 27100 Pavia, Italy
| |
Collapse
|
3
|
Mehrabian A, Mashreghi M, Dadpour S, Badiee A, Arabi L, Hoda Alavizadeh S, Alia Moosavian S, Reza Jaafari M. Nanocarriers Call the Last Shot in the Treatment of Brain Cancers. Technol Cancer Res Treat 2022; 21:15330338221080974. [PMID: 35253549 PMCID: PMC8905056 DOI: 10.1177/15330338221080974] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Our brain is protected by physio-biological barriers. The blood–brain barrier (BBB) main mechanism of protection relates to the abundance of tight junctions (TJs) and efflux pumps. Although BBB is crucial for healthy brain protection against toxins, it also leads to failure in a devastating disease like brain cancer. Recently, nanocarriers have been shown to pass through the BBB and improve patients’ survival rates, thus becoming promising treatment strategies. Among nanocarriers, inorganic nanocarriers, solid lipid nanoparticles, liposomes, polymers, micelles, and dendrimers have reached clinical trials after delivering promising results in preclinical investigations. The size of these nanocarriers is between 10 and 1000 nm and is modified by surface attachment of proteins, peptides, antibodies, or surfactants. Multiple research groups have reported transcellular entrance as the main mechanism allowing for these nanocarriers to cross BBB. Transport proteins and transcellular lipophilic pathways exist in BBB for small and lipophilic molecules. Nanocarriers cannot enter via the paracellular route, which is limited to water-soluble agents due to the TJs and their small pore size. There are currently several nanocarriers in clinical trials for the treatment of brain cancer. This article reviews challenges as well as fitting attributes of nanocarriers for brain tumor treatment in preclinical and clinical studies.
Collapse
Affiliation(s)
- Amin Mehrabian
- School of Pharmacy, Biotechnology Research Center, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Warwick Medical School, University of Warwick, Coventry, UK
| | - Mohammad Mashreghi
- School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saba Dadpour
- School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Student Research Committee, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Alia Moosavian
- School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- School of Pharmacy, Biotechnology Research Center, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Al Qtaish N, Gallego I, Villate-Beitia I, Sainz-Ramos M, Martínez-Navarrete G, Soto-Sánchez C, Fernández E, Gálvez-Martín P, Lopez-Mendez TB, Puras G, Luis Pedraz J. Sphingolipid extracts enhance gene delivery of cationic lipid vesicles into retina and brain. Eur J Pharm Biopharm 2021; 169:103-112. [PMID: 34606927 DOI: 10.1016/j.ejpb.2021.09.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 11/15/2022]
Abstract
The aim was to evaluate relevant biophysic processes related to the physicochemical features and gene transfection mechanism when sphingolipids are incorporated into a cationic niosome formulation for non-viral gene delivery to central nervous system. For that, two formulations named niosphingosomes and niosomes devoid of sphingolipid extracts, as control, were developed by the oil-in water emulsion technique. Both formulations and the corresponding complexes, obtained upon the addition of the reporter EGFP plasmid, were physicochemically and biologically characterized and evaluated. Compared to niosomes, niosphingosomes, and the corresponding complexes decreased particle size and increased superficial charge. Although there were not significant differences in the cellular uptake, cell viability and transfection efficiency increased when human retinal pigment epithelial (ARPE-19) cells were exposed to niosphingoplexes. Endocytosis via caveolae decreased in the case of niosphingoplexes, which showed higher co-localization with lysosomal compartment, and endosomal escape properties. Moreover, niosphingoplexes transfected not only primary central nervous system cells, but also different cells in mouse retina, depending on the administration route, and brain cortex. These preliminary results suggest that niosphingosomes represent a promising non-viral vector formulation purposed for the treatment of both retinal and brain diseases by gene therapy approach.
Collapse
Affiliation(s)
- Nuseibah Al Qtaish
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain.
| | - Idoia Gallego
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain.
| | - Ilia Villate-Beitia
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain.
| | - Myriam Sainz-Ramos
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain.
| | - Gema Martínez-Navarrete
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Neuroprothesis and Neuroengineering Research Group, Institute of Bioengineering, Miguel Hernández University, Avenida de la Universidad, 03202 Elche, Spain.
| | - Cristina Soto-Sánchez
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Neuroprothesis and Neuroengineering Research Group, Institute of Bioengineering, Miguel Hernández University, Avenida de la Universidad, 03202 Elche, Spain.
| | - Eduardo Fernández
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Neuroprothesis and Neuroengineering Research Group, Institute of Bioengineering, Miguel Hernández University, Avenida de la Universidad, 03202 Elche, Spain.
| | | | - Tania B Lopez-Mendez
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain.
| | - Gustavo Puras
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain.
| | - José Luis Pedraz
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology. Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain.
| |
Collapse
|
5
|
How Far Are Non-Viral Vectors to Come of Age and Reach Clinical Translation in Gene Therapy? Int J Mol Sci 2021; 22:ijms22147545. [PMID: 34299164 PMCID: PMC8304344 DOI: 10.3390/ijms22147545] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 07/10/2021] [Indexed: 01/14/2023] Open
Abstract
Efficient delivery of genetic material into cells is a critical process to translate gene therapy into clinical practice. In this sense, the increased knowledge acquired during past years in the molecular biology and nanotechnology fields has contributed to the development of different kinds of non-viral vector systems as a promising alternative to virus-based gene delivery counterparts. Consequently, the development of non-viral vectors has gained attention, and nowadays, gene delivery mediated by these systems is considered as the cornerstone of modern gene therapy due to relevant advantages such as low toxicity, poor immunogenicity and high packing capacity. However, despite these relevant advantages, non-viral vectors have been poorly translated into clinical success. This review addresses some critical issues that need to be considered for clinical practice application of non-viral vectors in mainstream medicine, such as efficiency, biocompatibility, long-lasting effect, route of administration, design of experimental condition or commercialization process. In addition, potential strategies for overcoming main hurdles are also addressed. Overall, this review aims to raise awareness among the scientific community and help researchers gain knowledge in the design of safe and efficient non-viral gene delivery systems for clinical applications to progress in the gene therapy field.
Collapse
|
6
|
Grijalvo S, Ocampo SM, Perales JC, Eritja R. Preparation of Lipid-Conjugated siRNA Oligonucleotides for Enhanced Gene Inhibition in Mammalian Cells. Methods Mol Biol 2021; 2282:119-136. [PMID: 33928573 DOI: 10.1007/978-1-0716-1298-9_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nucleic acid conjugates are promising drugs for treating gene-related diseases. Conjugating specific units like lipids, cell-penetrating peptides, polymers, antibodies, and aptamers either at the 3'- or 5'-termini of a siRNA duplex molecule has resulted in a plethora of siRNA bioconjugates with improved stabilities in bloodstream and better pharmacokinetic values than unmodified siRNAs. In this sense, lipid-siRNA conjugates have attracted a remarkable interest for their potential value in facilitating cellular uptake. In this chapter, we describe a series of protocols involving the synthesis of siRNA oligonucleotides carrying either neutral or cationic lipids at the 3'- and 5'-termini. The resulting lipid-siRNA conjugates are aimed to be used as exogenous effectors for inhibiting gene expression by RNA interference. A protocol for the formulation of lipid siRNA using sonication in the presence of serum is described yielding interesting transfection properties for cell culture without the use of transfecting agents.
Collapse
Affiliation(s)
- Santiago Grijalvo
- Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER BBN), Barcelona, Spain. .,Institute for Advanced Chemistry of Catalonia (IQAC), Spanish Research Council (CSIC), Barcelona, Spain.
| | - Sandra M Ocampo
- Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER BBN), Barcelona, Spain.,Institute for Advanced Chemistry of Catalonia (IQAC), Spanish Research Council (CSIC), Barcelona, Spain.,Department of Physiologic Sciences, School of Medicine, University of Barcelona, Barcelona, Spain
| | - José Carlos Perales
- Department of Physiologic Sciences, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Ramon Eritja
- Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER BBN), Barcelona, Spain. .,Institute for Advanced Chemistry of Catalonia (IQAC), Spanish Research Council (CSIC), Barcelona, Spain.
| |
Collapse
|
7
|
Mendonça MCP, Kont A, Aburto MR, Cryan JF, O'Driscoll CM. Advances in the Design of (Nano)Formulations for Delivery of Antisense Oligonucleotides and Small Interfering RNA: Focus on the Central Nervous System. Mol Pharm 2021; 18:1491-1506. [PMID: 33734715 PMCID: PMC8824433 DOI: 10.1021/acs.molpharmaceut.0c01238] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
RNA-based therapeutics have emerged
as one of the most powerful
therapeutic options used for the modulation of gene/protein expression
and gene editing with the potential to treat neurodegenerative diseases.
However, the delivery of nucleic acids to the central nervous system
(CNS), in particular by the systemic route, remains a major hurdle.
This review will focus on the strategies for systemic delivery of
therapeutic nucleic acids designed to overcome these barriers. Pathways
and mechanisms of transport across the blood–brain barrier
which could be exploited for delivery are described, focusing in particular
on smaller nucleic acids including antisense oligonucleotides (ASOs)
and small interfering RNA (siRNA). Approaches used to enhance delivery
including chemical modifications, nanocarrier systems, and target
selection (cell-specific delivery) are critically analyzed. Learnings
achieved from a comparison of the successes and failures reported
for CNS delivery of ASOs versus siRNA will help identify opportunities
for a wider range of nucleic acids and accelerate the clinical translation
of these innovative therapies.
Collapse
Affiliation(s)
- Monique C P Mendonça
- Pharmacodelivery Group, School of Pharmacy, University College Cork, T12 YT20 Cork, Ireland
| | - Ayse Kont
- Pharmacodelivery Group, School of Pharmacy, University College Cork, T12 YT20 Cork, Ireland
| | - Maria Rodriguez Aburto
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, T12 XF62 Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, T12 XF62 Cork, Ireland
| | - Caitriona M O'Driscoll
- Pharmacodelivery Group, School of Pharmacy, University College Cork, T12 YT20 Cork, Ireland
| |
Collapse
|
8
|
Hamad MIK, Daoud S, Petrova P, Rabaya O, Jbara A, Melliti N, Stichmann S, Reiss G, Herz J, Förster E. Biolistic transfection and expression analysis of acute cortical slices. J Neurosci Methods 2020; 337:108666. [PMID: 32119875 DOI: 10.1016/j.jneumeth.2020.108666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 02/21/2020] [Accepted: 02/27/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Biolistic gene gun transfection has been used to transfect organotypic cultures (OTCs) or dissociated cultures in vitro. Here, we modified this technique to allow successful transfection of acute brain slices, followed by measurement of neuronal activity within a few hours. NEW METHOD We established biolistic transfection of murine acute cortical slices to measure calcium signals. Acute slices are mounted on plasma/thrombin coagulate and transfected with a calcium sensor. Imaging can be performed within 4 h post transfection without affecting cell viability. RESULTS Four hours after GCaMP6s transfection, acute slices display remarkable fluorescent protein expression level allowing to study spontaneous activity and receptor pharmacology. While optimal gas pressure (150 psi) and gold particle size used (1 μm) confirm previously published protocols, the amount of 5 μg DNA was found to be optimal for particle coating. COMPARISON WITH EXISTING METHODS The major advantage of this technique is the rapid disposition of acute slices for calcium imaging. No transgenic GECI expressing animals or OTC for long periods are required. In acute slices, network interaction and connectivity are preserved. The method allows to obtain physiological readouts within 4 h, before functional tissue modifications might come into effect. Limitations of this technique are random transfection, low expression efficiency when using specific promotors, and preclusion or genetic manipulations that require a prolonged time before physiological changes become measurable, such as expression of recombinant proteins that require transport to distant subcellular localizations. CONCLUSION The method is optimal for short-time investigation of calcium signals in acute slices.
Collapse
Affiliation(s)
- Mohammad I K Hamad
- Institute for Anatomy and Clinical Morphology, School of Medicine, Faculty of Health, University of Witten/Herdecke, Witten, Germany; Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany.
| | - Solieman Daoud
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| | - Petya Petrova
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| | - Obada Rabaya
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| | - Abdalrahim Jbara
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| | - Nesrine Melliti
- Institute for Anatomy and Clinical Morphology, School of Medicine, Faculty of Health, University of Witten/Herdecke, Witten, Germany
| | - Sarah Stichmann
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| | - Gebhard Reiss
- Institute for Anatomy and Clinical Morphology, School of Medicine, Faculty of Health, University of Witten/Herdecke, Witten, Germany
| | - Joachim Herz
- Departments of Molecular Genetics, Neuroscience, Neurology and Neurotherapeutics, Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| |
Collapse
|
9
|
AL Qtaish N, Gallego I, Villate-Beitia I, Sainz-Ramos M, López-Méndez TB, Grijalvo S, Eritja R, Soto-Sánchez C, Martínez-Navarrete G, Fernández E, Puras G, Pedraz JL. Niosome-Based Approach for In Situ Gene Delivery to Retina and Brain Cortex as Immune-Privileged Tissues. Pharmaceutics 2020; 12:E198. [PMID: 32106545 PMCID: PMC7150807 DOI: 10.3390/pharmaceutics12030198] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/21/2020] [Accepted: 02/21/2020] [Indexed: 01/02/2023] Open
Abstract
Non-viral vectors have emerged as a promising alternative to viral gene delivery systems due to their safer profile. Among non-viral vectors, recently, niosomes have shown favorable properties for gene delivery, including low toxicity, high stability, and easy production. The three main components of niosome formulations include a cationic lipid that is responsible for the electrostatic interactions with the negatively charged genetic material, a non-ionic surfactant that enhances the long-term stability of the niosome, and a helper component that can be added to improve its physicochemical properties and biological performance. This review is aimed at providing recent information about niosome-based non-viral vectors for gene delivery purposes. Specially, we will discuss the composition, preparation methods, physicochemical properties, and biological evaluation of niosomes and corresponding nioplexes that result from the addition of the genetic material onto their cationic surface. Next, we will focus on the in situ application of such niosomes to deliver the genetic material into immune-privileged tissues such as the brain cortex and the retina. Finally, as future perspectives, non-invasive administration routes and different targeting strategies will be discussed.
Collapse
Affiliation(s)
- Nuseibah AL Qtaish
- NanoBioCel group, University of the Basque Country (UPV/EHU), E-01006 Vitoria-Gasteiz, Spain; (N.A.Q.); (I.G.); (I.V.-B.); (M.S.-R.); (T.B.L.-M.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-01006 Vitoria-Gasteiz, Spain
| | - Idoia Gallego
- NanoBioCel group, University of the Basque Country (UPV/EHU), E-01006 Vitoria-Gasteiz, Spain; (N.A.Q.); (I.G.); (I.V.-B.); (M.S.-R.); (T.B.L.-M.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-01006 Vitoria-Gasteiz, Spain
| | - Ilia Villate-Beitia
- NanoBioCel group, University of the Basque Country (UPV/EHU), E-01006 Vitoria-Gasteiz, Spain; (N.A.Q.); (I.G.); (I.V.-B.); (M.S.-R.); (T.B.L.-M.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-01006 Vitoria-Gasteiz, Spain
| | - Myriam Sainz-Ramos
- NanoBioCel group, University of the Basque Country (UPV/EHU), E-01006 Vitoria-Gasteiz, Spain; (N.A.Q.); (I.G.); (I.V.-B.); (M.S.-R.); (T.B.L.-M.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-01006 Vitoria-Gasteiz, Spain
| | - Tania Belén López-Méndez
- NanoBioCel group, University of the Basque Country (UPV/EHU), E-01006 Vitoria-Gasteiz, Spain; (N.A.Q.); (I.G.); (I.V.-B.); (M.S.-R.); (T.B.L.-M.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-01006 Vitoria-Gasteiz, Spain
| | - Santiago Grijalvo
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-08034 Barcelona, Spain; (S.G.); (R.E.)
- Institute for Advanced Chemistry of Catalonia, (IQAC-CSIC), E-08034 Barcelona, Spain
| | - Ramón Eritja
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-08034 Barcelona, Spain; (S.G.); (R.E.)
- Institute for Advanced Chemistry of Catalonia, (IQAC-CSIC), E-08034 Barcelona, Spain
| | - Cristina Soto-Sánchez
- Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, E-03202 Elche, Spain; (C.S.-S.); (G.M.-N.); (E.F.)
| | - Gema Martínez-Navarrete
- Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, E-03202 Elche, Spain; (C.S.-S.); (G.M.-N.); (E.F.)
- Networking Research Centre for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-03202 Elche, Spain
| | - Eduardo Fernández
- Neuroprothesis and Neuroengineering Research Group, Miguel Hernández University, E-03202 Elche, Spain; (C.S.-S.); (G.M.-N.); (E.F.)
- Networking Research Centre for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-03202 Elche, Spain
| | - Gustavo Puras
- NanoBioCel group, University of the Basque Country (UPV/EHU), E-01006 Vitoria-Gasteiz, Spain; (N.A.Q.); (I.G.); (I.V.-B.); (M.S.-R.); (T.B.L.-M.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-01006 Vitoria-Gasteiz, Spain
| | - José Luis Pedraz
- NanoBioCel group, University of the Basque Country (UPV/EHU), E-01006 Vitoria-Gasteiz, Spain; (N.A.Q.); (I.G.); (I.V.-B.); (M.S.-R.); (T.B.L.-M.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-01006 Vitoria-Gasteiz, Spain
| |
Collapse
|
10
|
Nanomaterials for direct and indirect immunomodulation: A review of applications. Eur J Pharm Sci 2020; 142:105139. [DOI: 10.1016/j.ejps.2019.105139] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/14/2019] [Accepted: 11/03/2019] [Indexed: 01/03/2023]
|
11
|
Singh RP, Hidalgo T, Cazade PA, Darcy R, Cronin MF, Dorin I, O’Driscoll CM, Thompson D. Self-Assembled Cationic β-Cyclodextrin Nanostructures for siRNA Delivery. Mol Pharm 2019; 16:1358-1366. [DOI: 10.1021/acs.molpharmaceut.8b01307] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | - Tania Hidalgo
- School of Pharmacy, Cavanagh Pharmacy Building, University College Cork, Cork, Ireland
| | - Pierre-Andre Cazade
- Department of Physics, Bernal Institute, University of Limerick, V94 T9PX Limerick, Ireland
| | - Raphael Darcy
- School of Pharmacy, Cavanagh Pharmacy Building, University College Cork, Cork, Ireland
| | - Michael F. Cronin
- School of Pharmacy, Cavanagh Pharmacy Building, University College Cork, Cork, Ireland
| | - Irina Dorin
- Malvern Panalytical Ltd., Grovewood Road, Malvern, Worcestershire WR14 1XZ, U.K
| | | | - Damien Thompson
- Department of Physics, Bernal Institute, University of Limerick, V94 T9PX Limerick, Ireland
| |
Collapse
|
12
|
Yang J, Zhang J, Liu Y, Shi Z, Han H, Li Q. Phenylboronic acid-modified polyamidoamine-mediated delivery of short GC rich DNA for hepatocarcinoma gene therapy. Biomater Sci 2019; 7:3348-3358. [DOI: 10.1039/c9bm00394k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Phenylboronic acid was introduced on the surface of polyamidoamine to construct a derivative PP, which was further used as a tumor-targeting carrier for realizing the delivery of short GC rich DNA (GCD).
Collapse
Affiliation(s)
- Jiebing Yang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education
- School of Life Sciences
- Jilin University
- Changchun 130012
- China
| | - Jiayuan Zhang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education
- School of Life Sciences
- Jilin University
- Changchun 130012
- China
| | - Yong Liu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education
- School of Life Sciences
- Jilin University
- Changchun 130012
- China
| | - Zhiyuan Shi
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education
- School of Life Sciences
- Jilin University
- Changchun 130012
- China
| | - Haobo Han
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education
- School of Life Sciences
- Jilin University
- Changchun 130012
- China
| | - Quanshun Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education
- School of Life Sciences
- Jilin University
- Changchun 130012
- China
| |
Collapse
|
13
|
Nanodelivery systems for overcoming limited transportation of therapeutic molecules through the blood-brain barrier. Future Med Chem 2018; 10:2659-2674. [PMID: 30499740 DOI: 10.4155/fmc-2018-0208] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Due to the impermeable structure and barrier function of the blood-brain barrier (BBB), the delivery of therapeutic molecules into the CNS is extremely limited. Nanodelivery systems are regarded as the most effective and versatile carriers for the CNS, as they can transport cargo molecules across the BBB via various mechanisms. This review emphasizes the multi-functionalization strategies of nanodelivery systems and combinatorial approaches for the delivery of therapeutic drugs and genes into the CNS. The characteristics and functions of the BBB and underlying mechanisms of molecular translocation across the BBB are also described.
Collapse
|
14
|
Madrid M, Saklayen N, Shen W, Huber M, Vogel N, Mazur E. Laser-Activated Self-Assembled Thermoplasmonic Nanocavity Substrates for Intracellular Delivery. ACS APPLIED BIO MATERIALS 2018; 1:1793-1799. [DOI: 10.1021/acsabm.8b00447] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | | | | | - Marinus Huber
- Department of Physics, Ludwig Maximilian University of Munich, Munich, 80539, Germany
| | - Nicolas Vogel
- Institute of Particle Technology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, 91058, Germany
| | | |
Collapse
|
15
|
Lossi L, Merighi A. The Use of ex Vivo Rodent Platforms in Neuroscience Translational Research With Attention to the 3Rs Philosophy. Front Vet Sci 2018; 5:164. [PMID: 30073174 PMCID: PMC6060265 DOI: 10.3389/fvets.2018.00164] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/29/2018] [Indexed: 01/08/2023] Open
Abstract
The principles of the 3Rs—Replacement, Reduction, and Refinement—are at the basis of most advanced national and supranational (EU) regulations on animal experimentation and welfare. In the perspective to reduce and refine the use of these animals in translational research, we here discuss the use of rodent acute and organotypically cultured central nervous system slices. We describe novel applications of these ex vivo platforms in medium-throughput screening of neuroactive molecules of potential pharmacological interest, with particular attention to more recent developments that permit to fully exploit the potential of direct genetic engineering of organotypic cultures using transfection techniques. We then describe the perspectives for expanding the use ex vivo platforms in neuroscience studies under the 3Rs philosophy using the following approaches: (1) Use of co-cultures of two brain regions physiologically connected to each other (source-target) to analyze axon regeneration and reconstruction of circuitries; (2) Microinjection or co-cultures of primary cells and/or cell lines releasing one or more neuroactive molecules to screen their physiological and/or pharmacological effects onto neuronal survival and slice circuitry. Microinjected or co-cultured cells are ideally made fluorescent after transfection with a plasmid construct encoding green or red fluorescent protein under the control of a general promoter such as hCMV; (3) Use of “sniffer” cells sensing the release of biologically active molecules from organotypic cultures by means of fluorescent probes. These cells can be prepared with activatable green fluorescent protein, a unique chromophore that remains in a “dark” state because its maturation is inhibited, and can be made fluorescent (de-quenched) if specific cellular enzymes, such as proteases or kinases, are activated.
Collapse
Affiliation(s)
- Laura Lossi
- Laboratory of Neurobiology, Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Adalberto Merighi
- Laboratory of Neurobiology, Department of Veterinary Sciences, University of Turin, Turin, Italy
| |
Collapse
|
16
|
Targeted Drug Delivery via Folate Receptors for the Treatment of Brain Cancer: Can the Promise Deliver? J Pharm Sci 2017; 106:3413-3420. [DOI: 10.1016/j.xphs.2017.08.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/14/2017] [Accepted: 08/14/2017] [Indexed: 12/25/2022]
|
17
|
Peng J, Rao Y, Yang X, Jia J, Wu Y, Lu J, Tao Y, Tu W. Targeting neuronal nitric oxide synthase by a cell penetrating peptide Tat-LK15/siRNA bioconjugate. Neurosci Lett 2017; 650:153-160. [PMID: 28450191 DOI: 10.1016/j.neulet.2017.04.045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/11/2017] [Accepted: 04/21/2017] [Indexed: 02/01/2023]
Abstract
We developed a cell penetrating peptide (CPP) Tat-LK15, as a siRNA carrier to target nNOS. The feasibility, stability, efficiency and selectivity of this peptide-siRNA complex were evaluated in rat neuronal cells. We also compared the new method with conventional siRNA carrier Lipofectamine™. It was found that the CPP Tat-LK15 effectively and specifically delivered nNOS-siRNA into Rat retinal ganglia (RGC-5) cells and silenced the expression of nNOS. The CPP Tat-LK15 can conjugate with siRNA to form stable complex at a ratio of 2:1 (peptide/siRNA, w/w), which maintained stable in serum for as long as 4h. The CPP Tat-LK15 was low-toxicity to cells, as the apoptosis rate of treat cells was not increased significantly when the used peptide lower than 10μg/mL. Moreover, the cellular uptake of nNOS siRNA by Rat Neurons-dorsal spinal cord (RNdsc) cells was also significantly more than naked siRNA by RNdsc cells. The CPP Tat-LK15 was an efficient and stable, and non-cytotoxic siRNA delivery to neurons and effectively silenced the nNOS expression. The CPP Tat-LK15 mediated siRNA delivery was a potential tool to treat neuropathic diseases involving NO or nNOS neurotoxic cascades.
Collapse
Affiliation(s)
- Jie Peng
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China
| | - Yun Rao
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China; Department of Anesthesiology, Jiangxi Maternal and Child Health Hospital, Nanchang, 330000, Jiangxi Province, China
| | - Xue Yang
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China; Department of Anesthesiology, Second Affiliated Hospital, Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510120, Guangdong Province, China
| | - Ji Jia
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China
| | - Youping Wu
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China
| | - Jianhua Lu
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China.
| | - Yuanxiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07013, USA
| | - Weifeng Tu
- Department of Anesthesiology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, Guangdong Province, China
| |
Collapse
|
18
|
Brain-Targeted Polymers for Gene Delivery in the Treatment of Brain Diseases. Top Curr Chem (Cham) 2017; 375:48. [PMID: 28397188 DOI: 10.1007/s41061-017-0138-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/27/2017] [Indexed: 10/19/2022]
Abstract
Gene therapies have become a promising strategy for treating neurological disorders, such as brain cancer and neurodegenerative diseases, with the help of molecular biology interpreting the underlying pathological mechanisms. Successful cellular manipulation against these diseases requires efficient delivery of nucleic acids into brain and further into specific neurons or cancer cells. Compared with viral vectors, non-viral polymeric carriers provide a safer and more flexible way of gene delivery, although suffering from significantly lower transfection efficiency. Researchers have been devoted to solving this defect, which is attributed to the multiple barriers existing for gene therapeutics in vivo, such as systemic degradation, blood-brain barrier, and endosome trapping. This review will be mainly focused on systemically administrated brain-targeted polymers developed so far, including PEI, dendrimers, and synthetic polymers with various functions. We will discuss in detail how they are designed to overcome these barriers and how they efficiently deliver therapeutic nucleic acids into targeted cells.
Collapse
|
19
|
Wang F, Gao L, Meng LY, Xie JM, Xiong JW, Luo Y. A Neutralized Noncharged Polyethylenimine-Based System for Efficient Delivery of siRNA into Heart without Toxicity. ACS APPLIED MATERIALS & INTERFACES 2016; 8:33529-33538. [PMID: 27960377 DOI: 10.1021/acsami.6b13295] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Cationic polymers constitute an important class of materials in development of delivery vehicles for nucleic acid-based therapeutics. Among them, polyethylenimine (PEI) has been a classical cationic carrier intensively studied for therapeutic delivery of DNA, RNA, and short RNA molecules to treat diseases. However, the development of PEI for in vivo applications has been hampered by the inherent problems associated with the material, particularly its cytotoxicity and the instability of the nucleic acid complexation systems formed via electrostatic interactions. Here, we demonstrate a strategy to modify PEI polymers via hydrazidation to create neutralized, stable, and multifunctional system for delivering siRNA molecules. Through substitution of the primary amino groups of PEI with neutral hydrazide groups, cross-linked nanoparticles with surface decorated with a model targeting ligands were generated. The neutral cross-linked siRNA nanoparticles not only showed favorable biocompatibility and cell internalization efficiency in vitro but also allowed for significant tissue uptake and gene silencing efficiency in zebrafish heart in vivo. Our study suggests transformation of conventional branched PEI into a neutral polymer that can lead to a new category of nonviral carriers, and the resulting functional delivery systems may be further explored for development of siRNA therapeutics for treating cardiovascular disease/injury.
Collapse
Affiliation(s)
- Fang Wang
- Department of Biomedical Engineering, College of Engineering, ‡Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, and §School of Life Sciences, Peking University , Beijing, China 100871
| | - Lu Gao
- Department of Biomedical Engineering, College of Engineering, ‡Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, and §School of Life Sciences, Peking University , Beijing, China 100871
| | - Liu-Yi Meng
- Department of Biomedical Engineering, College of Engineering, ‡Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, and §School of Life Sciences, Peking University , Beijing, China 100871
| | - Jing-Ming Xie
- Department of Biomedical Engineering, College of Engineering, ‡Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, and §School of Life Sciences, Peking University , Beijing, China 100871
| | - Jing-Wei Xiong
- Department of Biomedical Engineering, College of Engineering, ‡Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, and §School of Life Sciences, Peking University , Beijing, China 100871
| | - Ying Luo
- Department of Biomedical Engineering, College of Engineering, ‡Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, and §School of Life Sciences, Peking University , Beijing, China 100871
| |
Collapse
|
20
|
Chang MF, Hsieh JH, Chiang H, Kan HW, Huang CM, Chellis L, Lin BS, Miaw SC, Pan CL, Chao CC, Hsieh ST. Effective gene expression in the rat dorsal root ganglia with a non-viral vector delivered via spinal nerve injection. Sci Rep 2016; 6:35612. [PMID: 27748450 PMCID: PMC5066268 DOI: 10.1038/srep35612] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 10/04/2016] [Indexed: 12/13/2022] Open
Abstract
Delivering gene constructs into the dorsal root ganglia (DRG) is a powerful but challenging therapeutic strategy for sensory disorders affecting the DRG and their peripheral processes. The current delivery methods of direct intra-DRG injection and intrathecal injection have several disadvantages, including potential injury to DRG neurons and low transfection efficiency, respectively. This study aimed to develop a spinal nerve injection strategy to deliver polyethylenimine mixed with plasmid (PEI/DNA polyplexes) containing green fluorescent protein (GFP). Using this spinal nerve injection approach, PEI/DNA polyplexes were delivered to DRG neurons without nerve injury. Within one week of the delivery, GFP expression was detected in 82.8% ± 1.70% of DRG neurons, comparable to the levels obtained by intra-DRG injection (81.3% ± 5.1%, p = 0.82) but much higher than those obtained by intrathecal injection. The degree of GFP expression by neurofilament(+) and peripherin(+) DRG neurons was similar. The safety of this approach was documented by the absence of injury marker expression, including activation transcription factor 3 and ionized calcium binding adaptor molecule 1 for neurons and glia, respectively, as well as the absence of behavioral changes. These results demonstrated the efficacy and safety of delivering PEI/DNA polyplexes to DRG neurons via spinal nerve injection.
Collapse
Affiliation(s)
- Ming-Fong Chang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Jung-Hsien Hsieh
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Departments of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Hao Chiang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Hung-Wei Kan
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Cho-Min Huang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Luke Chellis
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, USA
| | - Bo-Shiou Lin
- Department of Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Shi-Chuen Miaw
- Department of Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chun-Liang Pan
- Department of Graduate Institute of Molecular Medicine, College of Medicine, National Taiwan University, No. 7 Chung-Shan South Road, Taipei, 10002, Taiwan
| | - Chi-Chao Chao
- Departments of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Sung-Tsang Hsieh
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Graduate Institute of Brain and Mind Science, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Departments of Clinical Center for Neuroscience and Behavior, National Taiwan University Hospital, Taipei, Taiwan
- Department of Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| |
Collapse
|
21
|
Malhotra M, Toulouse A, Godinho BMDC, Mc Carthy DJ, Cryan JF, O'Driscoll CM. RNAi therapeutics for brain cancer: current advancements in RNAi delivery strategies. MOLECULAR BIOSYSTEMS 2016; 11:2635-57. [PMID: 26135606 DOI: 10.1039/c5mb00278h] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Malignant primary brain tumors are aggressive cancerous cells that invade the surrounding tissues of the central nervous system. The current treatment options for malignant brain tumors are limited due to the inability to cross the blood-brain barrier. The advancements in current research has identified and characterized certain molecular markers that are essential for tumor survival, progression, metastasis and angiogenesis. These molecular markers have served as therapeutic targets for the RNAi based therapies, which enable site-specific silencing of the gene responsible for tumor proliferation. However, to bring about therapeutic success, an efficient delivery carrier that can cross the blood-brain barrier and reach the targeted site is essential. The current review focuses on the potential of targeted, non-viral and viral particles containing RNAi therapeutic molecules as delivery strategies specifically for brain tumors.
Collapse
Affiliation(s)
- Meenakshi Malhotra
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | | | | | | | | | | |
Collapse
|
22
|
Mastorakos P, Song E, Zhang C, Berry S, Park HW, Kim YE, Park JS, Lee S, Suk JS, Hanes J. Biodegradable DNA Nanoparticles that Provide Widespread Gene Delivery in the Brain. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:678-85. [PMID: 26680637 PMCID: PMC4913277 DOI: 10.1002/smll.201502554] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/16/2015] [Indexed: 05/26/2023]
Abstract
Successful gene therapy of neurological disorders is predicated on achieving widespread and uniform transgene expression throughout the affected disease area in the brain. However, conventional gene vectors preferentially travel through low-resistance perivascular spaces and/or are confined to the administration site even with the aid of a pressure-driven flow provided by convection-enhanced delivery. Biodegradable DNA nanoparticles offer a safe gene delivery platform devoid of adverse effects associated with virus-based or synthetic nonbiodegradable systems. Using a state-of-the-art biodegradable polymer, poly(β-amino ester), colloidally stable sub-100 nm DNA nanoparticles are engineered with a nonadhesive polyethylene glycol corona that are able to avoid the adhesive and steric hindrances imposed by the extracellular matrix. Following convection enhanced delivery, these brain-penetrating nanoparticles are able to homogeneously distribute throughout the rodent striatum and mediate widespread and high-level transgene expression. These nanoparticles provide a biodegradable DNA nanoparticle platform enabling uniform transgene expression patterns in vivo and hold promise for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Panagiotis Mastorakos
- Center for Nanomedicine, at the Wilmer Eye Institute, Johns Hopkins School of Medicine, 400 N. Broadway, Baltimore, MD 21231, USA
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21297, USA
| | - Eric Song
- Center for Nanomedicine, at the Wilmer Eye Institute, Johns Hopkins School of Medicine, 400 N. Broadway, Baltimore, MD 21231, USA
- Center for Biotechnology Education, Krieger School of Arts and Sciences, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Clark Zhang
- Center for Nanomedicine, at the Wilmer Eye Institute, Johns Hopkins School of Medicine, 400 N. Broadway, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Av., Baltimore, MD 21205, USA
| | - Sneha Berry
- Center for Nanomedicine, at the Wilmer Eye Institute, Johns Hopkins School of Medicine, 400 N. Broadway, Baltimore, MD 21231, USA
- Center for Biotechnology Education, Krieger School of Arts and Sciences, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Hee Won Park
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Young Eun Kim
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Jong Sung Park
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, 601 N. Caroline St, Baltimore, MD 21287, USA
| | - Seulki Lee
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, 601 N. Caroline St, Baltimore, MD 21287, USA
| | | | | |
Collapse
|
23
|
An S, He D, Wagner E, Jiang C. Peptide-like Polymers Exerting Effective Glioma-Targeted siRNA Delivery and Release for Therapeutic Application. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015. [PMID: 26222334 DOI: 10.1002/smll.201501167] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Lipopolymer 49, a solid-phase synthesized T-shaped peptide-like oligoamide containing two central oleic acids, 20 aminoethane, and two terminal cysteine units, is identified as very potent and biocompatible small interfering RNA (siRNA) carrier for gene silencing in glioma cells. This carrier is combined with a novel targeting polymer 727, containing a precise sequence of Angiopep 2 targeting peptide, linked with 28 monomer units of ethylene glycol, 40 aminoethane, and two terminal cysteines in siRNA complex formation. Angiopep-polyethylene glycol (PEG)/siRNA polyplexes exhibit good nanoparticle features, effective glioma-targeting siRNA delivery, and intracellular siRNA release, resulting in an outstanding gene downregulation both in glioma cells and upon intravenous delivery in glioma model nude mice without significant biotoxicity. Therefore, this novel siRNA delivery system is expected to be a promising strategy for targeted and safe glioma therapy.
Collapse
Affiliation(s)
- Sai An
- Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | - Dongsheng He
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-University Munich, Butenandtstrasse 5-13, 81377, Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for Nanoscience, Ludwig-Maximilians-University Munich, Butenandtstrasse 5-13, 81377, Munich, Germany
| | - Chen Jiang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| |
Collapse
|
24
|
Rahme K, Guo J, Holmes JD, O'Driscoll CM. Evaluation of the physicochemical properties and the biocompatibility of polyethylene glycol-conjugated gold nanoparticles: A formulation strategy for siRNA delivery. Colloids Surf B Biointerfaces 2015; 135:604-612. [PMID: 26322474 DOI: 10.1016/j.colsurfb.2015.08.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 08/14/2015] [Accepted: 08/20/2015] [Indexed: 02/07/2023]
Abstract
The potential of RNA interference (RNAi)-based therapeutics for cancer has received much attention; however, delivery of RNAi effectors, such as small interfering RNA (siRNA), remains an obstacle to clinical translation. Non-viral delivery vectors have been used extensively to enhance siRNA delivery. Recently, the potential of gold nanoparticles (AuNPs) for transporting drugs, proteins and genetic materials has been demonstrated. Previously, our laboratory synthesised positively charged, surfactant-free AuNPs in water by the reduction of gold (III) chloride (AuCl3) using hydroxylamine hydrochloride (NH2OH·HCl) in the presence of L-cysteine methyl ester hydrochloride (HSCH2CH(NH2)COOCH3·HCl) as a capping agent. These AuNPs, which achieve higher cell viability in comparison to cetyl trimethyl ammonium bromide (CTAB, a surfactant)-capped counterparts, have demonstrated potential for siRNA delivery. However, it is well known that systemic administration of cationic delivery systems without biological stablising moieties causes non-specific binding with negatively charged serum proteins, resulting in particle aggregation and opsonisation. Consequently, highly stable AuNPs capped with l-cysteine methyl ester hydrochloride conjugated to poly(ethylene glycol) (PEG) were synthesised in this study. PEGylation enhanced the biocompatibility of the AuNPs by reducing toxicity in a range of cell types, by inhibiting interaction with serum proteins thus avoiding aggregation, and, by providing protection against degradation by nucleases. Moreover, these PEGylated AuNPs formed nanoparticles (NPs) with siRNA (which was first compacted with protamine), and had a diameter within the nanoscale range (∼ 250 nm) and a near neutral surface charge (∼ 10 mV). In the future a bifunctional PEG chain on the AuNPs (i.e., SH-PEG-NH2, SH-PEG-COOH) will be used to facilitate conjugation of a targeting ligand to enhance cell specific uptake.
Collapse
Affiliation(s)
- Kamil Rahme
- Materials Chemistry and Analysis Group, Department of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland; AMBER (Advanced Materials and Biological Engineering Research Centre), CRANN (Centre for Research on Adaptive Nanostructures and Nanodevices), Trinity College Dublin, Dublin, Ireland; Department of Sciences, Faculty of Natural and Applied Science, Notre Dame University (Louaize), Lebanon
| | - Jianfeng Guo
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Ireland
| | - Justin D Holmes
- Materials Chemistry and Analysis Group, Department of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland; AMBER (Advanced Materials and Biological Engineering Research Centre), CRANN (Centre for Research on Adaptive Nanostructures and Nanodevices), Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
25
|
Nguyen LH, Diao HJ, Chew SY. MicroRNAs and their potential therapeutic applications in neural tissue engineering. Adv Drug Deliv Rev 2015; 88:53-66. [PMID: 25980934 DOI: 10.1016/j.addr.2015.05.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 05/08/2015] [Accepted: 05/10/2015] [Indexed: 01/01/2023]
Abstract
The inherent poor regeneration capacity of nerve tissues, especially in the central nervous system, poses a grand challenge for neural tissue engineering. After injuries, the local microenvironment often contains potent inhibitory molecules and glial scars, which do not actively support axonal regrowth. MicroRNAs can direct fate of neural cells and are tightly controlled during nerve development. Thus, RNA interference using microRNAs is a promising method to enhance nerve regeneration. Although the physiological roles of microRNA expression levels in various cellular activities or disease conditions have been extensively investigated, the translational use of these understanding for neural tissue engineering remains limited. This review aims to highlight essential microRNAs that participate in cellular behaviors within the adult nervous system and their potential therapeutic applications. In addition, possible delivery methods are also suggested for effective gene silencing in neural tissue engineering.
Collapse
Affiliation(s)
- Lan Huong Nguyen
- Division of Chemical and Biomolecular Engineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Hua Jia Diao
- Division of Chemical and Biomolecular Engineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Sing Yian Chew
- Division of Chemical and Biomolecular Engineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore.
| |
Collapse
|
26
|
Gomes MJ, Martins S, Sarmento B. siRNA as a tool to improve the treatment of brain diseases: Mechanism, targets and delivery. Ageing Res Rev 2015; 21:43-54. [PMID: 25796492 DOI: 10.1016/j.arr.2015.03.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/10/2015] [Accepted: 03/16/2015] [Indexed: 10/23/2022]
Abstract
As the population ages, brain pathologies such as neurodegenerative diseases and brain cancer increase their incidence, being the need to find successful treatments of upmost importance. Drug delivery to the central nervous system (CNS) is required in order to reach diseases causes and treat them. However, biological barriers, mainly blood-brain barrier (BBB), are the key obstacles that prevent the effectiveness of possible treatments due to their ability to strongly limit the perfusion of compounds into the brain. Over the past decades, new approaches towards overcoming BBB and its efflux transporters had been proposed. One of these approaches here reviewed is through small interfering RNA (siRNA), which is capable to specifically target one gene and silence it in a post-transcriptional way. There are different possible functional proteins at the BBB, as the ones responsible for transport or just for its tightness, which could be a siRNA target. As important as the effective silence is the way to delivery siRNA to its anatomical site of action. This is where nanotechnology-based systems may help, by protecting siRNA circulation and providing cell/tissue-targeting and intracellular siRNA delivery. After an initial overview on incidence of brain diseases and basic features of the CNS, BBB and its efflux pumps, this review focuses on recent strategies to reach brain based on siRNA, and how to specifically target these approaches in order to treat brain diseases.
Collapse
|
27
|
Mastorakos P, Zhang C, Berry S, Oh Y, Lee S, Eberhart CG, Woodworth GF, Suk JS, Hanes J. Highly PEGylated DNA Nanoparticles Provide Uniform and Widespread Gene Transfer in the Brain. Adv Healthc Mater 2015; 4:1023-33. [PMID: 25761435 DOI: 10.1002/adhm.201400800] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/15/2015] [Indexed: 12/13/2022]
Abstract
Gene delivery to the central nervous system (CNS) has potential as a means for treating numerous debilitating neurological diseases. Nonviral gene vector platforms are tailorable and can overcome key limitations intrinsic to virus-mediated delivery; however, lack of clinical efficacy with nonviral systems to date may be attributed to limited gene vector dispersion and transfection in vivo. It is shown that the brain extracellular matrix (ECM) strongly limits penetration of polymer-based gene vector nanoparticles (NP) through the brain parenchyma, even when they are very small (<60 nm) and coated with a polyethylene glycol (PEG) corona of typical density. Following convection enhanced delivery (CED), conventional gene vectors are confined to the injection site, presumably by adhesive interactions with the brain ECM and do not provide gene expression beyond the point of administration. In contrast, it is found that incorporating highly PEGylated polymers allows the production of compacted (≈43 nm) and colloidally stable DNA NP that avoid adhesive trapping within the brain parenchyma. When administered by CED into the rat striatum, highly PEGylated DNA NP distribute throughout and provide broad transgene expression without vector-induced toxicity. The use of these brain-penetrating gene vectors, in conjunction with CED, offers an avenue to improve gene therapy for CNS diseases.
Collapse
Affiliation(s)
- Panagiotis Mastorakos
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Department of Ophthalmology; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 600 N. Wolfe Street Baltimore MD 21297 USA
| | - Clark Zhang
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Department of Biomedical Engineering; Johns Hopkins University School of Medicine; 720 Rutland Avenue Baltimore MD 21205 USA
| | - Sneha Berry
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Center for Biotechnology Education; Krieger School of Arts and Sciences; Johns Hopkins University; 3400 N. Charles Street Baltimore MD 21218 USA
| | - Yumin Oh
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Russell H. Morgan Department of Radiology and Radiological Science; Johns Hopkins University; 601 N. Caroline Street Baltimore MD 21287 USA
| | - Seulki Lee
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Russell H. Morgan Department of Radiology and Radiological Science; Johns Hopkins University; 601 N. Caroline Street Baltimore MD 21287 USA
| | - Charles G. Eberhart
- Department of Pathology; Johns Hopkins University School of Medicine; 600 N. Wolfe Street Baltimore MD 21287 USA
| | - Graeme F. Woodworth
- Departments of Neurological Surgery Anatomy and Neurobiology; University of Maryland School of Medicine; 22 S. Greene Street Baltimore MD 21201 USA
| | - Jung Soo Suk
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Department of Ophthalmology; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 600 N. Wolfe Street Baltimore MD 21297 USA
| | - Justin Hanes
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Department of Ophthalmology; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 600 N. Wolfe Street Baltimore MD 21297 USA
- Department of Biomedical Engineering; Johns Hopkins University School of Medicine; 720 Rutland Avenue Baltimore MD 21205 USA
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; 3400 N. Charles Street Baltimore MD 21218 USA
| |
Collapse
|
28
|
Gooding M, Malhotra M, McCarthy DJ, Godinho BMDC, Cryan JF, Darcy R, O'Driscoll CM. Synthesis and characterization of rabies virus glycoprotein-tagged amphiphilic cyclodextrins for siRNA delivery in human glioblastoma cells: in vitro analysis. Eur J Pharm Sci 2015; 71:80-92. [PMID: 25703259 DOI: 10.1016/j.ejps.2015.02.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 12/23/2014] [Accepted: 02/11/2015] [Indexed: 11/28/2022]
Abstract
In man brain cancer is an aggressive, malignant form of tumour, it is highly infiltrative in nature, is associated with cellular heterogeneity and affects cerebral hemispheres of the brain. Current drug therapies are inadequate and an unmet clinical need exists to develop new improved therapeutics. The ability to silence genes associated with disease progression by using short interfering RNA (siRNA) presents the potential to develop safe and effective therapies. In this work, in order to protect the siRNA from degradation, promote cell specific uptake and enhance gene silencing efficiency, a PEGylated cyclodextrin (CD)-based nanoparticle, tagged with a CNS-targeting peptide derived from the rabies virus glycoprotein (RVG) was formulated and characterized. The modified cyclodextrin derivatives were synthesized and co-formulated to form nanoparticles containing siRNA which were analysed for size, surface charge, stability, cellular uptake and gene-knockdown in brain cancer cells. The results identified an optimised co-formulation prototype at a molar ratio of 1:1.5:0.5 (cationic cyclodextrin:PEGylated cyclodextrin:RVG-tagged PEGylated cyclodextrin) with a size of 281 ± 39.72 nm, a surface charge of 26.73 ± 3 mV, with efficient cellular uptake and a 27% gene-knockdown ability. This CD-based formulation represents a potential nanocomplex for systemic delivery of siRNA targeting brain cancer.
Collapse
Affiliation(s)
- Matt Gooding
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland; Centre for Synthesis and Chemical Biology, University College Dublin, Dublin, Ireland
| | - Meenakshi Malhotra
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - David J McCarthy
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Bruno M D C Godinho
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Lisbon School of Health Technology, Polytechnic Institute of Lisbon, Lisbon, Portugal
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Raphael Darcy
- Centre for Synthesis and Chemical Biology, University College Dublin, Dublin, Ireland
| | | |
Collapse
|
29
|
Godinho BM, Malhotra M, O’Driscoll CM, Cryan JF. Delivering a disease-modifying treatment for Huntington's disease. Drug Discov Today 2015; 20:50-64. [DOI: 10.1016/j.drudis.2014.09.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/28/2014] [Accepted: 09/16/2014] [Indexed: 11/16/2022]
|
30
|
Nanoparticles and the blood-brain barrier: advancing from in-vitro models towards therapeutic significance. Pharm Res 2014; 32:1161-85. [PMID: 25446769 DOI: 10.1007/s11095-014-1545-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/06/2014] [Indexed: 01/12/2023]
Abstract
The blood-brain barrier is a unique cell-based restrictive barrier that prevents the entry of many substances, including most therapeutics, into the central nervous system. A wide range of nanoparticulate delivery systems have been investigated with the aim of targeting therapeutics (drugs, nucleic acids, proteins) to the brain following administration by various routes. This review provides a comprehensive description of the design and formulation of these nanoparticles including the rationale behind individual approaches. In addition, the ability of currently available in-vitro BBB models to accurately predict the in-vivo performance of targeted nanoparticles is critically assessed.
Collapse
|
31
|
Development of poly (I:C) modified doxorubicin loaded magnetic dendrimer nanoparticles for targeted combination therapy. Biomed Pharmacother 2014; 68:979-87. [PMID: 25458787 DOI: 10.1016/j.biopha.2014.10.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 10/16/2014] [Indexed: 11/20/2022] Open
Abstract
The objective of this study was to develop and evaluate the anticancer activity and the safety of a combinational drug delivery system using polyamidoamine (PAMAM) dendrimer-coated iron oxide nanoparticles for doxorubicin and poly I:C delivery in vitro. Dendrimer-coated magnetic nanoparticles (DcMNPs) are suitable for drug delivery system as nanocarriers with their following properties, such as surface functional groups, symmetry perfection, internal cavities, nano-size and magnetization. These nanoparticles could be targeted to the tumor site under a magnetic field since they have a magnetic core. DcMNPs were found as a convenient vehicle for targeted doxorubicin delivery in cancer therapy. Poly (I:C) binding on doxorubicin loaded DcMNPs (DcMNPs-Dox) was reported for the first time in the literature. It was also demonstrated that loading of doxorubicin into the cavities of DcMNPs increases the binding efficiency of poly (I:C) to the surface functional groups of dendrimer up to 10 times. When we compare the in vitro cytotoxic properties of doxorubicin, poly (I:C) and poly (I:C) bound doxorubicin loaded DcMNPs (PIC-DcMNPs-Dox), it was observed that PIC-DcMNPs-Dox show the highest cytotoxic effect by passing the cell resistance mechanisms on doxorubicin resistant MCF7 (MCF7/Dox) cells. Results demonstrated that applying PIC-DcMNPs-Dox would improve the efficacy by increasing the biocompatibility of system in blood stream and the toxicity inside tumor cells. These results provide invaluable information and new insight for the design and optimization of a novel combinational drug delivery system for targeted cancer therapy.
Collapse
|
32
|
Godinho BMDC, Ogier JR, Quinlan A, Darcy R, Griffin BT, Cryan JF, O'Driscoll CM. PEGylated cyclodextrins as novel siRNA nanosystems: correlations between polyethylene glycol length and nanoparticle stability. Int J Pharm 2014; 473:105-12. [PMID: 24992319 DOI: 10.1016/j.ijpharm.2014.06.054] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 06/25/2014] [Accepted: 06/27/2014] [Indexed: 10/25/2022]
Abstract
Silencing disease-related genes in the central nervous system (CNS) using short interfering RNA (siRNA) holds great promise for treating neurological disorders. Yet, delivery of RNAi therapeutics to the brain poses major challenges to non-viral systems, especially when considering systemic administration. Cationic nanoparticles have been widely investigated for siRNA delivery, but the tendency of these to aggregate in physiological environments limits their intravenous application. Thus, strategies to increase the stability of nanoparticles have been developed. Here, we investigated the ability of modified cationic amphiphilic or PEGylated amphiphilic cyclodextrins (CD) to formulate stable CD.siRNA nanoparticles. To this end, we describe a simple method for post-modification of pre-formed cationic CD.siRNA nanoparticles at their surface using PEGylated CDs of different PEG lengths. PEGylated CD.siRNA nanoparticles presented reduced surface charges and increased stability in physiological salt conditions. Stability of PEGylated CD.siRNA nanoparticles in vitro increased with both PEG length and PEG density at the surface. Furthermore, in a comparative pharmacokinetic study, increased systemic exposure and reduced clearance were achieved with CD-formulations when compared to naked siRNAs. However, no significant differences were observed among non-PEGylated and PEGylated CD.siRNAs suggesting that longer PEG lengths might be required for improving stability in vivo.
Collapse
Affiliation(s)
- Bruno M D C Godinho
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland; Department Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Julien R Ogier
- Centre for synthesis and Chemical Biology, University College Dublin, Dublin, Ireland
| | - Aoife Quinlan
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Raphael Darcy
- Centre for synthesis and Chemical Biology, University College Dublin, Dublin, Ireland
| | - Brendan T Griffin
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - John F Cryan
- Department Anatomy and Neuroscience, University College Cork, Cork, Ireland; Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, Cork, Ireland
| | | |
Collapse
|
33
|
O'Mahony AM, Cronin MF, Mcmahon A, Evans JC, Daly K, Darcy R, O'Driscoll CM. Biophysical and Structural Characterisation of Nucleic Acid Complexes with Modified Cyclodextrins Using Circular Dichroism. J Pharm Sci 2014; 103:1346-55. [DOI: 10.1002/jps.23922] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 02/14/2014] [Accepted: 02/17/2014] [Indexed: 12/22/2022]
|
34
|
Barry G. Integrating the roles of long and small non-coding RNA in brain function and disease. Mol Psychiatry 2014; 19:410-6. [PMID: 24468823 DOI: 10.1038/mp.2013.196] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 12/12/2013] [Accepted: 12/16/2013] [Indexed: 12/20/2022]
Abstract
Regulatory RNA is emerging as the major architect of cognitive evolution and innovation in the mammalian brain. While the protein machinery has remained largely constant throughout animal evolution, the non protein-coding transcriptome has expanded considerably to provide essential and widespread cellular regulation, partly through directing generic protein function. Both long (long non-coding RNA) and small non-coding RNAs (for example, microRNA) have been demonstrated to be essential for brain development and higher cognitive abilities, and to be involved in psychiatric disease. Long non-coding RNAs, highly expressed in the brain and expanded in mammalian genomes, provide tissue- and activity-specific epigenetic and transcriptional regulation, partly through functional control of evolutionary conserved effector small RNA activity. However, increased cognitive sophistication has likely introduced concomitant psychiatric vulnerabilities, predisposing to conditions such as autism and schizophrenia, and cooperation between regulatory and effector RNAs may underlie neural complexity and concomitant fragility in the human brain.
Collapse
Affiliation(s)
- G Barry
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| |
Collapse
|
35
|
Nanomedicine-based neuroprotective strategies in patient specific-iPSC and personalized medicine. Int J Mol Sci 2014; 15:3904-25. [PMID: 24599081 PMCID: PMC3975375 DOI: 10.3390/ijms15033904] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Revised: 02/07/2014] [Accepted: 02/19/2014] [Indexed: 01/16/2023] Open
Abstract
In recent decades, nanotechnology has attracted major interests in view of drug delivery systems and therapies against diseases, such as cancer, neurodegenerative diseases, and many others. Nanotechnology provides the opportunity for nanoscale particles or molecules (so called “Nanomedicine”) to be delivered to the targeted sites, thereby, reducing toxicity (or side effects) and improving drug bioavailability. Nowadays, a great deal of nano-structured particles/vehicles has been discovered, including polymeric nanoparticles, lipid-based nanoparticles, and mesoporous silica nanoparticles. Nanomedical utilizations have already been well developed in many different aspects, including disease treatment, diagnostic, medical devices designing, and visualization (i.e., cell trafficking). However, while quite a few successful progressions on chemotherapy using nanotechnology have been developed, the implementations of nanoparticles on stem cell research are still sparsely populated. Stem cell applications and therapies are being considered to offer an outstanding potential in the treatment for numbers of maladies. Human induced pluripotent stem cells (iPSCs) are adult cells that have been genetically reprogrammed to an embryonic stem cell-like state. Although the exact mechanisms underlying are still unclear, iPSCs are already being considered as useful tools for drug development/screening and modeling of diseases. Recently, personalized medicines have drawn great attentions in biological and pharmaceutical studies. Generally speaking, personalized medicine is a therapeutic model that offers a customized healthcare/cure being tailored to a specific patient based on his own genetic information. Consequently, the combination of nanomedicine and iPSCs could actually be the potent arms for remedies in transplantation medicine and personalized medicine. This review will focus on current use of nanoparticles on therapeutical applications, nanomedicine-based neuroprotective manipulations in patient specific-iPSCs and personalized medicine.
Collapse
|
36
|
Godinho BM, McCarthy DJ, Torres-Fuentes C, Beltrán CJ, McCarthy J, Quinlan A, Ogier JR, Darcy R, O'Driscoll CM, Cryan JF. Differential nanotoxicological and neuroinflammatory liabilities of non-viral vectors for RNA interference in the central nervous system. Biomaterials 2014; 35:489-99. [DOI: 10.1016/j.biomaterials.2013.09.068] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 09/20/2013] [Indexed: 12/20/2022]
|