Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
J Invest Dermatol ; 141(6): 1428-1437.e8, 2021 06.
Article in English | MEDLINE | ID: mdl-33385397

ABSTRACT

Scavenger receptors clear pathogens, transport lipid, and mediate polyanionic ligand uptake in macrophages, but their expression and role in the skin are poorly understood. Although the epidermal barrier typically excludes nucleic acid entry, topically applied, spherically arranged oligonucleotide nanoconjugates (spherical nucleic acids [SNAs]) penetrate mouse skin, three-dimensional (3D) skin equivalents, and human skin. We explored the mechanism of SNA uptake in normal human epidermal keratinocytes and 3D skin equivalents. Normal human epidermal keratinocytes and 3D raft treatment with SR-A inhibitors reduced SNA uptake by >80%. The human epidermis expresses SR-As SCARA3 and, to a lesser extent, MARCO. Simultaneous lentiviral knockdown of SCARA3 and MARCO reduced SNA uptake in normal human epidermal keratinocytes and 3D rafts after topical application, affirming a role for SR-As in SNA uptake and 3D raft penetration. Incubation of normal human epidermal keratinocytes at 4oC or with sodium azide prevented SNA uptake, suggesting active endocytosis. Endocytosis inhibitors, immunofluorescence, immunoprecipitation, and knockdown studies localized functional SR-As to FLOT-1-containing lipid rafts throughout the epidermis and CAV-1-containing rafts only in the upper epidermis. These studies suggest a central role for SR-A complexes in epidermal lipid rafts in mediating the uptake of nucleic acid‒laden nanoparticles.


Subject(s)
Epidermis/metabolism , Heat-Shock Proteins/metabolism , Membrane Microdomains/metabolism , Nucleic Acids/pharmacokinetics , Receptors, Immunologic/metabolism , Scavenger Receptors, Class A/metabolism , Cells, Cultured , Endocytosis , Gene Knockdown Techniques , Heat-Shock Proteins/genetics , Humans , Keratinocytes/cytology , Membrane Proteins/metabolism , Nanoparticles , Nucleic Acids/administration & dosage , Primary Cell Culture/methods , Receptors, Immunologic/genetics , Scavenger Receptors, Class A/genetics
2.
Mol Ther ; 29(2): 521-539, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33188937

ABSTRACT

Oligonucleotide therapeutics are a novel promising class of drugs designed to specifically target either coding or non-coding RNA molecules to revolutionize treatment of various diseases. During preclinical development, investigations of the pharmacokinetic characteristics of these oligonucleotide-based drug candidates are essential. Oligonucleotides possess a long history of chemical modifications to enhance their stability and binding affinity, as well as reducing toxicity. Phosphorothioate backbone modifications of oligonucleotides were a hallmark of this development process that greatly enhanced plasma stability and protein binding of these agents. Modifications such as 2'-O-methylation further improved stability, while other modifications of the ribose, such as locked nucleic acid (LNA) modification, significantly increased binding affinity, potency, and tissue half-life. These attributes render oligonucleotide therapeutics able to regulate protein expression in both directions depending on the target RNA. Thus, a growing interest has emerged using these oligonucleotides in the treatment of neurodegenerative and cardiac disorders as well as cancer, since the deregulation of certain coding and non-coding RNAs plays a key role in the development of these diseases. Cutting edge research is being performed in the field of non-coding RNAs, identifying potential therapeutic targets, and developing novel oligonucleotide-based agents that outperform classical drugs. Some of these agents are either in clinical trials showing promising results or are already US Food and Drug Administration (FDA) approved, with more oligonucleotides being developed for therapeutic purposes. This is the advent of mechanism-based next-generation therapeutics for a wide range of diseases.


Subject(s)
Genetic Therapy , Nucleic Acids/pharmacokinetics , Nucleic Acids/therapeutic use , Animals , Clinical Studies as Topic , Clinical Trials as Topic , Drug Approval , Drug Development , Genetic Therapy/methods , Humans , Treatment Outcome , United States , United States Food and Drug Administration
3.
J Mater Chem B ; 8(41): 9385-9403, 2020 10 28.
Article in English | MEDLINE | ID: mdl-33048107

ABSTRACT

Dynamic covalent polymers are materials formed by reversible covalent bonds and non-covalent interactions through an adaptive constitutional dynamic chemistry. The implementation of dynamic covalent polymers in gene delivery has recently emerged due to their responsive and adaptive features. Indeed, such an approach offers the alluring promise of discovering optimal delivery vectors self-fitted to their nucleic acid cargos and responsive to environmental changes (e.g. pH changes or the presence of a biomolecular target). This review will discuss more precisely the structural features of the molecular building blocks used so far, the architecture of the resulting dynamic covalent polymers from linear to 2D and 3D, and the covalent and supramolecular self-assembly processes at play in nucleic acid recognition and delivery, showcasing in particular the very few examples of adaptive self-assembly of dynamic covalent polymers templated by nucleic acids and responsive to the presence of biomolecular targets found in cell membranes that facilitate cell entry.


Subject(s)
Cations/chemistry , Nucleic Acids/administration & dosage , Polymers/chemistry , Transfection/methods , Animals , Cations/metabolism , Cell Membrane/metabolism , Gene Transfer Techniques , Humans , Nucleic Acids/genetics , Nucleic Acids/pharmacokinetics , Polymers/metabolism
4.
Article in English | MEDLINE | ID: mdl-32202079

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disease that affects 0.5-1% of the world population. Current treatments include on one hand non-steroidal anti-inflammatory drugs and glucocorticoids (GCs) for treating pain and on the other hand disease-modifying anti-rheumatic drugs such as methotrexate, Janus kinase inhibitors or biologics such as antibodies targeting mainly cytokine expression. More recently, nucleic acids such as siRNA, miRNA, or anti-miRNA have shown strong potentialities for the treatment of RA. This review discusses the way nanomedicines can target GCs and nucleic acids to inflammatory sites, increase drug penetration within inflammatory cells, achieve better subcellular distribution and finally protect drugs against degradation. For GCs such a targeting effect would allow the treatment to be more effective at lower doses and to reduce the administration frequency as well as to induce much fewer side-effects. In the case of nucleic acids, particularly siRNA, knocking down proteins involved in RA, could importantly be facilitated using nanomedicines. Finally, the combination of both siRNA and GCs in the same carrier allowed for the same cell to target both the GCs receptor as well as any other signaling pathway involved in RA. Nanomedicines appear to be very promising for the delivery of conventional and novel drugs in RA therapeutics. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid/drug therapy , Glucocorticoids , Nanomedicine , Nucleic Acids , Animals , Antirheumatic Agents/pharmacokinetics , Antirheumatic Agents/therapeutic use , Glucocorticoids/pharmacokinetics , Glucocorticoids/therapeutic use , Humans , Mice , Nucleic Acids/pharmacokinetics , Nucleic Acids/therapeutic use , RNA, Small Interfering , Rats
5.
Adv Mater ; 32(13): e1901081, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31222852

ABSTRACT

Nucleic acids are a promising type of therapeutic for the treatment of a wide range of conditions, including cancer, but they also pose many delivery challenges. For efficient and safe delivery to cancer cells, nucleic acids must generally be packaged into a vehicle, such as a nanoparticle, that will allow them to be taken up by the target cells and then released in the appropriate cellular compartment to function. As with other types of therapeutics, delivery vehicles for nucleic acids must also be designed to avoid unwanted side effects; thus, the ability of such carriers to target their cargo to cancer cells is crucial. Classes of nucleic acids, hurdles that must be overcome for effective intracellular delivery, types of nonviral nanomaterials used as delivery vehicles, and the different strategies that can be employed to target nucleic acid delivery specifically to tumor cells are discussed. Additonally, nanoparticle designs that facilitate multiplexed delivery of combinations of nucleic acids are reviewed.


Subject(s)
Gene Transfer Techniques , Nanoparticles , Neoplasms/therapy , Nucleic Acids/administration & dosage , Animals , Drug Delivery Systems/methods , Genetic Therapy/methods , Humans , Nanoparticles/chemistry , Nanoparticles/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Nucleic Acids/genetics , Nucleic Acids/pharmacokinetics , Nucleic Acids/therapeutic use
6.
Nanomedicine ; 23: 102094, 2020 01.
Article in English | MEDLINE | ID: mdl-31669854

ABSTRACT

Programmable nucleic acid nanoparticles (NANPs) provide controlled coordination of therapeutic nucleic acids (TNAs) and other biological functionalities. Beyond multivalence, recent reports demonstrate that NANP technology can also elicit a specific immune response, adding another layer of customizability to this innovative approach. While the delivery of nucleic acids remains a challenge, new carriers are introduced and tested continuously. Polymeric platforms have proven to be efficient in shielding nucleic acid cargos from nuclease degradation while promoting their delivery and intracellular release. Here, we venture beyond the delivery of conventional TNAs and combine the stable cationic poly-(lactide-co-glycolide)-graft-polyethylenimine with functionalized NANPs. Furthermore, we compare several representative NANPs to assess how their overall structures influence their delivery with the same carrier. An extensive study of various formulations both in vitro and in vivo reveals differences in their immunostimulatory activity, gene silencing efficiency, and biodistribution, with fibrous NANPs advancing for TNA delivery.


Subject(s)
Adjuvants, Immunologic , Gene Silencing , Nanoparticles/chemistry , Nucleic Acids , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacokinetics , Adjuvants, Immunologic/pharmacology , Cell Line, Tumor , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Humans , Nucleic Acids/chemistry , Nucleic Acids/pharmacokinetics , Nucleic Acids/pharmacology
7.
Theranostics ; 9(9): 2662-2677, 2019.
Article in English | MEDLINE | ID: mdl-31131060

ABSTRACT

Rationale: Within the field of personalized medicine there is an increasing focus on designing flexible, multifunctional drug delivery systems that combine high efficacy with minimal side effects, by tailoring treatment to the individual. Methods: We synthesized a chemically stabilized ~4 nm nucleic acid nanoscaffold, and characterized its assembly, stability and functional properties in vitro and in vivo. We tested its flexibility towards multifunctionalization by conjugating various biomolecules to the four modules of the system. The pharmacokinetics, targeting capability and bioimaging properties of the structure were investigated in mice. The role of avidity in targeted liver cell internalization was investigated by flow cytometry, confocal microscopy and in vivo by fluorescent scanning of the blood and organs of the animals. Results: We have developed a nanoscaffold that rapidly and with high efficiency can self-assemble four chemically conjugated functionalities into a stable, in vivo-applicable system with complete control of stoichiometry and site specificity. The circulation time of the nanoscaffold could be tuned by functionalization with various numbers of polyethylene glycol polymers or with albumin-binding fatty acids. Highly effective hepatocyte-specific internalization was achieved with increasing valencies of tri-antennary galactosamine (triGalNAc) in vitro and in vivo. Conclusion: With its facile functionalization, stoichiometric control, small size and high serum- and thermostability, the nanoscaffold presented here constitutes a novel and flexible platform technology for theranostics.


Subject(s)
Diagnostic Imaging/methods , Drug Carriers/administration & dosage , Drug Carriers/pharmacokinetics , Drug Delivery Systems/methods , Nucleic Acids/administration & dosage , Nucleic Acids/pharmacokinetics , Theranostic Nanomedicine/methods , Animals , Drug Carriers/chemical synthesis , Drug Stability , Mice , Nucleic Acids/chemical synthesis
8.
Nat Commun ; 10(1): 1147, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30850596

ABSTRACT

DNA nanostructures are promising drug carriers with their intrinsic biocompatibility, uniformity and versatility. However, rapid serum disintegration leads to low bioavailability at targeted sites following systemic administration, hindering their biomedical applications. Here we demonstrate transdermal delivery of framework nucleic acids (FNAs) through topical applications. By designing FNAs with distinct shapes and sizes, we interrogate their penetration on mice and human skin explant. Skin histology reveals size-dependent penetration, with FNAs ≤75 nm effectively reaching dermis layer. 17 nm-tetrahedral FNAs show greatest penetration to 350 µm from skin periphery. Importantly, structural integrity is maintained during the skin penetration. Employing a mouse melanoma model, topical application of doxorubicin-loaded FNAs accommodates ≥2-fold improvement in drug accumulation and tumor inhibition relative to topically-applied free doxorubicin, or doxorubicin loaded in liposomes and polymeric nanoparticles. Programmable penetration with minimal systemic biodistribution underlines FNA potential as localized transdermal drug delivery carriers.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Delayed-Action Preparations/pharmacokinetics , Doxorubicin/pharmacology , Drug Delivery Systems/methods , Melanoma, Experimental/drug therapy , Nucleic Acids/chemistry , Skin Neoplasms/drug therapy , Administration, Cutaneous , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Delayed-Action Preparations/chemistry , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Humans , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Nude , Nucleic Acids/pharmacokinetics , Permeability , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Swine
9.
Bioconjug Chem ; 30(2): 293-304, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30462487

ABSTRACT

Cell delivery reagents often exploit the endocytic pathway as a route of cell entry. Once endocytosed, these reagents must overcome endosomal entrapment to ensure the release of their macromolecular cargo into the cytosol of cells. In this review, we describe several examples of prototypical synthetic reagents that are capable of endosomal escape and examine their mechanisms of action, their efficiencies, and their effects on cells. Although these delivery systems are chemically distinct, some commonalities in how they interact with cellular membranes can be inferred. This, in turn, sheds some light on the process of endosomal escape, and may help guide the development and optimization of next-generation delivery tools.


Subject(s)
Cytosol/metabolism , Drug Carriers/metabolism , Endosomes/metabolism , Nucleic Acids/administration & dosage , Proteins/administration & dosage , Animals , Drug Carriers/chemistry , Drug Delivery Systems/methods , Endocytosis , Humans , Lipids/chemistry , Nucleic Acids/pharmacokinetics , Peptides/chemistry , Peptides/metabolism , Polymers/chemistry , Polymers/metabolism , Proteins/pharmacokinetics
10.
Nano Lett ; 18(12): 7590-7600, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30216729

ABSTRACT

Nanoparticles are often targeted to receptors expressed on specific cells, but few receptors are (i) highly expressed on one cell type and (ii) involved in endocytosis. One unexplored alternative is manipulating an endocytic gene expressed on multiple cell types; an ideal gene would inhibit delivery to cell type A more than cell type B, promoting delivery to cell type B. This would require a commonly expressed endocytic gene to alter nanoparticle delivery in a cell type-dependent manner in vivo; whether this can occur is unknown. Based on its microenvironmental regulation, we hypothesized Caveolin 1 (Cav1) would exert cell type-specific effects on nanoparticle delivery. Fluorescence was not sensitive enough to investigate this question, and as a result, we designed a platform named QUANT to study nanoparticle biodistribution. QUANT is 108× more sensitive than fluorescence and can be multiplexed. By measuring how 226 lipid nanoparticles (LNPs) delivered nucleic acids to multiple cell types in vivo in wild-type and Cav1 knockout mice, we found Cav1 altered delivery in a cell-type specific manner. Cav1 knockout did not alter LNP delivery to lung and kidney macrophages but substantially reduced LNP delivery to Kupffer cells, which are liver-resident macrophages. These data suggest caveolin-mediated endocytosis of nanomedicines by macrophages varies with tissue type. These results suggest manipulating receptors expressed on multiple cell types can tune drug delivery.


Subject(s)
Caveolin 1/metabolism , Drug Carriers/metabolism , Nanoparticles/metabolism , Nucleic Acids/administration & dosage , Animals , Caveolin 1/genetics , Cell Line , Cells, Cultured , Drug Carriers/chemistry , Drug Delivery Systems , Endocytosis , Kupffer Cells/metabolism , Lipid Metabolism , Lipids/chemistry , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nanoparticles/chemistry , Nucleic Acids/pharmacokinetics , Tissue Distribution
11.
Yakugaku Zasshi ; 138(5): 591-598, 2018.
Article in Japanese | MEDLINE | ID: mdl-29709998

ABSTRACT

 Nucleic acid therapy is expected to be a next generation medicine. We recently developed a multifunctional envelope-type nano device (MEND) for use as a novel delivery system. The modification of polyethylene glycol (PEG), i.e., PEGylation, is useful for achieving the delivery of MENDs to tumors via an enhanced permeability and retention (EPR) effect. However, PEGylation strongly inhibits the cellular uptake and endosomal escape of MEND, which results in significant loss of action, and therefore lost effectiveness, of the cargo therapeutic. For successful nucleic acid delivery in cancer treatment, the crucial problem associated with the use of PEG, known as the "PEG dilemma", must be solved. In this review, we describe the development and application of MEND in overcoming the PEG dilemma based on manipulating both the pharmacokinetics and intracellular trafficking of cellular uptake and endosomal release using a cleavable PEG lipid, a pH-sensitive fusogenic peptide, and a pH-sensitive cationic lipid. We also developed dual-ligand liposomes with a controlled diameter of around 300 nm, then modified these with a specific ligand and a cell penetrating peptide designed to target the neovasculature of tumors. Dual-ligand liposomes could induce an anti-tumor effect in drug resistant tumors by delivering drugs to tumor blood vessels, rather than to the cancer cells themselves. Here, we review our recent efforts to develop a novel liposomal drug delivery system (DDS) by manipulating pharmacokinetics and intracellular trafficking for drug therapy and nucleic acid medicine.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Drug Delivery Systems , Drug Design , Liposomes , Nucleic Acids/administration & dosage , Nucleic Acids/pharmacokinetics , Drug Delivery Systems/methods , Humans , Hydrogen-Ion Concentration , Neoplasms/metabolism , Polyethylene Glycols
12.
Nano Lett ; 18(3): 2148-2157, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29489381

ABSTRACT

Endothelial cells and macrophages play active roles in disease and as a result are important targets for nucleic acid therapies. While thousands of chemically distinct lipid nanoparticles (LNPs) can be synthesized to deliver nucleic acids, studying more than a few LNPs in vivo is challenging. As a result, it is difficult to understand how nanoparticles target these cells in vivo. Using high throughput LNP barcoding, we quantified how well LNPs delivered DNA barcodes to endothelial cells and macrophages in vitro, as well as endothelial cells and macrophages isolated from the lung, heart, and bone marrow in vivo. We focused on two fundamental questions in drug delivery. First, does in vitro LNP delivery predict in vivo LNP delivery? By comparing how 281 LNPs delivered barcodes to endothelial cells and macrophages in vitro and in vivo, we found in vitro delivery did not predict in vivo delivery. Second, does LNP delivery change within the microenvironment of a tissue? We quantified how 85 LNPs delivered barcodes to eight splenic cell populations, and found that cell types derived from myeloid progenitors tended to be targeted by similar LNPs, relative to cell types derived from lymphoid progenitors. These data demonstrate that barcoded LNPs can elucidate fundamental questions about in vivo nanoparticle delivery.


Subject(s)
Drug Delivery Systems , Lipids/chemistry , Nanoparticles/chemistry , Nucleic Acids/administration & dosage , Animals , Cell Line , Cells, Cultured , Endothelial Cells/metabolism , Female , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Nanotechnology , Nucleic Acids/pharmacokinetics
13.
Article in English | MEDLINE | ID: mdl-28544521

ABSTRACT

Nucleic acid-based therapeutics has the potential for treating numerous diseases by correcting abnormal expression of specific genes. Lack of safe and efficacious delivery strategies poses a major obstacle limiting clinical advancement of nucleic acid therapeutics. Oral route of drug administration has greater delivery challenges, because the administered genes or oligonucleotides have to bypass degrading environment of the gastrointestinal (GI) tract in addition to overcoming other cellular barriers preventing nucleic acid delivery. For efficient oral nucleic acid delivery, vector should be such that it can protect encapsulated material during transit through the GI tract, facilitate efficient uptake and intracellular trafficking at desired target sites, along with being safe and well tolerated. In this review, we have discussed multicompartmental systems for overcoming extracellular and intracellular barriers to oral delivery of nucleic acids. A nanoparticles-in-microsphere oral system-based multicompartmental system was developed and tested for in vivo gene and small interfering RNA delivery for treating colitis in mice. This system has shown efficient transgene expression or gene silencing when delivered orally along with favorable downstream anti-inflammatory effects, when tested in a mouse model of intestinal bowel disease. WIREs Nanomed Nanobiotechnol 2018, 10:e1478. doi: 10.1002/wnan.1478 This article is categorized under: Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.


Subject(s)
Administration, Oral , Drug Delivery Systems , Gastrointestinal Tract , Genetic Therapy , Nucleic Acids , Animals , Gastrointestinal Tract/chemistry , Gastrointestinal Tract/metabolism , Humans , Mice , Nucleic Acids/administration & dosage , Nucleic Acids/pharmacokinetics , Nucleic Acids/therapeutic use , Transfection
14.
Chem Pharm Bull (Tokyo) ; 65(7): 642-648, 2017.
Article in English | MEDLINE | ID: mdl-28674337

ABSTRACT

In this review, we have summarized evaluation methods for the analysis of external stimuli-mediated nucleic acid and gene delivery. Prior to reviewing these evaluation methods, we describe various delivery processes of nucleic acid and gene medicines (small interfering RNA (siRNA), micro RNA, mRNA, plasmid DNA, etc.), which include interaction with blood components, bio-distribution, disposition in the target tissue, cell entry, intracellular trafficking, nuclear localization, and dissociation from the carriers. Next, we discuss the advantages of external stimuli-mediated nucleic acid and gene delivery. External stimuli enable us to effectively deliver nucleic acids and genes to targeted regions. Evaluation methods are required to elucidate the behaviors of nucleic acid and gene medicines in the body. Quantitative analyses of the bio-distribution and in situ disposition in perfused organs, as well as visualization of bio-distribution, transgene expression in the body, and intracellular trafficking of nucleic acid and gene medicines, are all useful in evaluating not only the efficacy and safety of delivery, but also serve as guidelines for the further development of nucleic acid and gene medicines by elucidating delivery problems. Progress in evaluation methods, including tissue optical clearing and super resolution microscopy, will help to better elucidate the in vivo fate of nucleic acid and gene medicines.


Subject(s)
Genetic Therapy , Nucleic Acids/administration & dosage , Transfection , Animals , Humans , Nucleic Acids/pharmacokinetics , Tissue Distribution
15.
Curr Gene Ther ; 17(2): 154-169, 2017.
Article in English | MEDLINE | ID: mdl-28494740

ABSTRACT

The restless endeavors revealing the molecular pathways underlying many neurodegenerative diseases and brain tumors have paved the way for the introduction of the selective exogenous gene-based therapeutics. The implicated active biomolecules encompass mainly negatively-charged nucleic acids ranging from DNA, mRNA, non-coding RNAs (small-interfering RNA, siRNA, and microRNA, miRNA), to antisense oligonucleotides. They selectively interfere with the genes translational and/or transcriptional processes. Although many reviews previously addressed brain targeting, a thorough correlation between the molecular properties of these biomacromolecules, the nature of blood brain barrier (BBB) in the accompanying pathological condition, the intracellular targets, as well as the design of the delivery system which will transport the bioactive cargo to the target cells attempting efficient delivery to the active sites in the brain will be appraised. In this review, we will further discuss the tremendous advances in non-viral gene delivery nanosystems currently investigated (starting from self-assembled nanoplexes using cationic polymers or lipids and going through liposomes, aptamers, polymersomes, exosomes, dendrimers and nanoparticles). Unlike previous reviews on this topic, functionalization strategies of the nanocarriers promoting either surface receptor binding or intracellular targeting of the cranial cells will be highlighted, with special emphasis on tailoring smart nanomedicines according to the CNS disease condition. In addition, newly-developed evaluation approaches, cell culture models studying BBB permeability and manipulation of the barrier function of the brain via focused ultrasound will be addressed.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Delivery Systems/methods , Gene Transfer Techniques , Nanomedicine/methods , Nanoparticles/administration & dosage , Nucleic Acids/administration & dosage , Animals , Brain/blood supply , Brain/metabolism , Genetic Therapy/methods , Humans , Nanoparticles/chemistry , Nucleic Acids/genetics , Nucleic Acids/pharmacokinetics
16.
Biomater Sci ; 5(3): 412-416, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-28133665

ABSTRACT

Spherical nucleic acids (SNAs) are spherically arranged oligonucleotides on core inorganic nanoparticles and have great potential for intracellular delivery of bioactive molecules, since they have been found to be internalized into mammalian cells. Understanding the factors that influence the cellular uptake of SNAs would be beneficial to design SNAs with novel uptake properties. We here report the effect of the sugar backbone type of the oligonucleotides on the cellular internalization of SNAs. After the preparation of SNAs with the oligonucleotides of five different sugar backbones, we analyze the cellular uptake efficiency quantitatively by flow cytometry and inductively coupled plasma mass spectrometry (ICP-MS). The data reveal that the uptake efficiencies and the uptake mechanisms significantly rely on the backbone type. These results suggest that the backbone modification can provide a unique handle to tune the cellular uptake behavior of SNAs.


Subject(s)
Gold/chemistry , Nanoparticles/chemistry , Nucleic Acids/chemistry , Nucleic Acids/pharmacokinetics , Oligonucleotides/chemistry , Oligonucleotides/pharmacokinetics , Biological Transport , HeLa Cells , Humans , Nucleic Acid Conformation , Nucleic Acids/administration & dosage , Oligonucleotides/administration & dosage
17.
Philos Trans A Math Phys Eng Sci ; 374(2072)2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27298431

ABSTRACT

Cationic liposomes (CLs) are synthetic carriers of nucleic acids in gene delivery and gene silencing therapeutics. The introduction will describe the structures of distinct liquid crystalline phases of CL-nucleic acid complexes, which were revealed in earlier synchrotron small-angle X-ray scattering experiments. When mixed with plasmid DNA, CLs containing lipids with distinct shapes spontaneously undergo topological transitions into self-assembled lamellar, inverse hexagonal, and hexagonal CL-DNA phases. CLs containing cubic phase lipids are observed to readily mix with short interfering RNA (siRNA) molecules creating double gyroid CL-siRNA phases for gene silencing. Custom synthesis of multivalent lipids and a range of novel polyethylene glycol (PEG)-lipids with attached targeting ligands and hydrolysable moieties have led to functionalized equilibrium nanoparticles (NPs) optimized for cell targeting, uptake or endosomal escape. Very recent experiments are described with surface-functionalized PEGylated CL-DNA NPs, including fluorescence microscopy colocalization with members of the Rab family of GTPases, which directly reveal interactions with cell membranes and NP pathways. In vitro optimization of CL-DNA and CL-siRNA NPs with relevant primary cancer cells is expected to impact nucleic acid therapeutics in vivo. This article is part of the themed issue 'Soft interfacial materials: from fundamentals to formulation'.


Subject(s)
Cations , Gene Silencing , Liposomes , Nanoparticles/chemistry , Nucleic Acids , Transfection/methods , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacokinetics , Cations/chemistry , Cations/pharmacokinetics , Cell Line , Humans , Liposomes/chemistry , Liposomes/pharmacokinetics , Mice , Nanotechnology , Nucleic Acids/chemistry , Nucleic Acids/pharmacokinetics
18.
Nanoscale ; 8(16): 8955-65, 2016 Apr 28.
Article in English | MEDLINE | ID: mdl-27073992

ABSTRACT

Coiled coil interactions are strong protein-protein interactions that are involved in many biological processes, including intracellular trafficking and membrane fusion. A synthetic heterodimeric coiled-coil forming peptide pair, known as E3 (EIAALEK)3 and K3 (KIAALKE)3 was used to functionalize liposomes encapsulating a splice correcting oligonucleotide or siRNA. These peptide-functionalized vesicles are highly stable in solution but start to cluster when vesicles modified with complementary peptides are mixed together, demonstrating that the peptides quickly coil and crosslink the vesicles. When one of the peptides was anchored to the cell membrane using a hydrophobic cholesterol anchor, vesicles functionalized with the complementary peptide could be docked to these cells, whereas non-functionalized cells did not show any vesicle tethering. Although the anchored peptides do not have a downstream signaling pathway, microscopy pictures revealed that after four hours, the majority of the docked vesicles were internalized by endocytosis. Finally, for the first time, it was shown that the coiled coil assembly at the interface between the vesicles and the cell membrane induces active uptake and leads to cytosolic delivery of the nucleic acid cargo. Both the siRNA and the splice correcting oligonucleotide were functionally delivered, resulting respectively in the silencing or recovery of luciferase expression in the appropriate cell lines. These results demonstrate that the docking to the cell by coiled coil interaction can induce active uptake and achieve the successful intracellular delivery of otherwise membrane impermeable nucleic acids in a highly specific manner.


Subject(s)
Liposomes/administration & dosage , Liposomes/chemistry , Nucleic Acids/administration & dosage , Amino Acid Sequence , Cell Membrane Permeability , Drug Delivery Systems , Endocytosis , HeLa Cells , Humans , Nanotechnology , Nucleic Acids/pharmacokinetics , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Protein Interaction Domains and Motifs
19.
ACS Chem Biol ; 11(5): 1180-91, 2016 05 20.
Article in English | MEDLINE | ID: mdl-26950048

ABSTRACT

Oligonucleotides present a high therapeutic potential for a wide variety of diseases. However, their clinical development is limited by their degradation by nucleases and their poor blood circulation time. Depending on the administration mode and the cellular target, these macromolecules will have to cross the vascular endothelium, to diffuse through the extracellular matrix, to be transported through the cell membrane, and finally to reach the cytoplasm. To overcome these physiological barriers, many strategies have been developed. Here, we review different methods of DNA vectorization, discuss limitations and advantages of the various vectors, and provide new perspectives for future development.


Subject(s)
Drug Delivery Systems , Gene Transfer Techniques , Nucleic Acids/administration & dosage , Oligonucleotides/administration & dosage , Animals , Drug Delivery Systems/instrumentation , Drug Delivery Systems/methods , Gene Transfer Techniques/instrumentation , Humans , Nucleic Acids/chemistry , Nucleic Acids/pharmacokinetics , Nucleic Acids/therapeutic use , Oligonucleotides/chemistry , Oligonucleotides/pharmacokinetics , Oligonucleotides/therapeutic use
20.
Adv Colloid Interface Sci ; 233: 161-175, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26265376

ABSTRACT

Due to the potential use as transfecting agents of nucleic acids (DNA or RNA), multivalent cationic non-viral vectors have received special attention in the last decade. Much effort has been addressed to synthesize more efficient and biocompatible gene vectors able to transport nucleic acids into the cells without provoking an immune response. Among them, the mostly explored to compact and transfect nucleic acids are: (a) gemini and multivalent cationic lipids, mixed with a helper lipid, by forming lipoplexes; and (b) cationic polymers, polycations, and polyrotaxanes, by forming polyplexes. This review is focused on the progress and recent advances experimented in this area, mainly during the present decade, devoting special attention to the lipoplexes and polyplexes, as follows: (a) to its biophysical characterization (mainly electrostatics, structure, size and morphology) using a wide variety of experimental methods; and (b) to its biological activity (transfection efficacy and cytotoxicity) addressed to confirm the optimum formulations and viability of these complexes as very promising gene vectors of nucleic acids in nanomedicine.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/chemistry , Nucleic Acids/therapeutic use , Animals , Genetic Diseases, Inborn/pathology , Genetic Diseases, Inborn/therapy , Genetic Vectors/pharmacokinetics , Humans , Lipids/chemistry , Lipids/pharmacokinetics , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Nucleic Acids/pharmacokinetics , Polyamines/chemistry , Polyamines/pharmacokinetics , Polyelectrolytes , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/pharmacokinetics , Rotaxanes/chemistry , Rotaxanes/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...