Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Biomedicines ; 9(8)2021 Aug 19.
Article in English | MEDLINE | ID: mdl-34440250

ABSTRACT

Splice-switching therapy with splice-switching oligonucleotides (SSOs) has recently proven to be a clinically applicable strategy for the treatment of several mis-splice disorders. Despite this, wider application of SSOs is severely limited by the inherently poor bioavailability of SSO-based therapeutic compounds. Cell-penetrating peptides (CPPs) are a class of drug delivery systems (DDSs) that have recently gained considerable attention for improving the uptake of various oligonucleotide (ON)-based compounds, including SSOs. One strategy that has been successfully applied to develop effective CPP vectors is the introduction of various lipid modifications into the peptide. Here, we repurpose hydrocarbon-modified amino acids used in peptide stapling for the orthogonal introduction of hydrophobic modifications into the CPP structure during peptide synthesis. Our data show that α,α-disubstituted alkenyl-alanines can be successfully utilized to introduce hydrophobic modifications into CPPs to improve their ability to formulate SSOs into nanoparticles (NPs), and to mediate high delivery efficacy and tolerability both in vitro and in vivo. Conclusively, our results offer a new flexible approach for the sequence-specific introduction of hydrophobicity into the structure of CPPs and for improving their delivery properties.

2.
Pharmaceutics ; 13(6)2021 Jun 14.
Article in English | MEDLINE | ID: mdl-34198625

ABSTRACT

The toolbox for genetic engineering has quickly evolved from CRISPR/Cas9 to a myriad of different gene editors, each with promising properties and enormous clinical potential. However, a major challenge remains: delivering the CRISPR machinery to the nucleus of recipient cells in a nontoxic and efficient manner. In this article, we repurpose an RNA-delivering cell-penetrating peptide, PepFect14 (PF14), to deliver Cas9 ribonucleoprotein (RNP). The RNP-CPP complex achieved high editing rates, e.g., up to 80% in HEK293T cells, while being active at low nanomolar ranges without any apparent signs of toxicity. The editing efficiency was similar to or better compared to the commercially available reagents RNAiMAX and CRISPRMax. The efficiency was thoroughly evaluated in reporter cells and wild-type cells by restriction enzyme digest and next-generation sequencing. Furthermore, the CPP-Cas9-RNP complexes were demonstrated to withstand storage at different conditions, including freeze-thaw cycles and freeze-drying, without a loss in editing efficiency. This CPP-based delivery strategy complements existing technologies and further opens up new opportunities for Cas9 RNP delivery, which can likely be extended to other gene editors in the future.

3.
FASEB J ; 33(10): 11314-11325, 2019 10.
Article in English | MEDLINE | ID: mdl-31311315

ABSTRACT

Antisense oligonucleotides (ASOs) are a promising class of therapeutics that are starting to emerge in the clinic. Determination of intracellular concentrations required for biologic effects and identification of effective delivery vehicles are crucial for understanding the mode of action and required dosing. Here, we investigated which nuclear oligonucleotide concentration is needed for a therapeutic effect for a triplet repeat-targeting ASO in a muscle cell model of myotonic dystrophy type 1 (DM1). For cellular delivery, ASOs were complexed into nanoparticles using the cationic cell-penetrating peptides nona-arginine and PepFect14 (PF14). Although both peptides facilitated uptake, only PF14 led to a dose-dependent correction of disease-typical abnormal splicing. In line with this observation, time-lapse confocal microscopy demonstrated that only PF14 mediated translocation of the ASOs to the nucleus, which is the main site of action. Through fluorescence lifetime imaging, we could distinguish intact oligonucleotide from free fluorophore, showing that PF14 also shielded the ASOs from degradation. Finally, we employed a combination of live-cell fluorescence correlation spectroscopy and immunofluorescence microscopy and demonstrated that intranuclear blocking-type oligonucleotide concentrations in the upper nanomolar range were required to dissolve nuclear muscleblind-like protein 1 foci, a hallmark of DM1. Our findings have important implications for the clinical use of ASOs in DM1 and provide a basis for further research on other types of ASOs.-Van der Bent, M. L., Paulino da Silva Filho, O., Willemse, M., Hällbrink, M., Wansink, D. G., Brock, R. The nuclear concentration required for antisense oligonucleotide activity in myotonic dystrophy cells.


Subject(s)
Cell Nucleus/genetics , Myotonic Dystrophy/genetics , Oligonucleotides, Antisense/genetics , Cells, Cultured , Humans , Muscle, Skeletal/physiology , Myoblasts/physiology , Oligonucleotides/genetics
4.
Eur J Pharm Biopharm ; 141: 180-190, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31103743

ABSTRACT

Ovarian cancer is the most lethal gynecological malignancy in the developed world. In spite of intensive research, the mortality has hardly decreased over the past twenty years. This necessitates the exploration of novel therapeutic modalities. Transient protein expression through delivery of mRNA is emerging as a highly promising option. In contrast to gene therapy there is no risk of integration into the genome. Here, we explore the expression of mRNA in models of ovarian cancer of increasing complexity. The cell-penetrating peptide (CPP) PepFect 14 (PF14) was used to formulate CPP-mRNA nanoparticles. Efficient expression of a reporter protein was achieved in two-dimensional tissue cultures and in three-dimensional cancer cell spheroids. PF14 nanoparticles greatly outperformed a lipid-based transfection agent in vivo, leading to expression in various cell types of tumor associated tissue. Protein expression was restricted to the peritoneal cavity. Messenger RNA expression across different cell types was confirmed in primary ovarian cancer explants. As ovarian cancer is confined to the peritoneal cavity in most cases, the results create the basis for applications in which the tumor microenvironment is transiently modified through protein expression.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Lipopeptides/administration & dosage , Ovarian Neoplasms/therapy , RNA, Messenger/administration & dosage , Cell Line, Tumor , Female , Humans , Nanoparticles/administration & dosage , Transfection/methods , Tumor Microenvironment/genetics
5.
J Biomater Appl ; 33(3): 392-401, 2018 09.
Article in English | MEDLINE | ID: mdl-30223733

ABSTRACT

Gene-based therapies, including the delivery of oligonucleotides, offer promising methods for the treatment of cancer cells. However, they have various limitations including low efficiency. Herein, cell-penetrating peptides (CPPs)-conjugated chitosan-modified iron oxide magnetic nanoparticles (CPPs-CTS@MNPs) with high biocompatibility as well as high efficiency were tested for the delivery of oligonucleotides such as plasmid pGL3, splice correction oligonucleotides, and small-interfering RNA. A biocompatible nanocomposite, in which CTS@MNPs was incorporated in non-covalent complex with CPPs-oligonucleotide, is developed. Modifying the surface of magnetic nanoparticles with cationic chitosan-modified iron oxide improved the performance of magnetic nanoparticles-CPPs for oligonucleotide delivery. CPPs-CTS@MNPs complexes enhance oligonucleotide transfection compared to CPPs@MNPs or CPPs. The hydrophilic character of CTS@MNPs improves complexation with plasmid pGL3, splice correction oligonucleotides, and small-interfering RNA payload, which consequently resulted in not only strengthening the colloidal stability of the constructed complex but also improving their biocompatibility. Transfection using PF14-splice correction oligonucleotides-CTS@MNPs showed sixfold increase of the transfection compared to splice correction oligonucleotides-PF14 that showed higher transfection than the commercially available lipid-based vector Lipofectamine™ 2000. Nanoscaled CPPs-CTS@MNPs comprise a new family of biomaterials that can circumvent some of the limitations of CPPs or magnetic nanoparticles.


Subject(s)
Cell-Penetrating Peptides/chemistry , Chitosan/analogs & derivatives , Magnetite Nanoparticles/chemistry , Oligonucleotides/administration & dosage , RNA, Small Interfering/administration & dosage , Transfection/methods , Biocompatible Materials/chemistry , Gene Transfer Techniques , HeLa Cells , Humans , Oligonucleotides/genetics , Plasmids/administration & dosage , Plasmids/genetics , RNA Interference , RNA, Small Interfering/genetics
6.
Peptides ; 104: 62-69, 2018 06.
Article in English | MEDLINE | ID: mdl-29684592

ABSTRACT

Delivery of small interfering RNA (siRNA) to suppress glioblastoma growth is a hurdle due to the critical obstacles of the blood-brain barrier and the siRNA properties of such as high negative charges and instability in serum. Therefore, the passage of siRNA to targeted cells is limited. Several siRNA carriers have been constructed using cell-penetrating peptides (CPPs) since the CPPs have shown a high potential for oligonucleotide delivery into the cells. In this study, two CPPs, PepFect 14 (PF14) and the amphipathic peptide PepFect 28 (PF28), were modified with targeting peptides by covalent conjugation and non-covalent complex formation to improve glioma-targeted specificity and gene-silencing efficiency. In conclusion, we have established an efficient non-covalently complexed carrier (PF14:TG1) for siRNA delivery to human glioblastoma cells (U87), showing a significant two-fold increase in gene-silencing efficiency compared to the parent peptide PF14 and also improved specificity to U87 cells compared to non-glioma targeted cells.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Glioblastoma/metabolism , RNA, Small Interfering/administration & dosage , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Cell-Penetrating Peptides/chemistry , Dynamic Light Scattering , Gene Silencing/physiology , Humans , RNA, Small Interfering/chemistry
7.
Biochim Biophys Acta Biomembr ; 1860(2): 491-504, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28962904

ABSTRACT

Peptides able to translocate cell membranes while carrying macromolecular cargo, as cell-penetrating peptides (CPPs), can contribute to the field of drug delivery by enabling the transport of otherwise membrane impermeable molecules. Formation of non-covalent complexes between amphipathic peptides and oligonucleotides is driven by electrostatic and hydrophobic interactions. Here we investigate and quantify the coexistence of distinct molecular species in multiple equilibria, namely peptide monomer, peptide self-aggregates and peptide/oligonucleotide complexes. As a model for the complexes, we used a stearylated peptide from the PepFect family, PF14 and siRNA. PF14 has a cationic part and a lipid part, resembling some characteristics of cationic lipids. Fluorescence correlation spectroscopy (FCS) and fluorescence cross-correlation spectroscopy (FCCS) were used to detect distinct molecular entities in solution and at the plasma membrane of live cells. For that, we labeled the peptide with carboxyrhodamine 6G and the siRNA with Cyanine 5. We were able to detect fluorescent entities with diffusional properties characteristic of the peptide monomer as well as of peptide aggregates and peptide/oligonucleotide complexes. Strategies to avoid peptide adsorption to solid surfaces and self-aggregation were developed and allowed successful FCS measurements in solution and at the plasma membrane. The ratio between the detected molecular species was found to vary with pH, peptide concentration and the proximity to the plasma membrane. The present results suggest that the diverse cellular uptake mechanisms, often reported for amphipathic CPPs, might result from the synergistic effect of peptide monomers, self-aggregates and cargo complexes, distributed unevenly at the plasma membrane.


Subject(s)
Cell Membrane/metabolism , Cell-Penetrating Peptides/metabolism , Protein Aggregates , Spectrometry, Fluorescence/methods , Amino Acid Sequence , Animals , Biological Transport , Carbocyanines/chemistry , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/genetics , Hydrophobic and Hydrophilic Interactions , Lipopeptides/chemistry , Lipopeptides/genetics , Lipopeptides/metabolism , Microscopy, Fluorescence , PC12 Cells , Protein Binding , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rhodamines/chemistry
8.
Sci Rep ; 7(1): 12635, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28974718

ABSTRACT

Cell-penetrating peptides (CPPs) uptake mechanism is still in need of more clarification to have a better understanding of their action in the mediation of oligonucleotide transfection. In this study, the effect on early events (1 h treatment) in transfection by PepFect14 (PF14), with or without oligonucleotide cargo on gene expression, in HeLa cells, have been investigated. The RNA expression profile was characterized by RNA sequencing and confirmed by qPCR analysis. The gene regulations were then related to the biological processes by the study of signaling pathways that showed the induction of autophagy-related genes in early transfection. A ligand library interfering with the detected intracellular pathways showed concentration-dependent effects on the transfection efficiency of splice correction oligonucleotide complexed with PepFect14, proving that the autophagy process is induced upon the uptake of complexes. Finally, the autophagy induction and colocalization with autophagosomes have been confirmed by confocal microscopy and transmission electron microscopy. We conclude that autophagy, an inherent cellular response process, is triggered by the cellular uptake of CPP-based transfection system. This finding opens novel possibilities to use autophagy modifiers in future gene therapy.


Subject(s)
Autophagy/genetics , Cell-Penetrating Peptides/genetics , Lipopeptides/genetics , RNA, Small Interfering/genetics , Cell Membrane/genetics , Cell Membrane/ultrastructure , Genetic Therapy , HeLa Cells , Humans , Microscopy, Electron, Transmission , Oligonucleotides , Transfection
9.
Sci Rep ; 7(1): 9159, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28831162

ABSTRACT

Magnetic nanoparticles (MNPs, Fe3O4) incorporated into the complexes of cell penetrating peptides (CPPs)-oligonucleotides (ONs) promoted the cell transfection for plasmid transfection, splice correction, and gene silencing efficiencies. Six types of cell penetrating peptides (CPPs; PeptFect220 (denoted PF220), PF221, PF222, PF223, PF224 and PF14) and three types of gene therapeutic agents (plasmid (pGL3), splicing correcting oligonucleotides (SCO), and small interfering RNA (siRNA) were investigated. Magnetic nanoparticles incorporated into the complexes of CPPs-pGL3, CPPs-SCO, and CPPs-siRNA showed high cell biocompatibility and efficiently transfected the investigated cells with pGL3, SCO, and siRNA, respectively. Gene transfer vectors formed among PF14, SCO, and MNPs (PF14-SCO-MNPs) showed a superior transfection efficiency (up to 4-fold) compared to the noncovalent PF14-SCO complex, which was previously reported with a higher efficiency compared to commercial vector called Lipofectamine™2000. The high transfection efficiency of the new complexes (CPPs-SCO-MNPs) may be attributed to the morphology, low cytotoxicity, and the synergistic effect of MNPs and CPPs. PF14-pDNA-MNPs is an efficient complex for in vivo gene delivery upon systemic administration. The conjugation of CPPs-ONs with inorganic magnetic nanoparticles (Fe3O4) may open new venues for selective and efficient gene therapy.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Gene Transfer Techniques , Magnetite Nanoparticles/administration & dosage , Oligonucleotides/genetics , Animals , Cell Line , Cell-Penetrating Peptides/chemistry , Drug Delivery Systems , Female , Gene Silencing , HeLa Cells , Humans , Magnetite Nanoparticles/chemistry , Mice , Plasmids/genetics , RNA, Small Interfering/genetics , Transfection
10.
Biochim Biophys Acta Gen Subj ; 1861(9): 2334-2341, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28689990

ABSTRACT

A new strategy for gene transfection using the nanocarrier of cell penetrating peptides (CPPs; PepFect14 (PF14) or PepFect14 (PF14) (PF221)) in complex with graphene oxide (GO) is reported. GO complexed with CPPs and plasmid (pGL3), splice correction oligonucleotides (SCO) or small interfering RNA (siRNA) are performed. Data show adsorption of CPPs and oligonucleotides on the top of the graphenic lamellar without any observed change of the particle size of GO. GO mitigates the cytotoxicity of CPPs and improves the material biocompatibility. Complexes of GO-pGL3-CPPs (CPPs; PF14 or PF221) offer 2.1-2.5 fold increase of the cell transfection compared to pGL3-CPPs (CPPs; PF14 or PF221). GO-SCO-PF14 assemblies effectively transfect the cells with an increase of >10-25 fold compared to the transfection using PF14. The concentration of GO plays a significant role in the material nanotoxicity and the transfection efficiency. The results open a new horizon in the gene treatment using CPPs and offer a simple strategy for further investigations.


Subject(s)
Cell-Penetrating Peptides/chemistry , Graphite/chemistry , Oligonucleotides/administration & dosage , Transfection/methods , Cell Survival , HeLa Cells , Humans , Nanoparticles , Particle Size , Receptors, Scavenger/metabolism
11.
Nucleic Acids Res ; 45(9): 5153-5169, 2017 May 19.
Article in English | MEDLINE | ID: mdl-28334749

ABSTRACT

Huntington's disease (HD) is a fatal, neurodegenerative disorder in which patients suffer from mobility, psychological and cognitive impairments. Existing therapeutics are only symptomatic and do not significantly alter the disease progression or increase life expectancy. HD is caused by expansion of the CAG trinucleotide repeat region in exon 1 of the Huntingtin gene (HTT), leading to the formation of mutant HTT transcripts (muHTT). The toxic gain-of-function of muHTT protein is a major cause of the disease. In addition, it has been suggested that the muHTT transcript contributes to the toxicity. Thus, reduction of both muHTT mRNA and protein levels would ideally be the most useful therapeutic option. We herein present a novel strategy for HD treatment using oligonucleotides (ONs) directly targeting the HTT trinucleotide repeat DNA. A partial, but significant and potentially long-term, HTT knock-down of both mRNA and protein was successfully achieved. Diminished phosphorylation of HTT gene-associated RNA-polymerase II is demonstrated, suggestive of reduced transcription downstream the ON-targeted repeat. Different backbone chemistries were found to have a strong impact on the ON efficiency. We also successfully use different delivery vehicles as well as naked uptake of the ONs, demonstrating versatility and possibly providing insights for in vivo applications.


Subject(s)
Down-Regulation/drug effects , Huntingtin Protein/genetics , Phosphorothioate Oligonucleotides/pharmacology , Trinucleotide Repeat Expansion/genetics , Alleles , DNA/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Knockdown Techniques , Humans , Huntingtin Protein/metabolism , Nucleic Acid Denaturation/drug effects , Peptides/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Restriction Mapping , Ultraviolet Rays
12.
Bioconjug Chem ; 28(3): 782-792, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28209057

ABSTRACT

Modifying cell-penetrating peptides (CPPs) with fatty acids has long been used to improve peptide-mediated nucleic acid delivery. In this study we have revisited this phenomenon with a systematic approach where we developed a structure-activity relationship to describe the role of the acyl chain length in the transfection process. For that we took a well-studied CPP, PepFect14, as the basis and varied its N-terminal acyl chain length from 2 to 22 carbons. To evaluate the delivery efficiency, the peptides were noncovalently complexed with a splice-correcting oligonucleotide (SCO) and tested in HeLa pLuc705 reporter cell line. Our results demonstrate that biological splice-correction activity emerges from acyl chain of 12 carbons and increases linearly with each additional carbon. To assess the underlying factors regarding how the transfection efficacy of these complexes is dependent on hydrophobicity, we used an array of different methods. For the functionally active peptides (C12-22) there was no apparent difference in their physicochemical properties, including complex formation efficiency, hydrodynamic size, and zeta potential. Moreover, membrane activity studies with peptides and their complexes with SCOs confirmed that the toxicity of the complexes at higher molar ratios is mainly caused by the free fraction of the peptide which is not incorporated into the peptide/oligonucleotide complexes. Finally, we show that the increase in splice-correcting activity correlates with the ability of the complexes to associate with the cells. Collectively these studies lay the ground work for how to design highly efficient CPPs and how to optimize their oligonucleotide complexes for lowest toxicity without losing efficiency.


Subject(s)
Cell-Penetrating Peptides/chemistry , Fatty Acids/chemistry , Lipopeptides/chemistry , Oligonucleotides/administration & dosage , Transfection/methods , Acylation , Amino Acid Sequence , Animals , Cattle , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Oligonucleotides/genetics
13.
Mol Ther Nucleic Acids ; 5: e290, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-27111416

ABSTRACT

The advantages of lipid-based transfection reagents have permitted their widespread use in molecular biology and gene therapy. This study outlines the effect of cryo-manipulation of a cationic lipid-based formulation, Lipofectamine 2000, which, after being frozen and thawed, showed orders of magnitude higher plasmid delivery efficiency throughout eight different cell lines, without compromising cell viability. Increased transfection efficiency with the freeze-thawed reagent was also seen with 2'-O-methyl phosphorothioate oligonucleotide delivery and in a splice-correction assay. Most importantly, a log-scale improvement in gene delivery using the freeze-thawed reagent was seen in vivo. Using three different methods, we detected considerable differences in the polydispersity of the different nucleic acid complexes as well as observed a clear difference in their surface spreading and sedimentation, with the freeze-thawed ones displaying substantially higher rate of dispersion and deposition on the glass surface. This hitherto overlooked elevated potency of the freeze-thawed reagent facilitates the targeting of hard-to-transfect cells, accomplishes higher transfection rates, and decreases the overall amount of reagent needed for delivery. Additionally, as we also saw a slight increase in plasmid delivery using other freeze-thawed transfection reagents, we postulate that freeze-thawing might prove to be useful for an even wider variety of transfection reagents.

14.
Int J Pharm ; 501(1-2): 32-8, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-26821060

ABSTRACT

A series of cell-penetrating PepFect peptide analogues was developed by substitutions of the galanin-derived N-terminal sequence. Histidine modifications were incorporated in order to make the peptides pH-responsive. The peptides were all able to form non-covalent complexes with an oligonucleotide cargo by co-incubation in buffer. The complexes were characterized by dynamic light scattering and circular dichroism, and an assay to evaluate the peptide-cargo affinity was developed. Cellular bioactivity was studied in HeLa cells using a luciferase-based splice correction assay. In addition, the membrane interactions of the peptides in large unilammelar vesicles was studied using a calcein leakage assay. The effects of substitutions were found to be dependent of the non-modified, C-terminal sequence of the peptides; for analogues of PepFect 3 we observed an increase in membrane activity and bioactivity for histidine-containing analogues, whereas the same modifications introduced to PepFect 14 lead to a decreased bioactivity. Peptides modified with a leucine/histidine sequence were found to be pH responsive, complexes formed from these peptides were small at pH 7 and grew under acidic conditions. The most promising of the novel PepFect 3 analogues, PepFect 132 has a significantly higher bioactivity and membrane activity than the parent peptide PepFect 3.


Subject(s)
Cell-Penetrating Peptides , Histidine/chemistry , Lipopeptides , Oligonucleotides , Cell Survival/drug effects , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Circular Dichroism , HeLa Cells , Humans , Hydrogen-Ion Concentration , Lipopeptides/administration & dosage , Lipopeptides/chemistry , Lipopeptides/pharmacology , Oligonucleotides/administration & dosage , Oligonucleotides/chemistry , Oligonucleotides/pharmacology
15.
Int J Pharm ; 500(1-2): 128-35, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26773601

ABSTRACT

Receptor-mediated transcytosis remains a major route for drug delivery across the blood-brain barrier (BBB). PepFect 32 (PF32), a peptide-based vector modified with targeting ligand (Angiopep-2) binding to low-density lipoprotein receptor-related protein-1 (LRP-1), was previously found to be a promising vector for plasmid delivery across an in vitro model of the BBB. Cellular uptake of PF32/plasmid DNA (pDNA) complexes was speculated the internalization via LRP-1 receptor. In this study, we prove that PF32/pDNA nanocomplexes are not only transported into brain endothelial cells via LRP-1 receptor-mediated endocytosis, but also via scavenger receptor class A and B (SCARA3, SCARA5, and SR-BI)-mediated endocytosis. SCARA3, SCARA5, and SR-BI are found to be expressed in the brain endothelial cells. Inhibition of these receptors leads to a reduction of the transfection. In conclusion, this study shows that scavenger receptors also play an essential role in the cellular uptake of the PF32/pDNA nanocomplexes.


Subject(s)
Blood-Brain Barrier/metabolism , DNA/administration & dosage , Peptides/administration & dosage , Receptors, Scavenger/metabolism , Animals , Cell Line , DNA/chemistry , Mice , Peptides/chemistry , Plasmids
16.
Methods Mol Biol ; 1324: 39-58, 2015.
Article in English | MEDLINE | ID: mdl-26202261

ABSTRACT

The in silico methods for the prediction of the cell-penetrating peptides are reviewed. Those include the multivariate statistical methods, machine-learning methods such as the artificial neural networks and support vector machines, and molecular modeling techniques including molecular docking and molecular dynamics.The applicability of the methods is demonstrated on the basis of the exemplary cases from the literature.


Subject(s)
Cell-Penetrating Peptides/chemistry , Computing Methodologies , Animals , Artificial Intelligence , Computer Simulation , Humans , Multivariate Analysis , Neural Networks, Computer , Support Vector Machine
17.
Methods Mol Biol ; 1324: 303-15, 2015.
Article in English | MEDLINE | ID: mdl-26202277

ABSTRACT

Nucleic acids can be utilized in gene therapy to restore, alter, or silence gene functions. In order to reveal the biological activity nucleic acids have to reach their intracellular targets by passing through the plasma membrane, which is impermeable for these large and negatively charged molecules. Cell-penetrating peptides (CPPs) condense nucleic acids into nanoparticles using non-covalent complexation strategy and mediate their delivery into the cell, whereas the physicochemical parameters of the nanoparticles determine the interactions with the membranes, uptake mechanism, and subsequent intracellular fate. The nanoparticles are mostly internalized by endocytosis that leads to the entrapment of them in endosomal vesicles. Therefore design of new CPPs that are applicable for non-covalent complex formation strategy and harness endosomolytic properties is highly vital. Here we demonstrate that PepFects and NickFects are efficient vectors for the intracellular delivery of various nucleic acids.This chapter describes how to form CPP/pDNA nanoparticles, evaluate stable nanoparticles formation, and assess gene delivery efficacy.


Subject(s)
Cell-Penetrating Peptides/chemistry , Gene Transfer Techniques , Nanoparticles/chemistry , Nucleic Acids/administration & dosage , Oligonucleotides/administration & dosage , Cell Culture Techniques/methods , Cell-Penetrating Peptides/metabolism , Endocytosis , HeLa Cells , Humans , Indicators and Reagents , Nanoparticles/metabolism , Nucleic Acids/chemistry , Nucleic Acids/genetics , Oligonucleotides/chemistry , Oligonucleotides/genetics , Plasmids/administration & dosage , Plasmids/chemistry , Plasmids/genetics
18.
Nanomedicine ; 11(4): 879-83, 2015 May.
Article in English | MEDLINE | ID: mdl-25659648

ABSTRACT

Extracellular vesicles (EVs) are natural nanoparticles that mediate intercellular transfer of RNA and proteins and are of great medical interest; serving as novel biomarkers and potential therapeutic agents. However, there is little consensus on the most appropriate method to isolate high-yield and high-purity EVs from various biological fluids. Here, we describe a systematic comparison between two protocols for EV purification: ultrafiltration with subsequent liquid chromatography (UF-LC) and differential ultracentrifugation (UC). A significantly higher EV yield resulted from UF-LC as compared to UC, without affecting vesicle protein composition. Importantly, we provide novel evidence that, in contrast to UC-purified EVs, the biophysical properties of UF-LC-purified EVs are preserved, leading to a different in vivo biodistribution, with less accumulation in lungs. Finally, we show that UF-LC is scalable and adaptable for EV isolation from complex media types such as stem cell media, which is of huge significance for future clinical applications involving EVs. FROM THE CLINICAL EDITOR: Recent evidence suggests extracellular vesicles (EVs) as another route of cellular communication. These EVs may be utilized for future therapeutics. In this article, the authors compared ultrafiltration with size-exclusion liquid chromatography (UF-LC) and ultra-centrifugation (UC) for EV recovery.


Subject(s)
Cell-Derived Microparticles/chemistry , Cell-Derived Microparticles/ultrastructure , Chromatography, Gel , HEK293 Cells , Humans , Ultrafiltration
19.
Nucleic Acid Ther ; 25(2): 65-77, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25594433

ABSTRACT

Oligonucleotide analogs have provided novel therapeutics targeting various disorders. However, their poor cellular uptake remains a major obstacle for their clinical development. Negatively charged oligonucleotides, such as 2'-O-Methyl RNA and locked nucleic acids have in recent years been delivered successfully into cells through complex formation with cationic polymers, peptides, liposomes, or similar nanoparticle delivery systems. However, due to the lack of electrostatic interactions, this promising delivery method has been unsuccessful to date using charge-neutral oligonucleotide analogs. We show here that lipid-functionalized cell-penetrating peptides can be efficiently exploited for cellular transfection of the charge-neutral oligonucleotide analog phosphorodiamidate morpholino. The lipopeptides form complexes with splice-switching phosphorodiamidate morpholino oligonucleotide and can be delivered into clinically relevant cell lines that are otherwise difficult to transfect while retaining biological activity. To our knowledge, this is the first study to show delivery through complex formation of biologically active charge-neutral oligonucleotides by cationic peptides.


Subject(s)
Agammaglobulinemia/drug therapy , Genetic Diseases, X-Linked/drug therapy , Morpholinos/administration & dosage , Muscular Atrophy, Spinal/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Nanoparticles , Peptides/administration & dosage , Amino Acid Sequence , Animals , Cells, Cultured , Disease Models, Animal , Mice , Molecular Sequence Data , Morpholinos/therapeutic use , Peptides/chemistry
20.
ACS Nano ; 7(5): 3797-807, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23600610

ABSTRACT

Cell-penetrating peptides (CPPs) are versatile tools for the intracellular delivery of various biomolecules, including siRNA. Recently, CPPs were introduced that showed greatly enhanced delivery efficiency. However, the molecular basis of this increased activity is poorly understood. Here, we performed a detailed analysis of the molecular and physicochemical properties of seven different siRNA-CPP nanoparticles. In addition, we determined which complexes are internalized most efficiently into the leukemia cell-line SKNO-1, and subsequently inhibited the expression of a luciferase reporter gene. We demonstrated effective complexation of siRNA for all tested CPPs, and optimal encapsulation of the siRNA was achieved at very similar molar ratios independent of peptide charge. However, CPPs with an extreme high or low overall charge proved to be exceptions, suggesting an optimal range of charge for CPP-siRNA nanoparticle formation based on opposite charge. The most active CPP (PepFect6) displayed high serum resistance but also high sensitivity to decomplexation by polyanionic macromolecules, indicating the necessity for partial decomplexation for efficient uptake. Surprisingly, CPP-siRNA complexes acquired a negative ζ-potential in the presence of serum. These novel insights shed light on the observation that cell association is necessary but not sufficient for activity and motivate new research into the nature of the nanoparticle-cell interaction. Overall, our results provide a comprehensive molecular basis for the further development of peptide-based oligonucleotide transfection agents.


Subject(s)
Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/metabolism , Drug Carriers/chemistry , Drug Carriers/metabolism , Nanostructures , RNA, Small Interfering/metabolism , Amino Acid Sequence , Base Sequence , Blood Proteins/metabolism , Cell Line, Tumor , Cell-Penetrating Peptides/toxicity , Drug Carriers/toxicity , Humans , Molecular Sequence Data , Protein Stability , RNA, Small Interfering/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...