Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
FASEB J ; 33(10): 11314-11325, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31311315

RESUMEN

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.


Asunto(s)
Núcleo Celular/genética , Distrofia Miotónica/genética , Oligonucleótidos Antisentido/genética , Células Cultivadas , Humanos , Músculo Esquelético/fisiología , Mioblastos/fisiología , Oligonucleótidos/genética
2.
Nucleic Acids Res ; 45(9): 5153-5169, 2017 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-28334749

RESUMEN

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.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Proteína Huntingtina/genética , Oligonucleótidos Fosforotioatos/farmacología , Expansión de Repetición de Trinucleótido/genética , Alelos , ADN/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Proteína Huntingtina/metabolismo , Desnaturalización de Ácido Nucleico/efectos de los fármacos , Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Mapeo Restrictivo , Rayos Ultravioleta
3.
Biochim Biophys Acta Biomembr ; 1860(2): 491-504, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28962904

RESUMEN

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.


Asunto(s)
Membrana Celular/metabolismo , Péptidos de Penetración Celular/metabolismo , Agregado de Proteínas , Espectrometría de Fluorescencia/métodos , Secuencia de Aminoácidos , Animales , Transporte Biológico , Carbocianinas/química , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/genética , Interacciones Hidrofóbicas e Hidrofílicas , Lipopéptidos/química , Lipopéptidos/genética , Lipopéptidos/metabolismo , Microscopía Fluorescente , Células PC12 , Unión Proteica , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Rodaminas/química
4.
Bioconjug Chem ; 28(3): 782-792, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28209057

RESUMEN

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.


Asunto(s)
Péptidos de Penetración Celular/química , Ácidos Grasos/química , Lipopéptidos/química , Oligonucleótidos/administración & dosificación , Transfección/métodos , Acilación , Secuencia de Aminoácidos , Animales , Bovinos , Células HeLa , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Oligonucleótidos/genética
5.
Biochim Biophys Acta Gen Subj ; 1861(9): 2334-2341, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28689990

RESUMEN

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.


Asunto(s)
Péptidos de Penetración Celular/química , Grafito/química , Oligonucleótidos/administración & dosificación , Transfección/métodos , Supervivencia Celular , Células HeLa , Humanos , Nanopartículas , Tamaño de la Partícula , Receptores Depuradores/metabolismo
6.
Nanomedicine ; 11(4): 879-83, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25659648

RESUMEN

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.


Asunto(s)
Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/ultraestructura , Cromatografía en Gel , Células HEK293 , Humanos , Ultrafiltración
7.
J Biol Chem ; 287(20): 16880-9, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22437827

RESUMEN

Cell-penetrating peptides (CPPs) promote the uptake of different cargo molecules, e.g. therapeutic compounds, making the harnessing of CPPs a promising strategy for drug design and delivery. However, the internalization mechanisms of CPPs are still under discussion, and it is not clear how cells compensate the disturbances induced by peptides in the plasma membrane. In this study, we demonstrate that the uptake of various CPPs enhances the intracellular Ca(2+) levels in Jurkat and HeLa cells. The elevated Ca(2+) concentration in turn triggers plasma membrane blebbing, lysosomal exocytosis, and membrane repair response. Our results indicate that CPPs split into two major classes: (i) amphipathic CPPs that modulate the plasma membrane integrity inducing influx of Ca(2+) and activating downstream responses starting from low concentrations; (ii) non-amphipathic CPPs that do not evoke changes at relevant concentrations. Triggering of the membrane repair response may help cells to replace distorted plasma membrane regions and cells can recover from the influx of Ca(2+) if its level is not drastically elevated.


Asunto(s)
Calcio/metabolismo , Membrana Celular/metabolismo , Péptidos de Penetración Celular/farmacología , Exocitosis/efectos de los fármacos , Lisosomas/metabolismo , Péptidos de Penetración Celular/síntesis química , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/farmacocinética , Células HeLa , Humanos , Células Jurkat
8.
Nucleic Acids Res ; 39(12): 5284-98, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21345932

RESUMEN

Numerous human genetic diseases are caused by mutations that give rise to aberrant alternative splicing. Recently, several of these debilitating disorders have been shown to be amenable for splice-correcting oligonucleotides (SCOs) that modify splicing patterns and restore the phenotype in experimental models. However, translational approaches are required to transform SCOs into usable drug products. In this study, we present a new cell-penetrating peptide, PepFect14 (PF14), which efficiently delivers SCOs to different cell models including HeLa pLuc705 and mdx mouse myotubes; a cell culture model of Duchenne's muscular dystrophy (DMD). Non-covalent PF14-SCO nanocomplexes induce splice-correction at rates higher than the commercially available lipid-based vector Lipofectamine 2000 (LF2000) and remain active in the presence of serum. Furthermore, we demonstrate the feasibility of incorporating this delivery system into solid formulations that could be suitable for several therapeutic applications. Solid dispersion technique is utilized and the formed solid formulations are as active as the freshly prepared nanocomplexes in solution even when stored at an elevated temperatures for several weeks. In contrast, LF2000 drastically loses activity after being subjected to same procedure. This shows that using PF14 is a very promising translational approach for the delivery of SCOs in different pharmaceutical forms.


Asunto(s)
Péptidos de Penetración Celular/química , Lipopéptidos/química , Oligonucleótidos Antisentido/administración & dosificación , Empalme Alternativo , Animales , Péptidos de Penetración Celular/metabolismo , Péptidos de Penetración Celular/toxicidad , Células Cultivadas , Medios de Cultivo , Medio de Cultivo Libre de Suero , Endocitosis , Células HeLa , Humanos , Cinética , Luz , Lipopéptidos/metabolismo , Lipopéptidos/toxicidad , Ratones , Fibras Musculares Esqueléticas/metabolismo , Nanoestructuras/química , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/metabolismo , Dispersión de Radiación , Soluciones , Temperatura
9.
Pharmaceutics ; 13(6)2021 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-34198625

RESUMEN

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.

10.
Biomedicines ; 9(8)2021 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-34440250

RESUMEN

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.

11.
Bioconjug Chem ; 21(4): 774-83, 2010 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-20205419

RESUMEN

Despite increasing interest in cell-penetrating peptides (CPPs) as carriers for drugs and in gene therapy, the current understanding of their exact internalization mechanism is still far from complete. The cellular translocation of CPPs and their payloads has been mostly described by fluorescence- and activity-based methods, leaving the more detailed characterization at the ultrastructural level almost out of attention. Herein, we used transmission electron microscopy to characterize the membrane interaction and internalization of a cell-penetrating peptide S4(13)-PV. We demonstrate that S4(13)-PV peptide forms spherical nanoparticle-like regular structures upon association with cell surface glycosaminoglycans on the plasma membrane. Insertion of S4(13)-PV particles into plasma membrane induces disturbances and leads to the vesicular uptake of peptides by cells. We propose that for efficient cellular translocation S4(13)-PV peptides have to assemble into particles of specific size and shape. The spherical peptide particles are not dissociated in intracellular vesicles but often retain their organization and remain associated with the membrane of vesicles, destabilizing them and promoting the escape of peptides into cytosol. Lowering the temperature and inhibition of dynamins' activity reduce the internalization of S4(13)-PV peptides, but do not block it completely. Our results provide an ultrastructural insight into the interaction mode of CPPs with the plasma membrane and the distribution in cells, which might help to better understand how CPPs cross the biological membranes and gain access into cells.


Asunto(s)
Membrana Celular/metabolismo , Nanopartículas/química , Péptidos/metabolismo , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Células HeLa , Humanos , Microscopía Electrónica de Transmisión , Tamaño de la Partícula , Péptidos/química , Conformación Proteica , Temperatura , Distribución Tisular
12.
FASEB J ; 23(1): 214-23, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18787109

RESUMEN

Although cell-penetrating peptides are able to deliver cargo into cells, their uptake mechanism is still not fully understood and needs to be elucidated to improve their delivery efficiency. Herein, we present evidence of a new mechanism involved in uptake, the membrane repair response. Recent studies have suggested that there might be a direct penetration of peptides in parallel with different forms of endocytosis. The direct penetration of hydrophilic peptides through the hydrophobic plasma membrane, however, is highly controversial. Three proteins involved in target cell apoptosis--perforin, granulysin, and granzymes--share many features common in uptake of cell-penetrating peptides (e.g., they bind proteoglycans). During perforin uptake, the protein activates the membrane repair response, a resealing mechanism triggered in cells with injured plasma membrane, because of extracellular calcium influx. On activation of the membrane repair response, internal vesicles are mobilized to the site of the disrupted plasma membrane, resealing it within seconds. In this study, we have used flow cytometry, fluorescence, and electron microscopy, together with high-performance liquid chromatography and mass spectrometry, to present evidence that the membrane repair response is able to mask damages caused during cell-penetrating peptide uptake, thus preventing leakage of endogenous molecules out of the cell.


Asunto(s)
Membrana Celular/fisiología , Péptidos/metabolismo , Animales , Antígenos de Diferenciación de Linfocitos T/metabolismo , Péptidos Catiónicos Antimicrobianos/metabolismo , Células CHO , Calcio/metabolismo , Calcio/farmacología , Proteínas Portadoras/metabolismo , Membrana Celular/efectos de los fármacos , Péptidos de Penetración Celular , Cricetinae , Cricetulus , Medios de Cultivo , Relación Dosis-Respuesta a Droga , Granzimas/metabolismo , Células HeLa , Humanos , Perforina/metabolismo
13.
Eur J Pharm Biopharm ; 141: 180-190, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31103743

RESUMEN

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.


Asunto(s)
Péptidos de Penetración Celular/administración & dosificación , Lipopéptidos/administración & dosificación , Neoplasias Ováricas/terapia , ARN Mensajero/administración & dosificación , Línea Celular Tumoral , Femenino , Humanos , Nanopartículas/administración & dosificación , Transfección/métodos , Microambiente Tumoral/genética
14.
Biochim Biophys Acta ; 1768(7): 1769-76, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17499577

RESUMEN

Cell-penetrating peptide mediated uptake of labels appears to follow an equilibrium-like process. However, this assumption is only valid if the peptides are stabile. Hence, in this study we investigate intracellular and extracellular peptide degradation kinetics of two fluorescein labeled cell-penetrating peptides, namely MAP and penetratin, in Chinese hamster ovarian cells. The degradation and uptake kinetics were assessed by RP-HPLC equipped with a fluorescence detector. We show that MAP and penetratin are rapidly degraded both extracellularly and intracellularly giving rise to several degradation products. Kinetics indicates that intracellularly, the peptides exist in (at least) two distinct pools: one that is immediately degraded and one that is stabile. Moreover, the degradation could be decreased by treating the peptides with BSA and phenanthroline and the uptake was significantly reduced by cytochalasin B, chloroquine and energy depletion. The results indicate that the extracellular degradation determines the intracellular peptide concentration in this system and therefore the stability of cell-penetrating peptides needs to be evaluated.


Asunto(s)
Membrana Celular/metabolismo , Péptidos/metabolismo , Androstadienos/farmacología , Animales , Células CHO , Proteínas Portadoras/metabolismo , Péptidos de Penetración Celular , Cloroquina/farmacología , Cromatografía Líquida de Alta Presión , Cricetinae , Cricetulus , Citocalasina B/farmacología , Desoxiglucosa/farmacología , Endocitosis/efectos de los fármacos , Leupeptinas/farmacología , Glicoproteínas de Membrana/metabolismo , Modelos Biológicos , Nocodazol/farmacología , Oligopéptidos/metabolismo , Fragmentos de Péptidos/metabolismo , Perforina , Fenantrolinas/farmacología , Fluoruro de Fenilmetilsulfonilo/farmacología , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Inhibidores de Proteasas/farmacología , Transporte de Proteínas , Azida Sódica/farmacología , Wortmanina
15.
J Biomater Appl ; 33(3): 392-401, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30223733

RESUMEN

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.


Asunto(s)
Péptidos de Penetración Celular/química , Quitosano/análogos & derivados , Nanopartículas de Magnetita/química , Oligonucleótidos/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Transfección/métodos , Materiales Biocompatibles/química , Técnicas de Transferencia de Gen , Células HeLa , Humanos , Oligonucleótidos/genética , Plásmidos/administración & dosificación , Plásmidos/genética , Interferencia de ARN , ARN Interferente Pequeño/genética
16.
Peptides ; 104: 62-69, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29684592

RESUMEN

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.


Asunto(s)
Péptidos de Penetración Celular/administración & dosificación , Glioblastoma/metabolismo , ARN Interferente Pequeño/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/genética , Proliferación Celular/fisiología , Péptidos de Penetración Celular/química , Dispersión Dinámica de Luz , Silenciador del Gen/fisiología , Humanos , ARN Interferente Pequeño/química
17.
Sci Rep ; 7(1): 9159, 2017 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-28831162

RESUMEN

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.


Asunto(s)
Péptidos de Penetración Celular/administración & dosificación , Técnicas de Transferencia de Gen , Nanopartículas de Magnetita/administración & dosificación , Oligonucleótidos/genética , Animales , Línea Celular , Péptidos de Penetración Celular/química , Sistemas de Liberación de Medicamentos , Femenino , Silenciador del Gen , Células HeLa , Humanos , Nanopartículas de Magnetita/química , Ratones , Plásmidos/genética , ARN Interferente Pequeño/genética , Transfección
18.
Sci Rep ; 7(1): 12635, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28974718

RESUMEN

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.


Asunto(s)
Autofagia/genética , Péptidos de Penetración Celular/genética , Lipopéptidos/genética , ARN Interferente Pequeño/genética , Membrana Celular/genética , Membrana Celular/ultraestructura , Terapia Genética , Células HeLa , Humanos , Microscopía Electrónica de Transmisión , Oligonucleótidos , Transfección
19.
Biochem Pharmacol ; 71(4): 416-25, 2006 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-16376307

RESUMEN

Resistance to chemotherapy limits the effectiveness of anti-cancer drug treatment. Here, we present a new approach to overcome the setback of drug resistance by designing a conjugate of a cell-penetrating peptide and the cytostatic agent methotrexate (MTX). Two different peptides, YTA2 and YTA4, were designed and their intracellular delivery efficiency was characterized by fluorescence microscopy and quantified by fluorometry. MTX was conjugated to the transport peptides and the ability of the peptide-MTX conjugates to inhibit dihydrofolate reductase, the target enzyme of MTX, was found to be 15 and 20 times less potent than MTX. In addition, in vitro studies were performed in a drug resistant cell model using the 100-fold MTX resistant breast cancer cells MDA-MB-231. At a concentration of 1 microM, the peptide-MTX conjugates were shown to overcome MTX resistance and kill the cells more efficiently than MTX alone. Estimated EC50's were determined for MTX, MTX-YTA2 and YTA2 to be 18.5, 3.8 and 20 microM, respectively. In summary, cell-penetrating peptide conjugation of MTX is a new way of increasing delivery, and thereby, the potency of already well-characterized therapeutic molecules into drug resistant tumour cells.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Metotrexato/farmacología , Oligopéptidos/farmacología , Antimetabolitos Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Fluorometría , Humanos , L-Lactato Deshidrogenasa/metabolismo , Espectrometría de Masas , Microscopía Fluorescente , Oligopéptidos/metabolismo , Péptidos/química , Péptidos/metabolismo , Transporte de Proteínas/efectos de los fármacos , Tetrahidrofolato Deshidrogenasa/metabolismo
20.
Peptides ; 27(7): 1710-6, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16500001

RESUMEN

Cell-penetrating peptides (CPPs) are carriers developed to improve mammalian cell uptake of important research tools such as antisense oligonucleotides and short interfering RNAs. However, the data on CPP uptake into non-mammalian cells are limited. We have studied the uptake and antimicrobial effects of the three representative peptides penetratin (derived from a non-mammalian protein), MAP (artificial peptide) and pVEC (derived from a mammalian protein) using fluorescence HPLC in four common model systems: insect cells (Sf9), gram-positive bacteria (Bacillus megaterium), gram-negative bacteria (Escherichia coli) and yeast (Saccharomyces cerevisiae). We demonstrate that non-mammalian cells internalize CPPs and a comparison of the uptake of the peptides show that the intracellular concentration and degradation of the peptides varies widely among organisms. In addition, these CPPs showed antimicrobial activity.


Asunto(s)
Antiinfecciosos/farmacología , Péptidos/química , Animales , Bacillus megaterium/metabolismo , Línea Celular , Permeabilidad de la Membrana Celular , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Escherichia coli/metabolismo , Insectos , Péptidos/farmacocinética , Saccharomyces cerevisiae/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA