Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
ACS Appl Mater Interfaces ; 7(43): 24322-9, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26470057

RESUMEN

Small interfering RNA (siRNA) offers a great potential for the treatment of various diseases and disorders. Nevertheless, inefficient in vivo siRNA delivery hampers its translation into the clinic. While numerous successful in vitro siRNA delivery stories exist in reduced-protein conditions, most studies so far overlook the influence of the biological fluids present in the in vivo environment. In this study, we compared the transfection efficiency of liposomal formulations in Opti-MEM (low protein content, routinely used for in vitro screening) and human undiluted ascites fluid obtained from a peritoneal carcinomatosis patient (high protein content, representing the in vivo situation). In Opti-MEM, all formulations are biologically active. In ascites fluid, however, the biological activity of all lipoplexes is lost except for lipofectamine RNAiMAX. The drop in transfection efficiency was not correlated to the physicochemical properties of the nanoparticles, such as premature siRNA release and aggregation of the nanoparticles in the human ascites fluid. Remarkably, however, all of the formulations except for lipofectamine RNAiMAX lost their ability to be taken up by cells following incubation in ascites fluid. To take into account the possible effects of a protein corona formed around the nanoparticles, we recommend always using undiluted biological fluids for the in vitro optimization of nanosized siRNA formulations next to conventional screening in low-protein content media. This should tighten the gap between in vitro and in vivo performance of nanoparticles and ensure the optimal selection of nanoparticles for further in vivo studies.


Asunto(s)
Ascitis/metabolismo , Liposomas/química , Nanopartículas/química , ARN Interferente Pequeño/metabolismo , Línea Celular Tumoral , Femenino , Terapia Genética/métodos , Humanos , Lípidos/química , Nanotecnología/métodos , Metástasis de la Neoplasia , Neoplasias Ováricas/metabolismo , Tamaño de la Partícula , Proteínas/química , Interferencia de ARN , Espectrometría de Fluorescencia , Transfección
2.
J Control Release ; 172(3): 885-93, 2013 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-24075924

RESUMEN

Local extravasation and triggered drug delivery by use of ultrasound and microbubbles is a promising strategy to target drugs to their sites of action. In the past we have developed drug loaded microbubbles by coupling drug containing liposomes to the surface of microbubbles. Until now the advantages of this drug loading strategy have only been demonstrated in vitro. Therefore, in this paper, microbubbles with indocyanine green (ICG) containing liposomes at their surface or a mixture of ICG-liposomes and microbubbles was injected intravenously in mice. Immediately after injection the left hind leg was exposed to 1 MHz ultrasound and the ICG deposition was monitored 1, 4 and 7 days post-treatment by in vivo fluorescence imaging. In mice that received the ICG-liposome loaded microbubbles the local ICG deposition was, at each time point, about 2-fold higher than in mice that received ICG-liposomes mixed with microbubbles. We also showed that the perforations in the blood vessels allow the passage of ICG-liposomes up to 5h after microbubble and ultrasound treatment. An increase in tissue temperature to 41°C was observed in all ultrasound treated mice. However, ultrasound tissue heating was excluded to cause the local ICG deposition. We concluded that coupling of drug containing liposomes to microbubbles may increase ultrasound mediated drug delivery in vivo.


Asunto(s)
Sistemas de Liberación de Medicamentos/instrumentación , Verde de Indocianina/administración & dosificación , Liposomas/química , Microburbujas , Ultrasonido/instrumentación , Animales , Femenino , Ratones
3.
Small ; 9(23): 4027-35, 2013 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-23737360

RESUMEN

One of the main problems in cancer treatment is disease relapse through metastatic colonization, which is caused by circulating tumor cells (CTCs). This work reports on liposome-loaded microbubbles targeted to N-cadherin, a cell-cell adhesion molecule expressed by CTCs. It is shown that such microbubbles can indeed bind to N-cadherin at the surface of HMB2 cells. Interestingly, in a mixture of cells with and without N-cadherin expression, binding of the liposome-loaded microbubbles mainly occurs to the N-cadherin-expressing cells. Importantly, applying ultrasound results in the intracellular delivery of a model drug (loaded in the liposomes) in the N-cadherin-expressing cells only. As described in this paper, such liposome-loaded microbubbles may find application as theranostics and in devices aimed for the specific killing of CTCs in blood.


Asunto(s)
Cadherinas/química , Sistemas de Liberación de Medicamentos/métodos , Liposomas/química , Microburbujas , Humanos
4.
Mol Pharm ; 10(3): 1070-9, 2013 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-23421924

RESUMEN

In cationic carrier-mediated gene delivery, the disproportional relationship between the quantity of delivered DNA and the amount of encoded protein produced is a well-known phenomenon. The numerous intracellular barriers which need to be overcome by pDNA to reach the nucleoplasm play a major role in it. In contrast to what one would expect, a partial replacement of coding pDNA by noncoding DNA does not lead to a decrease in transfection efficiency. The mechanism underlying this observation is still unclear. Therefore, we investigated which constituents of the transfection process might contribute to this phenomenon. Our data reveal that the topology of the noncoding plasmid DNA plays a major role. Noncoding pDNA can be used only in a supercoiled form to replace coding pDNA in Lipofectamine lipoplexes, without a loss in transfection levels. When noncoding pDNA is linearized or partly digested, it diminishes the transfection potential of coding pDNA, as does noncoding salmon DNA. The difference in transfection efficiencies could not be attributed to diverse physicochemical characteristics of the Lipofectamine lipoplexes containing different types of noncoding DNA or to the extent of their internalization. At the level of endosomal release, however, nucleic acid release from the endosomal compartment proceeds faster when lipoplexes contain noncoding salmon DNA. Since the half-life of pDNA in the cytosol hardly exceeds 90 min, it is conceivable that prolonged release of coding pDNA from complexes carrying supercoiled noncoding pDNA may explain its positive effect on transfection, while this depot effect does not exist when noncoding salmon DNA is used.


Asunto(s)
ADN/química , Liposomas/química , Transfección/métodos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos
5.
Nanomedicine (Lond) ; 8(6): 935-49, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23035662

RESUMEN

AIM: The aim of this study was to evaluate the effect of the surface functionalization of model nanoparticles on their mobility in bacterial biofilms and cystic fibrosis sputum. MATERIALS & METHODS: With single-particle tracking microscopy, the mobility of 0.1- and 0.2-µm fluorescent polyethylene glycol (PEG) modified, carboxylate- and N,N-dimethylethylenediamine-modified polystyrene nanospheres were evaluated in fresh cystic fibrosis sputum, as well as Burkholderia multivorans and Pseudomonas aeruginosa biofilms. RESULTS: PEGylation increased the mobility of the particles in sputum and biofilms, while the charged nanospheres were strongly immobilized. However, the transport of the PEGylated nanoparticles was lower in sputum compared with biofilms. Furthermore, the particle transport showed heterogeneity in samples originating from different patients. CONCLUSION: This study's data suggest that for future nanocarrier design it will be essential to combine PEGylation with a targeting moiety to ensure sufficient mobility in mucus and a better accumulation of the nanoparticles in the biofilm.


Asunto(s)
Biopelículas , Burkholderia/fisiología , Fibrosis Quística/metabolismo , Nanopartículas/metabolismo , Pseudomonas aeruginosa/fisiología , Esputo/metabolismo , Adulto , Infecciones por Burkholderia/microbiología , Niño , Femenino , Humanos , Masculino , Movimiento (Física) , Nanopartículas/análisis , Polietilenglicoles/análisis , Polietilenglicoles/metabolismo , Infecciones por Pseudomonas/microbiología , Propiedades de Superficie , Adulto Joven
6.
Bioconjug Chem ; 23(6): 1276-89, 2012 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-22668084

RESUMEN

In this study, we aimed at specific targeting of polycationic amphiphilic cyclodextrins (paCDs) to HepG2 cells via the asialoglycoprotein receptor (ASGPr). The transfection efficiencies of paCDs modified with galactose moieties were evaluated. In preliminary experiments, attempts to transfect HepG2 cells with pDNA complexed with different modified paCDs resulted in very low transfection levels. In additional series of experiments, we found out that nucleic acid/cyclodextrin complexes (CDplexes) were efficiently taken up by the cells and that photochemical internalization, which facilitates release from endosomes, did not improve transfection. Further experiments showed that pDNA can be readily released from the CDplexes when exposed to negatively charged vesicles. These observations imply that the lack of transfection cannot be attributed to a lack of internalization, release of CDplexes from the endosomal compartment, or release of free pDNA from the CDplexes. This in turn suggests that the nuclear entry of the pDNA represents the main limiting factor in the transfection process. To verify that HepG2 cells were transfected with targeted CDplexes containing mRNA, which does not require entry into the nucleus for being translated. With mRNA encoding the green fluorescent protein, fractions of GFP-positive cells of up to 31% were obtained. The results confirmed that the galactosylated complexes are specifically internalized via the ASGPr.


Asunto(s)
Ciclodextrinas/química , Galactosa/química , Hepatocitos/metabolismo , ARN Mensajero/administración & dosificación , Transfección , Receptor de Asialoglicoproteína/metabolismo , Ciclodextrinas/metabolismo , ADN/administración & dosificación , Galactosa/metabolismo , Proteínas Fluorescentes Verdes/genética , Células Hep G2 , Humanos , Plásmidos/administración & dosificación , ARN Mensajero/genética
7.
Biomaterials ; 33(16): 4059-68, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22381475

RESUMEN

Delivery of reprogramming factor-encoding mRNAs by means of lipofection in somatic cells is a desirable method for deriving integration-free iPSCs. However, the lack of reproducibility implies there are major hurdles to overcome before this protocol becomes universally accepted. This study demonstrates the functionality of our in-house synthesized mRNAs expressing the reprogramming factors (OCT4, SOX2, KLF4, c-MYC) within the nucleus of human fibroblasts. However, upon repeated transfections, the mRNAs induced severe loss of cell viability as demonstrated by MTT cytotoxicity assays. Microarray-derived transcriptome data revealed that the poor cell survival was mainly due to the innate immune response triggered by the exogenous mRNAs. We validated the influence of mRNA transfection on key immune response-associated transcript levels, including IFNB1, RIG-I, PKR, IL12A, IRF7 and CCL5, by quantitative real-time PCR and directly compared these with the levels induced by other methods previously published to mediate reprogramming in somatic cells. Finally, we evaluated chemical compounds (B18R, chloroquine, TSA, Pepinh-TRIF, Pepinh-MYD), known for their ability to suppress cellular innate immune responses. However, none of these had the desired effect. The data presented here should provide the basis for further investigations into other immunosuppressing strategies that might facilitate efficient mRNA-mediated cellular reprogramming in human cells.


Asunto(s)
Fibroblastos/citología , ARN Mensajero/genética , Supervivencia Celular , Células Cultivadas , Fibroblastos/inmunología , Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunidad Innata , Factor 4 Similar a Kruppel , Liposomas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
8.
J Control Release ; 158(3): 362-70, 2012 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-22023849

RESUMEN

siRNA therapeutics are currently regarded as promising candidates to make a leap forward in the search for treatments of various hard to cure diseases. In order to exploit the full potential of siRNA based therapeutics, development of delivery systems that can efficiently guide the siRNA molecules to their target without major side effects will be the key to success. Lipid based delivery systems, originating from earlier research in the fields of gene delivery, are the most studied candidates for siRNA delivery. Here we discuss the requirements that need to be met by these siRNA delivery systems to ensure adequate stability after systemic application and subsequent deposition in the target tissue. The encountered hurdles in the blood stream and the solutions proposed in literature are discussed.


Asunto(s)
Portadores de Fármacos/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Animales , Portadores de Fármacos/química , Estabilidad de Medicamentos , Humanos , Liposomas , ARN Interferente Pequeño/química
9.
Biomaterials ; 33(2): 412-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21993235

RESUMEN

The first successful reprogramming of differentiated cells to a pluripotent state was done by retroviral introduction of four transcription factors (Oct4, Sox2, Klf4, cMyc) by the group of Yamanaka in 2006. Since then, scientists all over the world have attempted various methods to avoid insertional mutagenesis, a major limitation of the retrovirus-based method, however no technique was found to completely avoid DNA integration. Recently, a non-viral mRNA-based approach, inherent to avoid genomic integration, was implemented to generate stem cell-like cells, yet, seventeen daily transfections were required, inducing substantial stress on the cells. In this work, we demonstrate successful activation of pluripotency-associated genes in mouse embryonic fibroblasts by means of cationic lipid-mediated introduction of mRNAs encoding the four factors. Moreover, our transfection protocol required maximally three transfections. Up-regulation of the transfected factors as well as Nanog and SSEA-1, typical mouse pluripotency markers, was detected already after the first transfection. Nuclear localization of the introduced factors was confirmed. Positive alkaline phosphatase staining of cell clusters further confirmed the onset of the reprogramming process. In conclusion, the transfection method presented here holds great promise for safe generation of induced pluripotent stem cells of mouse origin.


Asunto(s)
Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , ARN Mensajero/genética , Transfección/métodos , Fosfatasa Alcalina/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Reprogramación Celular , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Antígeno Lewis X/genética , Antígeno Lewis X/metabolismo , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Regulación hacia Arriba
10.
Mol Pharm ; 8(6): 2244-51, 2011 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-22014166

RESUMEN

Microbubbles are Food and Drug Administration (FDA) approved contrast agents for ultrasound imaging. It has been reported that applying ultrasound on drug-loaded microbubbles facilitates drug uptake by cells, due to so-named sonoporation. However, the biophysics behind sonoporation are not fully understood. It is believed that sonoporation results in a "direct" delivery of drugs in the cytoplasm of cells, though it has been suggested as well that sonoporation facilitates endocytosis which would improve the internalization of drugs by cells. To get a better understanding of sonoporation, this study reports on the ultrasound assisted delivery of adeno-associated virus (AAV) loaded on the surface of microbubbles. AAVs rely on endocytosis for efficient transduction of cells and are, consequently, an elegant tool to evaluate whether endocytosis is involved in ultrasound-induced sonoporation. Applying ultrasound on AAV-loaded microbubbles clearly improved the internalization of AAVs by cells, though transduction of the cells did not occur, indicating that by sonoporation substances become directly delivered in the cytosol of cells.


Asunto(s)
Dependovirus , Sistemas de Liberación de Medicamentos , Microburbujas , Terapia por Ultrasonido , Línea Celular Tumoral , Medios de Contraste/química , Endosomas/diagnóstico por imagen , Endosomas/metabolismo , Vectores Genéticos/farmacología , Humanos , Microscopía Confocal , Modelos Biológicos , Estructura Molecular , Polietilenglicoles/química , Sonicación , Ultrasonografía
11.
Mol Pharm ; 8(5): 1757-66, 2011 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-21859089

RESUMEN

The nuclear membrane is one of the major cellular barriers in the delivery of plasmid DNA (pDNA). Cell division has a positive influence on the expression efficiency since, at the end of mitosis, pDNA or pDNA containing complexes near the chromatin are probably included by a random process in the nuclei of the daughter cells. However, very little is known about the nuclear inclusion of nanoparticles during cell division. Using the Xenopus nuclear envelope reassembly (XNER) assay, we found that the nuclear enclosure of nanoparticles was dependent on size (with 100 and 200 nm particles being better included than the 500 nm ones) and charge (with positively charged particles being better included than negatively charged or polyethyleneglycolated (PEGylated) ones) of the beads. Also, coupling chromatin-targeting peptides to the polystyrene beads or pDNA complexes improved their inclusion by 2- to 3-fold. Upon microinjection in living HeLa cells, however, nanoparticles were never observed in the nuclei of cells postdivision but accumulated in a specific perinuclear region, which was identified as the lysosomal compartment. This indicates that nanoparticles can end up in the lysosomes even when they were not delivered through endocytosis. To elucidate if the chromatin binding peptides also have potential in living cells, this additional barrier first has to be tackled, since it prevents free particles from being present near the chromatin at the moment of cell division.


Asunto(s)
Núcleo Celular/metabolismo , Cromatina/metabolismo , ADN/metabolismo , Técnicas de Transferencia de Gen , Nanopartículas/química , Plásmidos/metabolismo , Poliestirenos/química , Animales , Transporte Biológico , Núcleo Celular/ultraestructura , Cromatina/química , Ensamble y Desensamble de Cromatina , ADN/química , Femenino , Células HeLa , Humanos , Masculino , Ensayo de Materiales , Microinyecciones , Microesferas , Membrana Nuclear/metabolismo , Membrana Nuclear/ultraestructura , Óvulo/metabolismo , Tamaño de la Partícula , Espermatozoides/metabolismo , Xenopus laevis
12.
J Control Release ; 152(2): 249-56, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21362448

RESUMEN

Liposome-loaded microbubbles have been recently introduced as a promising drug delivery platform for ultrasound guided drug delivery. In this paper we design liposome-loaded (lipid-shelled) microbubbles through the simple self-assembly of the involved compounds in a single step process. We thoroughly characterized the liposome-loading of the microbubbles and evaluated the cell killing efficiency of this material using doxorubicin (DOX) as a model drug. Importantly, we observed that the DOX liposome-loaded microbubbles allowed killing of melanoma cells even at very low doses of DOX. These findings clearly prove the potential of liposome-loaded microbubbles for ultrasound targeted drug delivery to cancer tissues.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Liposomas , Melanoma/tratamiento farmacológico , Microburbujas , Ultrasonido/métodos , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Humanos , Liposomas/química , Liposomas/metabolismo , Plasma/metabolismo
13.
J Control Release ; 147(3): 385-91, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20708647

RESUMEN

Messenger RNA encoding luciferase (mLUC) was complexed to the cationic lipids Lipofectamine or DOTAP/DOPE, and to the cationic polymer linear poly(ethyleneimine) (linPEI). The complexes were incubated with HeLa cells and luciferase expression was assessed. The type of non-viral carrier used determined the extent and duration of protein expression. Maximal duration of mRNA expression was about 9 days for Lipofectamine complexes, i.e. not very much shorter than with pDNA polyplexes. Interestingly, luciferase activity was already detected 30 min after adding the mRNA complexes to the cells, independent on the type of carrier. We also assessed the proportion of cells that become transfected by means of transfection with an mRNA encoding GFP. For both cationic lipids transfection with mRNA yielded a substantially larger fraction of transfected cells (more than 80%) than transfection with pDNA (40%). In addition we tested the carriers for their ability to mediate delivery of mRNA encoding CXCR4 into mesenchymal stem cells. The fraction of CXCR4-positive cells obtained with the mRNA-cationic lipid complexes was around 80%, as compared to 40% for the linPEI polyplexes. Our results demonstrate that the advantage of the use of mRNA over that of pDNA may under certain conditions outweigh the disadvantage of the somewhat shorter expression period.


Asunto(s)
Lípidos/química , Células Madre Mesenquimatosas/metabolismo , Poliaminas/química , ARN Mensajero/metabolismo , Transfección/métodos , Neoplasias del Cuello Uterino/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Ácidos Grasos Monoinsaturados/química , Femenino , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Células HeLa , Humanos , Cinética , Lípidos/toxicidad , Luciferasas/biosíntesis , Luciferasas/genética , Fosfatidiletanolaminas/química , Plásmidos/metabolismo , Poliaminas/toxicidad , Polielectrolitos , Polietileneimina/química , Compuestos de Amonio Cuaternario/química , ARN Mensajero/química , Receptores CXCR4/biosíntesis , Receptores CXCR4/genética , Neoplasias del Cuello Uterino/genética
14.
Mol Ther ; 18(1): 101-8, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19623162

RESUMEN

Drug delivery with microbubbles and ultrasound is gaining more and more attention in the drug delivery field due to its noninvasiveness, local applicability, and proven safety in ultrasonic imaging techniques. In this article, we tried to improve the cytotoxicity of doxorubicin (DOX)-containing liposomes by preparing DOX-liposome-containing microbubbles for drug delivery with therapeutic ultrasound. In this way, the DOX release and uptake can be restricted to ultrasound-treated areas. Compared to DOX-liposomes, DOX-loaded microbubbles killed at least two times more melanoma cells after exposure to ultrasound. After treatment of the melanoma cells with DOX-liposome-loaded microbubbles and ultrasound, DOX was mainly present in the nuclei of the cancer cells, whereas it was mainly detected in the cytoplasm of cells treated with DOX-liposomes. Exposure of cells to DOX-liposome-loaded microbubbles and ultrasound caused an almost instantaneous cellular entry of the DOX. At least two mechanisms were identified that explain the fast uptake of DOX and the superior cell killing of DOX-liposome-loaded microbubbles and ultrasound. First, exposure of DOX-liposome-loaded microbubbles to ultrasound results in the release of free DOX that is more cytotoxic than DOX-liposomes. Second, the cellular entry of the released DOX is facilitated due to sonoporation of the cell membranes. The in vitro results shown in this article indicate that DOX-liposome-loaded microbubbles could be a very interesting tool to obtain an efficient ultrasound-controlled DOX delivery in vivo.


Asunto(s)
Doxorrubicina/administración & dosificación , Doxorrubicina/farmacología , Microburbujas , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/química , Liposomas/administración & dosificación , Liposomas/química , Melanoma/patología , Ratones , Microscopía Confocal , Modelos Teóricos
15.
Cell Mol Life Sci ; 67(6): 907-18, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19960225

RESUMEN

The present study was set up to investigate the fate of connexin32 and its channels in hepatocellular apoptosis. Primary hepatocyte cultures were exposed to Fas ligand and cycloheximide, and modifications in connexin32 expression and localization, and gap junction functionality were studied. We found that gap junction functionality rapidly declined upon progression of cell death, which was associated with a decay of the gap junctional connexin32 protein pool. Simultaneously, levels of newly synthesized connexin32 protein increased and gathered in a hemichannel configuration. This became particularly evident towards the end stages of the cell death process and was not reflected at the transcriptional level. We next either silenced connexin32 expression or inhibited connexin32 hemichannel activity prior to cell death induction. Both approaches resulted in a delayed termination of the cell death response. We conclude that connexin32 hemichannels facilitate the apoptotic-to-necrotic transition, which typically occurs in the final stage of hepatocellular apoptosis.


Asunto(s)
Apoptosis , Conexinas/metabolismo , Proteína Ligando Fas/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Activación del Canal Iónico , Animales , Apoptosis/efectos de los fármacos , Cicloheximida/farmacología , Hepatocitos/efectos de los fármacos , Humanos , Masculino , Necrosis/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína beta1 de Unión Comunicante
16.
Mol Ther ; 18(3): 561-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20010917

RESUMEN

Nonviral gene complexes can enter mammalian cells through different endocytic pathways. For efficient optimization of the gene carrier it is important to profile its cellular uptake, because this largely determines its intracellular processing and subsequent transfection efficiency. Most of the current information on uptake of these gene-delivery vehicles is based on data following the use of chemical inhibitors of endocytic pathways. Here, we have performed a detailed characterization of four commonly used endocytosis inhibitors [chlorpromazine, genistein, methyl-beta-cyclodextrin (MbetaCD), and potassium depletion] on cell viability and endocytosis in five well-described cell lines. We found that chlorpromazine and to a lesser extent MbetaCD significantly decreased cell viability of some cell lines even after short incubation periods and at concentrations that are routinely used to inhibit endocytosis. Through analyzing the uptake and subcellular distribution of two fluorescent endocytic probes transferrin and lactosylceramide (LacCer) that are reported to enter cells via clathrin-dependent (CDE) and clathrin-independent (CIE) mechanisms, respectively, we showed poor specificity of these agents for inhibiting distinct endocytic pathways. Finally, we demonstrate that any inhibitory effects are highly cell line dependent. Overall, the data question the significance of performing endocytosis studies with these agents in the absence of very stringent controls.


Asunto(s)
Endocitosis , Técnicas de Transferencia de Gen , Animales , Antígenos CD/química , Células COS , Supervivencia Celular , Chlorocebus aethiops , Clatrina/química , Colorantes Fluorescentes/farmacología , Vectores Genéticos , Humanos , Lactosilceramidos/química , Microscopía Fluorescente/métodos , Modelos Genéticos , Transferrina/química , beta-Ciclodextrinas/química
17.
J Control Release ; 141(1): 38-41, 2010 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-19737587

RESUMEN

To exploit the full therapeutic potential of short interfering RNA (siRNA), efficient delivery vehicles are needed as siRNA fails to enter cells spontaneously. Such carriers should also protect siRNA against degradation while it is on its way to the cytosol of the target cells. Cationic polymers are widely investigated as siRNA carriers. Cationic polymers and siRNA self-assemble into siRNA polyplexes which have been shown to silence genes in cell cultures. While siRNA polyplexes will become exposed to full blood after intravenous injection, in vitro gene knockdown is mostly evaluated in serum free media or media containing only a few percent of serum. Little knowledge is currently available on the stability of siRNA polyplexes in blood, while there are no methods available which allow a quantitative measurement of the disassembly of nucleic acid containing nanoparticles in such complex biological media. This paper shows that fluorescence fluctuation spectroscopy allows us to quantitatively monitor the disassembly of siRNA containing nanoparticles in full serum. It further shows that the gene silencing efficacy of siRNA polyplexes in serum containing media can be very well explained by their disassembling behavior in these media. Our findings are important for the further development of siRNA polyplexes and also other nanoparticulate nucleic acid delivery systems.


Asunto(s)
Materiales Biocompatibles/química , Portadores de Fármacos/química , Nanopartículas/química , ARN Interferente Pequeño/sangre , Sistemas de Liberación de Medicamentos , Estabilidad de Medicamentos , Expresión Génica , Silenciador del Gen , Luciferasas de Luciérnaga/genética , Valor Predictivo de las Pruebas , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Espectrometría de Fluorescencia
20.
Langmuir ; 25(9): 4886-91, 2009 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-19341292

RESUMEN

Short interfering RNA (siRNA) holds great potential for the treatment of hard-to-cure diseases. One of the major challenges to translate siRNA into drugs is its efficient delivery to its site-of-action, namely the cytoplasm of the target cells. Cationic liposomes have been shown to do the trick, but their short circulation lifetime and potential aggregation in blood limit their applicability for intravenous administration. These hurdles might be overcome by attaching poly(ethylene glycol) (PEG) at the surface of the cationic liposomes through the use of PEGylated lipids. However, this paper reveals that the classical mixing of siRNA with preformed PEGylated cationic liposomes, as frequently done to load PEGylated liposomes with siRNA, prevents an efficient encapsulation of the siRNA in the liposomes. We show that only a minor fraction of the siRNA becomes encapsulated in the core of the PEGylated liposomes, whereas a major part of the siRNA becomes bound at the liposome's outer surface. In serum, the surface-bound siRNA is immediately released and becomes degraded by serum nucleases. By contrast, hydrating a lipid film (containing PEGylated and cationic lipids) directly with a concentrated solution of siRNA (so-called HYDRA protocol), instead of mixing the siRNA with preformed PEGylated liposomes, encapsulates almost 50% of the siRNA in the core of the PEGylated liposomes, which is the maximal encapsulation efficiency for this type of complexes. We show that the siRNA encapsulated in the core of the thus obtained "HYDRA siPLexes" remains fully encapsulated upon dispersing the PEGylated liposomes in human serum.


Asunto(s)
Liposomas/química , Polietilenglicoles/química , ARN Interferente Pequeño/química , Cápsulas/química , Cationes/química , Humanos , Cinética , Nanoestructuras/química , ARN Interferente Pequeño/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA