Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Control Release ; 305: 155-164, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31121282

RESUMEN

Anti-angiogenic therapies are promising options for diseases with enhanced vessel formation such as tumors or retinopathies. In most cases, a site-specific local effect on vessel growth is required, while the current focus on systemic distribution of angiogenesis inhibitors may cause severe unwanted side-effects. Therefore, in the current study we have developed an approach for the local inhibition of vascularization, using complexes of lentivirus and magnetic nanoparticles in combination with magnetic fields. Using this strategy in the murine embryonic stem cell (ESC) system, we were able to site-specifically downregulate the protein tyrosine phosphatase SHP2 by RNAi technology in areas with active vessel formation. This resulted in a reduction of vessel development, as shown by reduced vascular tube length, branching points and vascular loops. The anti-angiogenic effect could also be recapitulated in the dorsal skinfold chamber of mice in vivo. Here, site-specific downregulation of SHP2 reduced re-vascularization after wound induction. Thus, we have developed a magnet-assisted, RNAi-based strategy for the efficient local inhibition of angiogenesis in ESCs in vitro and also in vivo.


Asunto(s)
Regulación hacia Abajo , Vectores Genéticos/genética , Lentivirus/genética , Células Madre Embrionarias de Ratones/metabolismo , Neovascularización Fisiológica , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Animales , Línea Celular , Vectores Genéticos/administración & dosificación , Imanes/química , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/terapia , Interferencia de ARN , Transducción Genética/métodos
2.
Biomaterials ; 155: 176-190, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29179133

RESUMEN

Cell replacement in the heart is considered a promising strategy for the treatment of post-infarct heart failure. Direct intramyocardial injection of cells proved to be the most effective application route, however, engraftment rates are very low (<5%) strongly hampering its efficacy. Herein we combine magnetic nanoparticle (MNP) loading of EGFP labeled embryonic cardiomyocytes (eCM) and embryonic stem cell-derived cardiomyocytes (ES-CM) with application of custom designed magnets to enhance their short and long-term engraftment. To optimize cellular MNP uptake and magnetic force within the infarct area, first numerical simulations and experiments were performed in vitro. All tested cell types could be loaded efficiently with SOMag5-MNP (200 pg/cell) without toxic side effects. Application of a 1.3 T magnet at 5 mm distance from the heart for 10 min enhanced engraftment of both eCM and ES-CM by approximately 7 fold at 2 weeks and 3.4 fold (eCM) at 8 weeks after treatment respectively and also strongly improved left ventricular function at all time points. As underlying mechanisms we found that application of the magnetic field prevented the initial dramatic loss of cells via the injection channel. In addition, grafted eCM displayed higher proliferation and lower apoptosis rates. Electron microscopy revealed better differentiation of engrafted eCM, formation of cell to cell contacts and more physiological matrix formation in magnet-treated grafts. These results were corroborated by gene expression data. Thus, combination of MNP-loaded cells and magnet-application strongly increases long-term engraftment of cells addressing a major shortcoming of cardiomyoplasty.


Asunto(s)
Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Animales , Nanopartículas de Magnetita/efectos adversos , Trasplante de Células Madre
3.
Mol Ther ; 25(7): 1616-1627, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28434868

RESUMEN

Hypoxia promotes vascularization by stabilization and activation of the hypoxia inducible factor 1α (HIF-1α), which constitutes a target for angiogenic gene therapy. However, gene therapy is hampered by low gene delivery efficiency and non-specific side effects. Here, we developed a gene transfer technique based on magnetic targeting of magnetic nanoparticle-lentivirus (MNP-LV) complexes allowing site-directed gene delivery to individual wounds in the dorsal skin of mice. Using this technique, we were able to control HIF-1α dependent wound healing angiogenesis in vivo via site-specific modulation of the tyrosine phosphatase activity of SHP-2. We thus uncover a novel physiological role of SHP-2 in protecting HIF-1α from proteasomal degradation via a Src kinase dependent mechanism, resulting in HIF-1α DNA-binding and transcriptional activity in vitro and in vivo. Excitingly, using targeting of MNP-LV complexes, we achieved simultaneous expression of constitutively active as well as inactive SHP-2 mutant proteins in separate wounds in vivo and hereby specifically and locally controlled HIF-1α activity as well as the angiogenic wound healing response in vivo. Therefore, magnetically targeted lentiviral induced modulation of SHP-2 activity may be an attractive approach for controlling patho-physiological conditions relying on hypoxic vessel growth at specific sites.


Asunto(s)
Portadores de Fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Nanopartículas de Magnetita/administración & dosificación , Neovascularización Fisiológica , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Cicatrización de Heridas/genética , Animales , Línea Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Nanopartículas de Magnetita/química , Ratones , Terapia Molecular Dirigida , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteolisis , Piel/lesiones , Piel/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
4.
Methods Mol Biol ; 1522: 257-272, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27837546

RESUMEN

Targeted delivery systems for anticancer drugs are urgently needed to achieve maximum therapeutic efficacy by site-specific accumulation and thereby minimizing adverse effects resulting from systemic distribution of many potent anticancer drugs. We have prepared folate receptor-targeted magnetic liposomes loaded with doxorubicin, which are designed for tumor targeting through a combination of magnetic and biological targeting. Furthermore, these liposomes are designed for hyperthermia-induced drug release to be mediated by an alternating magnetic field and to be traceable by magnetic resonance imaging (MRI). Here, detailed preparation and relevant characterization techniques of targeted magnetic liposomes encapsulating doxorubicin are described.


Asunto(s)
Doxorrubicina/farmacología , Liposomas/química , Magnetismo , Muerte Celular , Citometría de Flujo , Receptores de Folato Anclados a GPI/metabolismo , Ácido Fólico/química , Células HeLa , Humanos , Microscopía Fluorescente , Nanopartículas/química , Fosfatidiletanolaminas/química , Fosfolípidos/química , Polietilenglicoles/química
5.
Sci Rep ; 6: 39149, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27974853

RESUMEN

Modified nucleotide chemistries that increase the half-life (T1/2) of transfected recombinant mRNA and the use of non-native 5'- and 3'-untranslated region (UTR) sequences that enhance protein translation are advancing the prospects of transcript therapy. To this end, a set of UTR sequences that are present in mRNAs with long cellular T1/2 were synthesized and cloned as five different recombinant sequence set combinations as upstream 5'-UTR and/or downstream 3'-UTR regions flanking a reporter gene. Initial screening in two different cell systems in vitro revealed that cytochrome b-245 alpha chain (CYBA) combinations performed the best among all other UTR combinations and were characterized in detail. The presence or absence of CYBA UTRs had no impact on the mRNA stability of transfected mRNAs, but appeared to enhance the productivity of transfected transcripts based on the measurement of mRNA and protein levels in cells. When CYBA UTRs were fused to human bone morphogenetic protein 2 (hBMP2) coding sequence, the recombinant mRNA transcripts upon transfection produced higher levels of protein as compared to control transcripts. Moreover, transfection of human adipose mesenchymal stem cells with recombinant hBMP2-CYBA UTR transcripts induced bone differentiation demonstrating the osteogenic and therapeutic potential for transcript therapy based on hybrid UTR designs.


Asunto(s)
NADPH Oxidasas/genética , ARN Mensajero/metabolismo , Regiones no Traducidas 3' , Regiones no Traducidas 5' , Células A549 , Tejido Adiposo/citología , Animales , Área Bajo la Curva , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Genes Reporteros , Semivida , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , NADPH Oxidasas/metabolismo , Células 3T3 NIH , Osteogénesis , Biosíntesis de Proteínas , Estabilidad del ARN , Curva ROC , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Transfección
6.
J Control Release ; 241: 164-173, 2016 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-27667178

RESUMEN

Gene therapy is a promising approach for chronic disorders that require continuous treatment such as cardiovascular disease. Overexpression of vasoprotective genes has generated encouraging results in animal models, but not in clinical trials. One major problem in humans is the delivery of sufficient amounts of genetic vectors to the endothelium which is impeded by blood flow, whereas prolonged stop-flow conditions impose the risk of ischemia. In the current study we have therefore developed a strategy for the efficient circumferential lentiviral gene transfer in the native endothelium under constant flow conditions. For that purpose we perfused vessels that were exposed to specially designed magnetic fields with complexes of lentivirus and magnetic nanoparticles thereby enabling overexpression of therapeutic genes such as endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF). This treatment enhanced NO and VEGF production in the transduced endothelium and resulted in a reduction of vascular tone and increased angiogenesis. Thus, the combination of MNPs with magnetic fields is an innovative strategy for site-specific and efficient vascular gene therapy.


Asunto(s)
Endotelio Vascular/fisiología , Técnicas de Transferencia de Gen , Vectores Genéticos , Lentivirus/genética , Nanopartículas de Magnetita/química , Animales , Aorta/metabolismo , Aorta/fisiología , Endotelio Vascular/enzimología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Tono Muscular/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , Neovascularización Fisiológica/genética , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
7.
Nanomedicine (Lond) ; 11(14): 1787-800, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27388974

RESUMEN

AIM: To develop a safe and efficient method for targeted, anti-apoptotic gene therapy of corneal endothelial cells (CECs). MATERIALS & METHODS: Magnetofection (MF), a combination of lipofection with magnetic nanoparticles (MNPs; PEI-Mag2, SO-Mag5, PalD1-Mag1), was tested in human CECs and in explanted human corneas. Effects on cell viability and function were investigated. Immunocompatibility was assessed in human peripheral blood mononuclear cells. RESULTS: Silica iron-oxide MNPs (SO-Mag5) combined with X-tremeGENE-HP achieved high transfection efficiency in human CECs and explanted human corneas, without altering cell viability or function. Magnetofection caused no immunomodulatory effects in human peripheral blood mononuclear cells. Magnetofection with anti-apoptotic P35 gene effectively blocked apoptosis in CECs. CONCLUSION: Magnetofection is a promising tool for gene therapy of corneal endothelial cells with potential for targeted on-site delivery.


Asunto(s)
Endotelio Corneal/metabolismo , Magnetismo/métodos , Nanopartículas de Magnetita/química , Dióxido de Silicio/química , Transfección/métodos , Antígenos CD/análisis , Antígenos de Diferenciación de Linfocitos T/análisis , Línea Celular , Línea Celular Tumoral , Supervivencia Celular , ADN/administración & dosificación , ADN/genética , Humanos , Subunidad alfa del Receptor de Interleucina-2/análisis , Lectinas Tipo C/análisis , Leucocitos Mononucleares/citología , Campos Magnéticos , Plásmidos/administración & dosificación , Plásmidos/genética , Proteínas Virales/genética
8.
J Nanomed Nanotechnol ; 7(2)2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27274908

RESUMEN

The goal of magnetic field-assisted gene transfer is to enhance internalization of exogenous nucleic acids by association with magnetic nanoparticles (MNPs). This technique named magnetofection is particularly useful in difficult-to-transfect cells. It is well known that human, mouse, and rat skeletal muscle cells suffer a maturation-dependent loss of susceptibility to Recombinant Adenoviral vector (RAd) uptake. In postnatal, fully differentiated myofibers, the expression of the primary Coxsackie and Adenoviral membrane receptor (CAR) is severely downregulated representing a main hurdle for the use of these vectors in gene transfer/therapy. Here we demonstrate that assembling of Recombinant Adenoviral vectors with suitable iron oxide MNPs into magneto-adenovectors (RAd-MNP) and further exposure to a gradient magnetic field enables to efficiently overcome transduction resistance in skeletal muscle cells. Expression of Green Fluorescent Protein and Insulin-like Growth Factor 1 was significantly enhanced after magnetofection with RAd-MNPs complexes in C2C12 myotubes in vitro and mouse skeletal muscle in vivo when compared to transduction with naked virus. These results provide evidence that magnetofection, mainly due to its membrane-receptor independent mechanism, constitutes a simple and effective alternative to current methods for gene transfer into traditionally hard-to-transfect biological models.

9.
Theranostics ; 5(7): 667-85, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25897333

RESUMEN

Oncolytic viruses are promising new agents in cancer therapy. Success of tumor lysis is often hampered by low intra-tumoral titers due to a strong anti-viral host immune response and insufficient tumor targeting. Previous work on the co-assembly of oncolytic virus particles (VPs) with magnetic nanoparticles (MNPs) was shown to provide shielding from inactivating immune response and improve targeting by external field gradients. In addition, MNPs are detected by magnet resonance imaging (MRI) enabling non-invasive therapy monitoring. In this study two selected core-shell type iron oxide MNPs were assembled with adenovirus (Ad) or vesicular stomatitis virus (VSV). The selected MNPs were characterized by high r2 and r2(*) relaxivities and thus could be quantified non-invasively by 1.5 and 3.0 tesla MRI with a detection limit below 0.001 mM iron in tissue-mimicking phantoms. Assembly and cell internalization of MNP-VP complexes resulted in 81 - 97 % reduction of r2 and 35 - 82 % increase of r2(*) compared to free MNPs. The relaxivity changes could be attributed to the clusterization of particles and complexes shown by transmission electron microscopy (TEM). In a proof-of-principle study the non-invasive detection of MNP-VPs by MRI was shown in vivo in an orthotopic rat hepatocellular carcinoma model. In conclusion, MNP assembly and compartmentalization have a major impact on relaxivities, therefore calibration measurements are required for the correct quantification in biodistribution studies. Furthermore, our study provides first evidence of the in vivo applicability of selected MNP-VPs in cancer therapy.


Asunto(s)
Adenoviridae/metabolismo , Nanopartículas de Magnetita , Viroterapia Oncolítica/métodos , Vesiculovirus/metabolismo , Adenoviridae/genética , Animales , Línea Celular Tumoral , Cricetinae , Cricetulus , Humanos , Masculino , Ratas , Vesiculovirus/genética
10.
Methods Mol Biol ; 1218: 53-106, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25319646

RESUMEN

This chapter describes how to design and conduct experiments to deliver siRNA to adherent cell cultures in vitro by magnetic force-assisted transfection using self-assembled complexes of small interfering RNA (siRNA) and cationic lipids or polymers that are associated with magnetic nanoparticles (MNPs). These magnetic complexes are targeted to the cell surface by the application of a gradient magnetic field. A further development of the magnetic drug-targeting concept is combining it with an ultrasound-triggered delivery using magnetic microbubbles as a carrier for gene or drug delivery. For this purpose, selected MNPs, phospholipids, and siRNAs are assembled in the presence of perfluorocarbon gas into flexible formulations of magnetic lipospheres (microbubbles). Methods are described how to accomplish the synthesis of magnetic nanoparticles for magnetofection and how to test the association of siRNA with the magnetic components of the transfection vector. A simple method is described to evaluate magnetic responsiveness of the magnetic siRNA transfection complexes and estimate the complex loading with magnetic nanoparticles. Procedures are provided for the preparation of magnetic lipoplexes and polyplexes of siRNA as well as magnetic microbubbles for magnetofection and downregulation of the target gene expression analysis with account for the toxicity determined using an MTT-based respiration activity test. A modification of the magnetic transfection triplexes with INF-7, fusogenic peptide, is described resulting in reporter gene silencing improvement in HeLa, Caco-2, and ARPE-19 cells. The methods described can also be useful for screening vector compositions and novel magnetic nanoparticle preparations for optimized siRNA transfection by magnetofection in any cell type.


Asunto(s)
Portadores de Fármacos/química , Nanopartículas de Magnetita/química , Interferencia de ARN , ARN Interferente Pequeño/química , Transfección/métodos , Células CACO-2 , Línea Celular Tumoral , Respiración de la Célula , Células Epiteliales/citología , Células Epiteliales/metabolismo , Fluorocarburos/química , Vectores Genéticos , Células HeLa , Humanos , Iminas/química , Radioisótopos de Yodo , Campos Magnéticos , Microburbujas , Fosfolípidos/química , Plásmidos/química , Plásmidos/metabolismo , Polietilenos/química , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ultrasonido
11.
Neuro Oncol ; 14(10): 1215-26, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22962413

RESUMEN

The Notch pathway is dysregulated and a potential target in glioblastoma multiforme (GBM). Currently available Notch inhibitors block γ-secretase, which is necessary for Notch processing. However, Notch is first cleaved by α-secretase outside the plasma membrane, via a disintegrin and metalloproteinase-10 and -17. In this work, we used a potent α-secretase inhibitor (ASI) to test inhibition of glioblastoma growth and inhibition of Notch and of both novel and known Notch targets. Featured in this study are luciferase reporter assays and immunoblot, microarray analysis, chromatin immunoprecipitation (ChIP), quantitative real-time PCR, cell number assay, bromodeoxyuridine incorporation, plasmid rescue, orthotopic xenograft model, and local delivery of treatment with convection-enhanced delivery using nanoparticles, as well as survival, MRI, and ex vivo luciferase assay. A CBF1-luciferase reporter assay as well as an immunoblot of endogenous Notch revealed Notch inhibition by the ASI. Microarray analysis, quantitative real-time PCR, and ChIP of ASI and γ-secretase inhibitor (GSI) treatment of GBM cells identified known Notch pathway targets, as well as novel Notch targets, including YKL-40 and leukemia inhibitory factor. Finally, we found that local nanoparticle delivery of ASIs but not GSIs increased survival time significantly in a GBM stem cell xenograft treatment model, and ASI treatment resulted in decreased tumor size and Notch activity. This work indicates α-secretase as an alternative to γ-secretase for inhibition of Notch in GBM and possibly other cancers as well, and it identifies novel Notch targets with biologic relevance and potential as biomarkers.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Neoplasias Encefálicas/patología , Proliferación Celular , Glioblastoma/patología , Células Madre Neoplásicas/metabolismo , Receptores Notch/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Ciclo Celular , Inmunoprecipitación de Cromatina , Perfilación de la Expresión Génica , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Humanos , Técnicas In Vitro , Luciferasas/metabolismo , Magnetismo , Ratones , Ratones Endogámicos BALB C , Nanopartículas , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Piperidinas/farmacología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Notch/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Compuestos de Espiro/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Pharm Res ; 29(5): 1308-18, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22271050

RESUMEN

PURPOSE: Targeted delivery of aerosols could not only improve efficacy of inhaled drugs but also reduce side effects resulting from their accumulation in healthy tissue. Here we investigated the impact of magnetized aerosols on model drug accumulation and transgene expression in magnetically targeted lung regions of unanesthetized mice. METHODS: Solutions containing superparamagnetic iron oxide nanoparticles (SPIONs) and model drugs (fluorescein or complexed plasmid DNA) were nebulized to unanesthetized mice under the influence of an external magnetic gradient directed to the lungs. Drug accumulation and transgene expression was subsequently measured at different time points. RESULTS: We could demonstrate 2-3 fold higher accumulation of the model drug fluorescein and specific transgene expression in lung regions of mice which had been exposed to an external magnetic gradient during nebulization compared to the control mice without any exposure to magnetic gradient. CONCLUSIONS: Magnetized aerosols present themselves as an efficient approach for targeted pulmonary delivery of drugs and gene therapeutic agents in order to treat localized diseases of the deeper airways.


Asunto(s)
Aerosoles/química , Sistemas de Liberación de Medicamentos , Compuestos Férricos , Técnicas de Transferencia de Gen , Pulmón/metabolismo , Magnetismo , Nanopartículas del Metal , Animales , Femenino , Fluoresceína/farmacocinética , Fluoresceína/farmacología , Regulación de la Expresión Génica , Vectores Genéticos/genética , Ratones , Ratones Endogámicos BALB C , Plásmidos/genética , Transgenes/genética
13.
Pharm Res ; 29(5): 1344-65, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22222384

RESUMEN

PURPOSE: To optimize silica-iron oxide magnetic nanoparticles with surface phosphonate groups decorated with 25-kD branched polyethylenimine (PEI) for gene delivery. METHODS: Surface composition, charge, colloidal stabilities, associations with adenovirus, magneto-tranduction efficiencies, cell internalizations, in vitro toxicities and MRI relaxivities were tested for the particles decorated with varying amounts of PEI. RESULTS: Moderate PEI-decoration of MNPs results in charge reversal and destabilization. Analysis of space and time resolved concentration changes during centrifugation clearly revealed that at >5% PEI loading flocculation gradually decreases and sufficient stabilization is achieved at >10%. The association with adenovirus occurred efficiently at levels over 5% PEI, resulting in the complexes stable in 50% FCS at a PEI-to-iron w/w ratio of ≥7%; the maximum magneto-transduction efficiency was achieved at 9-12% PEI. Primary silica iron oxide nanoparticles and those with 11.5% PEI demonstrated excellent r(2)* relaxivity values (>600 s(-1)(mM Fe)(-1)) for the free and cell-internalized particles. CONCLUSIONS: Surface decoration of the silica-iron oxide nanoparticles with a PEI-to-iron w/w ratio of 10-12% yields stable aqueous suspensions, allows for efficient viral gene delivery and labeled cell detection by MRI.


Asunto(s)
Compuestos Férricos/química , Técnicas de Transferencia de Gen , Vectores Genéticos/química , Magnetismo , Nanopartículas/química , Dióxido de Silicio/química , Adenoviridae/genética , Animales , Línea Celular , Coloides/química , Estabilidad de Medicamentos , Vectores Genéticos/genética , Humanos , Lentivirus/genética , Imagen por Resonancia Magnética , Ratones , Microscopía Electrónica de Transmisión , Modelos Moleculares , Polietileneimina/química , Ratas , Propiedades de Superficie , Difracción de Rayos X
14.
Pharm Res ; 29(5): 1219-31, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22207207

RESUMEN

PURPOSE: To target adenoviral vectors to cells of the vasculature and shielding vectors from inactivation by the immune system. METHODS: Complexes of reporter gene expressing adenoviral vectors with positively charged magnetic nanoparticles were formed by electrostatic interaction in presence or absence of additional negatively charged poly(ethylene glycol)-based polymer. Transduction of HUVEC was analyzed in vitro under flow. Protection from inactivation by the immune system was analyzed by pre-incubation of AdV and complexes with neutralizing antibodies and subsequent reporter protein analysis of infected cells. RESULTS: Physical association of AdV with MNP and polymers was demonstrated by radioactive labelling of components and co-sedimentation in a magnetic field. Ad-MNP+/-polymer resulted in efficient transduction of HUVEC, depending on MOI and flow rate in presence of magnetic field, whereas no transduction was observed without complex formation with MNP or in absence of magnetic field. Association with MNP did result in protection from neutralizing antibodies, with slightly increased protection provided by the polymer. CONCLUSIONS: Complex formation of AdV with MNP is a viable means for targeting of vectors to areas of magnetic field gradient. Additional coating with polymer might proof useful in protection from inactivation by the immune system.


Asunto(s)
Adenoviridae/genética , Células Endoteliales/fisiología , Magnetismo , Nanopartículas , Transducción Genética/métodos , Adenoviridae/química , Células Endoteliales/química , Células Endoteliales/virología , Eritrocitos/química , Eritrocitos/metabolismo , Vectores Genéticos/química , Vectores Genéticos/genética , Humanos , Nanopartículas/química , Polietilenglicoles/química , Electricidad Estática
15.
Biomaterials ; 33(1): 256-69, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21978891

RESUMEN

Limitations to adenovirus infectivity can be overcome by association with magnetic nanoparticles and enforced infection by magnetic field influence. Here we examined three core-shell-type iron oxide magnetic nanoparticles differing in their surface coatings, particle sizes and magnetic properties for their ability to enhance the oncolytic potency of adenovirus Ad520 and to stabilize it against the inhibitory effects of serum or a neutralizing antibody. It was found that the physicochemical properties of magnetic nanoparticles are critical determinants of the properties which govern the oncolytic productivities of their complexes with Ad520. Although high serum concentration during infection or a neutralizing antibody had strong inhibitory influence on the uptake or oncolytic productivity of the naked virus, one particle type was identified which conferred high protection against both inhibitory factors while enhancing the oncolytic productivity of the internalized virus. This particle type equipped with a silica coating and adsorbed polyethylenimine, displaying a high magnetic moment and high saturation magnetization, mediated a 50% reduction of tumor growth rate versus control upon intratumoral injection of its complex with Ad520 and magnetic field influence, whereas Ad520 alone was inefficient. The correlations between physical properties of the magnetic particles or virus complexes and oncolytic potency are described herein.


Asunto(s)
Adenoviridae/fisiología , Nanopartículas de Magnetita/química , Nanopartículas/química , Virus Oncolíticos/fisiología , Adenoviridae/química , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Nanopartículas de Magnetita/efectos adversos , Nanopartículas de Magnetita/uso terapéutico , Ratones , Ratones Desnudos , Nanopartículas/efectos adversos , Nanopartículas/uso terapéutico , Virus Oncolíticos/química
16.
Adv Drug Deliv Rev ; 63(14-15): 1300-31, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21893135

RESUMEN

Nucleic acids carry the building plans of living systems. As such, they can be exploited to make cells produce a desired protein, or to shut down the expression of endogenous genes or even to repair defective genes. Hence, nucleic acids are unique substances for research and therapy. To exploit their potential, they need to be delivered into cells which can be a challenging task in many respects. During the last decade, nanomagnetic methods for delivering and targeting nucleic acids have been developed, methods which are often referred to as magnetofection. In this review we summarize the progress and achievements in this field of research. We discuss magnetic formulations of vectors for nucleic acid delivery and their characterization, mechanisms of magnetofection, and the application of magnetofection in viral and nonviral nucleic acid delivery in cell culture and in animal models. We summarize results that have been obtained with using magnetofection in basic research and in preclinical animal models. Finally, we describe some of our recent work and end with some conclusions and perspectives.


Asunto(s)
Portadores de Fármacos/química , Nanopartículas de Magnetita/química , Ácidos Nucleicos/administración & dosificación , Transfección , Adenoviridae/genética , Animales , Vectores Genéticos , Humanos , Lentivirus/genética , Magnetismo , Ácidos Nucleicos/genética , Electricidad Estática , Propiedades de Superficie , Transfección/métodos , Transfección/tendencias
17.
Blood ; 117(16): e171-81, 2011 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-21357765

RESUMEN

Research applications and cell therapies involving genetically modified cells require reliable, standardized, and cost-effective methods for cell manipulation. We report a novel nanomagnetic method for integrated cell separation and gene delivery. Gene vectors associated with magnetic nanoparticles are used to transfect/transduce target cells while being passaged and separated through a high gradient magnetic field cell separation column. The integrated method yields excellent target cell purity and recovery. Nonviral and lentiviral magselectofection is efficient and highly specific for the target cell population as demonstrated with a K562/Jurkat T-cell mixture. Both mouse and human enriched hematopoietic stem cell pools were effectively transduced by lentiviral magselectofection, which did not affect the hematopoietic progenitor cell number determined by in vitro colony assays. Highly effective reconstitution of T and B lymphocytes was achieved by magselectofected murine wild-type lineage-negative Sca-1(+) cells transplanted into Il2rg(-/-) mice, stably expressing GFP in erythroid, myeloid, T-, and B-cell lineages. Furthermore, nonviral, lentiviral, and adenoviral magselectofection yielded high transfection/transduction efficiency in human umbilical cord mesenchymal stem cells and was fully compatible with their differentiation potential. Upscaling to a clinically approved automated cell separation device was feasible. Hence, once optimized, validated, and approved, the method may greatly facilitate the generation of genetically engineered cells for cell therapies.


Asunto(s)
Separación Celular/métodos , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Células Madre Hematopoyéticas/citología , Células Madre Mesenquimatosas/citología , Animales , Antígenos Ly/genética , Vectores Genéticos/química , Células Madre Hematopoyéticas/metabolismo , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Células Jurkat , Células K562 , Magnetismo , Proteínas de la Membrana/genética , Células Madre Mesenquimatosas/metabolismo , Ratones , Nanopartículas/química , Transfección
18.
Ther Deliv ; 2(6): 717-26, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22822504

RESUMEN

Nucleic acid delivery to cells to make them produce a desired protein or to shut down the expression of endogenous genes opens unique possibilities for research and therapy. During the last decade, to realize the potential of this approach, nanomagnetic methods for delivering and targeting nucleic acids have been developed, methods which are often referred to as Magnetofection. Our research group at the Institute of Experimental Oncology and Therapy Research, located at the University Hospital Klinikum rechts der Isar in the center of Munich, Germany, develops new magnetic nanomaterials and, their formulations with gene-delivery vectors and technologies to allow localized and efficient gene delivery in vitro and in vivo for a variety of research, diagnostic and therapeutic applications.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Campos Magnéticos , Nanopartículas del Metal/química , Adenoviridae/metabolismo , Terapia Genética/veterinaria , Vectores Genéticos/metabolismo , Humanos , Nanopartículas del Metal/uso terapéutico , Ingeniería de Tejidos/métodos
19.
J Gene Med ; 12(9): 747-54, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20821745

RESUMEN

BACKGROUND: Lentiviral (LV) vectors are able to only slowly and inefficiently transduce nondividing cells such as those of the airway epithelium. To address this issue, we have exploited the magnetofection technique in in vitro models of airway epithelium. METHODS: Magnetofectins were formed by noncovalent interaction between LV particles and polycation-coated iron oxide nanoparticles. Efficiency of LV-mediated transduction (as evaluated through green fluorescent protein (GFP) expression by cytofluorimetric analysis) was measured in bronchial epithelial cells in the presence or absence of a magnetic field. Cytotoxicity was evaluated by lactate dehydrogenase (LDH) release; cell monolayer integrity by measurement of transepithelial resistance (TER) and evaluation of correct zonula occludens-1 (ZO-1) localization at tight junctions (TJs) by immunofluorescence and confocal microscopy. RESULTS: In nonpolarized cells, magnetofectins enhanced LV-mediated transduction at multiplicity of infection (MOI) of 50 up to 3.9-fold upon a 24-h incubation, to levels that approached those achieved at MOI of 200 for LV alone, in the presence or absence of the magnetic field. Magnetofection significantly increased the percentage of transduced cells up to 186-fold already after 15 min of incubation. In polarized cells, magnetofection increased GFP+ cells up to 24-fold compared to LV alone. Magnetofection did not enhance LDH release and slightly altered TER but not ZO-1 localization at the TJs. CONCLUSIONS: We conclude that magnetofection can facilitate in vitro LV-mediated transduction of airway epithelial cells, in the absence of overt cytotoxicity and maintaining epithelial integrity, by lowering the necessary vector dose and reducing the incubation time required to achieve efficient transduction.


Asunto(s)
Células Epiteliales/metabolismo , Técnicas de Transferencia de Gen , Lentivirus/genética , Magnetismo , Mucosa Respiratoria/metabolismo , Polaridad Celular , Supervivencia Celular , Células Cultivadas , Vectores Genéticos/administración & dosificación , Proteínas Fluorescentes Verdes/metabolismo , Humanos , L-Lactato Deshidrogenasa/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteína de la Zonula Occludens-1
20.
Mol Pharm ; 7(4): 1069-89, 2010 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-20550160

RESUMEN

Oncolytic adenoviruses rank among the most promising innovative agents in cancer therapy. We examined the potential of boosting the efficacy of the oncolytic adenovirus dl520 by associating it with magnetic nanoparticles and magnetic-field-guided infection in multidrug-resistant (MDR) cancer cells in vitro and upon intratumoral injection in vivo. The virus was complexed by self-assembly with core-shell nanoparticles having a magnetite core of about 10 nm and stabilized by a shell containing 68 mass % lithium 3-[2-(perfluoroalkyl)ethylthio]propionate) and 32 mass % 25 kDa branched polyethylenimine. Optimized virus binding, sufficiently stable in 50% fetal calf serum, was found at nanoparticle-to-virus ratios of 5 fg of Fe per physical virus particle (VP) and above. As estimated from magnetophoretic mobility measurements, 3,600 to 4,500 magnetite nanocrystallites were associated per virus particle. Ultrastructural analysis by electron and atomic force microscopy showed structurally intact viruses surrounded by magnetic particles that occasionally bridged several virus particles. Viral uptake into cells at a given virus dose was enhanced 10-fold compared to nonmagnetic virus when infections were carried out under the influence of a magnetic field. Increased virus internalization resulted in a 10-fold enhancement of the oncolytic potency in terms of the dose required for killing 50% of the target cells (IC(50) value) and an enhancement of 4 orders of magnitude in virus progeny formation at equal input virus doses compared to nonmagnetic viruses. Furthermore, the full oncolytic effect developed within two days postinfection compared with six days in a nonmagnetic virus as a reference. Plotting target cell viability versus internalized virus particles for magnetic and nonmagnetic virus showed that the inherent oncolytic productivity of the virus remained unchanged upon association with magnetic nanoparticles. Hence, we conclude that the mechanism of boosting the oncolytic effect by magnetic force is mainly due to the improved internalization of magnetic virus complexes resulting in potentiated virus progeny formation. Upon intratumoral injection and application of a gradient magnetic field in a murine xenograft model, magnetic virus complexes exhibited a stronger oncolytic effect than adenovirus alone. We propose that this approach would be useful during in vivo administration to tumor-feeding blood vessels to boost the efficacy of the primary infection cycle within the tumor. For systemic application, further modification of magnetic adenovirus complexes for shielding and retargeting of the whole magnetic virus complex entity is needed.


Asunto(s)
Adenoviridae/fisiología , Magnetismo , Nanopartículas , Virus Oncolíticos/fisiología , Neoplasias Pancreáticas/terapia , Adenoviridae/genética , Animales , Southern Blotting , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Electrónica de Transmisión , Virus Oncolíticos/genética , Neoplasias Pancreáticas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA