Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hum Gene Ther ; 35(1-2): 59-69, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38062776

RESUMEN

Despite decades of research in adeno-associated virus (AAV) and the role of adenovirus in production, the interplay of AAV and adenovirus is not fully understood. Specific regions of the adenoviral genome containing E1, E2a, E4 open reading frame (ORF), and VA RNA have been demonstrated as necessary for AAV production; however, incorporating these regions into either a producer cell line or subcloning into an Ad helper plasmid may lead to inclusion of neighboring adenoviral sequence or ORFs with unknown function. Because AAV is frequently used in gene therapies, removing excessive adenovirus sequences improves the Ad helper plasmid size and manufacturability, and may lead to safer vectors for patients. Furthermore, deepening our understanding of the helper virus genes required for recombinant AAV (rAAV) production has the potential to increase yields and manufacturability of rAAV for clinical and commercial applications. One region continuously included in various Ad helper plasmid iterations is the adenoviral E2a promoter region that appears to be necessary for E2a expression. Due to the compact nature of viral genomes, the E2a promoter region overlaps with the Hexon Assembly/100K protein and the L4 region. The L4 region, which contains the coding sequences for 22K and 33K proteins, had not been thought to be necessary for AAV production. Through molecular techniques, this study demonstrates that the adenoviral 22K protein is essential for rAAV production in HEK293 cells by triple transfection and that the 33K protein synergistically increases rAAV yield.


Asunto(s)
Adenoviridae , Dependovirus , Humanos , Dependovirus/genética , Dependovirus/metabolismo , Adenoviridae/genética , Células HEK293 , Plásmidos , Transfección , Proteínas Virales/genética , Vectores Genéticos/genética
2.
J Control Release ; 358: 498-509, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37127076

RESUMEN

The therapeutic potential of many gene therapies is limited by their inability to cross the blood brain barrier (BBB). While intranasal administration of plasmid DNA nanoparticles (NPs) offers a non-invasive approach to bypass the BBB, it is not targeted to disease-relevant brain regions. Here, our goal was to determine whether focused ultrasound (FUS) can enrich intranasal delivery of our plasmid DNA NPs to target deeper brain regions, in this case the regions most affected in Parkinson's disease. Combining FUS with intranasal administration resulted in enhanced delivery of DNA NPs to the rodent brain, by recruitment and transfection of microglia. FUS increased transgene expression by over 3-fold after intranasal administration compared to intravenous administration. Additionally, FUS with intranasal delivery increased transgene expression in the sonicated hemisphere by over 80%, altered cellular transfection patterns at the sonication sites, and improved penetration of plasmid NPs into the brain parenchyma (with a 1-fold and 3-fold increase in proximity of transgene expression to neurons in the forebrain and midbrain respectively, and a 40% increase in proximity of transgene expression to dopaminergic neurons in the substantia nigra). These results provide evidence in support of using FUS to improve transgene expression after intranasal delivery of non-viral gene therapies.


Asunto(s)
Encéfalo , Nanopartículas , Administración Intranasal , Encéfalo/metabolismo , Barrera Hematoencefálica/metabolismo , ADN , Transgenes , Microburbujas , Sistemas de Liberación de Medicamentos/métodos
3.
Nanomedicine ; 16: 20-33, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30472323

RESUMEN

The intranasal route of administration allows large therapeutics to circumvent the blood-brain barrier and be delivered directly to the CNS. Here we examined the distribution and pattern of cellular transfection, and the time course of transgene expression, in the rat brain after intranasal delivery of plasmid DNA nanoparticles (NPs) encoding hGDNF fused with eGFP. Intranasal administration of these NPs resulted in transfection and transgene expression throughout the rat brain, as indicated by eGFP ELISA and eGFP-positive cell counts. Most of the transfected cells were abluminal and immediately adjacent to capillaries and are likely pericytes, consistent with their distribution by perivascular transport. Intranasal administration of these plasmid DNA NPs resulted in significant, long-term transgene expression in rat brain, with highest levels at 1 week and continued expression for 6 months. These results provide evidence in support of intranasal DNA NPs as a non-invasive, long-term gene therapy approach for various CNS disorders.


Asunto(s)
Encéfalo/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Plásmidos/genética , Administración Intranasal , Animales , Sistemas de Liberación de Medicamentos , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Masculino , Microscopía , Ratas , Ratas Sprague-Dawley , Transfección/métodos
4.
Mol Neurobiol ; 56(1): 688-701, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29779176

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) gene therapy could offer a disease-modifying treatment for Parkinson's disease (PD). Here, we report that plasmid DNA nanoparticles (NPs) encoding human GDNF administered intranasally to rats induce transgene expression in the brain and protect dopamine neurons in a model of PD. To first test whether intranasal administration could transfect cells in the brain, rats were sacrificed 1 week after intranasal pGDNF NPs or the naked plasmid. GDNF ELISA revealed significant increases in GDNF expression throughout the brain for both treatments. To assess whether expression was sufficient to protect dopamine neurons, naked pGDNF and pGDNF DNA NPs were given intranasally 1 week before a unilateral 6-hydroxydopamine lesion in a rat model of PD. Three to four weeks after the lesion, amphetamine-induced rotational behavior was reduced, and dopaminergic fiber density and cell counts in the lesioned substantia nigra and nerve terminal density in the lesioned striatum were significantly preserved in rats given intranasal pGDNF. The NPs afforded a greater level of neuroprotection than the naked plasmid. These results provide proof-of-principle that intranasal administration of pGDNF DNA NPs can offer a non-invasive, non-viral gene therapy approach for early-stage PD.


Asunto(s)
ADN/administración & dosificación , Factor Neurotrófico Derivado de la Línea Celular Glial/administración & dosificación , Factor Neurotrófico Derivado de la Línea Celular Glial/uso terapéutico , Nanopartículas/administración & dosificación , Neuroprotección , Enfermedad de Parkinson/prevención & control , Plásmidos/administración & dosificación , Administración Intranasal , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Humanos , Masculino , Factores de Crecimiento Nervioso , Oxidopamina , Ratas Sprague-Dawley , Sustancia Negra/metabolismo , Sustancia Negra/patología
5.
Adv Exp Med Biol ; 1074: 109-115, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721934

RESUMEN

Considerable progress has been made in the design and delivery of non-viral gene therapy vectors, but, like their viral counterparts, therapeutic levels of transgenes have not met the requirements for successful clinical applications so far. The biggest advantage of polymer-based nanoparticle vectors is the ease with which they can be modified to increase their ability to penetrate the cell membrane and target specific cells by simply changing the formulation of the nanoparticle compaction. We took advantage of this characteristic to improve transfection rates of our particles to meet the transgene levels which will be needed for future treatment of patients. For this study, we successfully investigated the possibility of our established pegylated polylysine particles to be administered via intravitreal rather than subretinal route to ease the damage during injection. We also demonstrated that our particles are flexible enough to sustain changes in the formulation to accommodate additional targeting sequences without losing their efficiency in transfecting neuronal cells in the retina. Together, these results give us the opportunity to even further improve our particles.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Inyecciones Intraoculares/métodos , Nanopartículas/administración & dosificación , Polilisina/administración & dosificación , Epitelio Pigmentado de la Retina/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Animales , ADN Recombinante/administración & dosificación , Inyecciones Intravítreas , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Tamaño de la Partícula , Polietilenglicoles/administración & dosificación , Epitelio Pigmentado de la Retina/citología , Transgenes
6.
Nanomedicine ; 13(7): 2209-2217, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28666950

RESUMEN

This study reports proof of concept for administering compacted DNA nanoparticles (DNPs) intracerebrally as a means to protect against neurotoxin-induced neurodegeneration of dopamine (DA) neurons. In this study we used DNPs that encoded for human glial cell line-derived neurotrophic factor (hGDNF); GDNF is a potent neurotrophic factor for DA neurons. Intracerebral injections of DNPs into the striatum and/or substantia nigra were performed 1 week before treatment with a neurotoxin. We observed that the number of surviving DA cells, the density of DA fiber terminals and recovery of motor function were greater in animals injected with GDNF-encoding DNPs than in control animals receiving DNPs encoding for an inert reporter gene. The results of these studies are one of the first to demonstrate that a non-viral, synthetic nanoparticle can be used to deliver therapeutic genes to cells in the brain as a means to protect cells against neurotoxin-induced neurodegeneration.


Asunto(s)
ADN/administración & dosificación , ADN/genética , Neuronas Dopaminérgicas/citología , Técnicas de Transferencia de Gen , Terapia Genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Neuroprotección , Animales , Supervivencia Celular , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Terapia Genética/métodos , Humanos , Masculino , Nanopartículas/administración & dosificación , Ratas Sprague-Dawley
7.
Mol Ther ; 20(1): 63-72, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21952168

RESUMEN

Efficient and prolonged human cystic fibrosis transmembrane conductance regulator (hCFTR) expression is a major goal for cystic fibrosis (CF) lung therapy. A hCFTR expression plasmid was optimized as a payload for compacted DNA nanoparticles formulated with polyethylene glycol (PEG)-substituted 30-mer lysine peptides. A codon-optimized and CpG-reduced hCFTR synthetic gene (CO-CFTR) was placed in a polyubiquitin C expression plasmid. Compared to hCFTR complementary DNA (cDNA), CO-CFTR produced a ninefold increased level of hCFTR protein in transfected HEK293 cells and, when compacted as DNA nanoparticles, produced a similar improvement in lung mRNA expression in Balb/c and fatty acid binding protein promoter (FABP) CF mice, although expression duration was transient. Various vector modifications were tested to extend duration of CO-CFTR expression. A novel prolonged expression (PE) element derived from the bovine growth hormone (BGH) gene 3' flanking sequence produced prolonged expression of CO-CFTR mRNA at biologically relevant levels. A time course study in the mouse lung revealed that CO-CFTR mRNA did not change significantly, with CO-CFTR/mCFTR geometric mean ratios of 94% on day 2, 71% on day 14, 53% on day 30, and 14% on day 59. Prolonged CO-CFTR expression is dependent on the orientation of the PE element and its transcription, is not specific to the UbC promoter, and is less dependent on other vector backbone elements.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , ADN/química , Vectores Genéticos/genética , Pulmón/metabolismo , Región de Flanqueo 3' , Administración Intranasal , Animales , Fibrosis Quística/genética , Fibrosis Quística/terapia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Proteínas de Unión a Ácidos Grasos/genética , Expresión Génica , Regulación de la Expresión Génica , Orden Génico , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/química , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Nanoconjugados/química , Nanoconjugados/uso terapéutico , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Transcripción Genética
8.
Mol Imaging ; 10(5): 327-39, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21521549

RESUMEN

In this study, we used bioluminescence imaging (BLI) to track long-term transgene activity following the transfection of brain cells using a nonviral gene therapy technique. Formulations of deoxyribonucleic acid (DNA) combined with 30-mer lysine polymers (substituted with 10 kDa polyethylene glycol) form nanoparticles that transfect brain cells in vivo and produce transgene activity. Here we show that a single intracerebral injection of these DNA nanoparticles (DNPs) into the rat cortex, striatum, or substantia nigra results in long-term and persistent luciferase transgene activity over an 8- to 11-week period as evaluated by in vivo BLI analysis, and single injections of DNPs into the mouse striatum showed stable luciferase transgene activity for 1 year. Compacted DNPs produced in vivo signals 7- to 34-fold higher than DNA alone. In contrast, ex vivo BLI analysis, which is subject to less signal quenching from surrounding tissues, demonstrated a DNP to DNA alone ratio of 76- to 280-fold. Moreover, the ex vivo BLI analysis confirmed that signals originated from the targeted brain structures. In summary, BLI permits serial analysis of luciferase transgene activity at multiple brain locations following gene transfer with DNPs. Ex vivo analysis may permit more accurate determination of relative activities of gene transfer vectors.


Asunto(s)
Encéfalo/fisiología , ADN/administración & dosificación , Técnicas de Transferencia de Gen , Mediciones Luminiscentes/métodos , Nanopartículas/administración & dosificación , Análisis de Varianza , Animales , Encéfalo/metabolismo , Química Encefálica , Vectores Genéticos , Histocitoquímica/métodos , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Microinyecciones , Ratas , Ratas Sprague-Dawley , Transgenes
9.
Cell Transplant ; 18(10): 1183-96, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19650971

RESUMEN

Previously it was established that infusion of glial cell line-derived neurotrophic factor (GDNF) protein into grafts of embryonic dopamine cells has a neurotrophic effect on the grafted cells. In this study we used a nonviral technique to transfer the gene encoding for GDNF to striatal cells. Plasmid DNA encoding for GDNF was compacted into DNA nanoparticles (DNPs) by 10 kDa polyethylene glycol (PEG)-substituted lysine 30-mers (CK(30)PEG10k) and then injected into the denervated striatum of rats with unilateral 6-hydroxydopamine lesions. Sham controls were injected with saline. One week later, experimental animals received either a ventral mesencephalic (VM) tissue chunk graft or a cell suspension VM graft implanted into the denervated striatum. Grafts were allowed to integrate for 4-6 weeks and during this period we monitored spontaneous and drug-induced motor activity. Using stereological cell counting we observed a 16-fold increase in the number of surviving TH(+) cells within tissue chunk grafts placed into the striatum pretreated with pGDNF DNPs (14,923 +/- 4,326) when compared to grafts placed into striatum pretreated with saline (955 +/- 343). Similarly, we observed a sevenfold increase in the number of TH(+) cells within cell suspension grafts placed into the striatum treated with pGDNF DNPs when compared to cell suspension grafts placed into the saline dosed striatum. Behaviorally, we observed significant improvement in rotational scores and in spontaneous forepaw usage of the affected forelimb in grafted animals receiving prior treatment with compacted pGDNF DNPs when compared to grafted animals receiving saline control pretreatment. Data analysis for protein, morphological, and behavioral measures suggests that compacted pGDNF DNPs injected into the striatum can result in transfected cells overexpressing GDNF protein at levels that provide neurotrophic support for grafted embryonic dopamine neurons.


Asunto(s)
Trasplante de Tejido Encefálico , Cuerpo Estriado/metabolismo , ADN/administración & dosificación , Dopamina/metabolismo , Trasplante de Tejido Fetal , Técnicas de Transferencia de Gen , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Nanopartículas/química , Neuronas/trasplante , Animales , Conducta Animal , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Masculino , Mesencéfalo/trasplante , Actividad Motora/efectos de los fármacos , Neuronas/citología , Neuronas/metabolismo , Enfermedad de Parkinson/terapia , Plásmidos/metabolismo , Polietilenglicoles/química , Polilisina/química , Ratas , Ratas Sprague-Dawley , Prueba de Desempeño de Rotación con Aceleración Constante
10.
Mol Ther ; 17(4): 641-50, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19223866

RESUMEN

This study demonstrates proof of concept for delivery and expression of compacted plasmid DNA in the central nervous system. Plasmid DNA was compacted with polyethylene glycol substituted lysine 30-mer peptides, forming rod-like nanoparticles with diameters between 8 and 11 nm. Here we show that an intracerebral injection of compacted DNA can transfect both neurons and glia, and can produce transgene expression in the striatum for up to 8 weeks, which was at least 100-fold greater than intracerebral injections of naked DNA plasmids. Bioluminescent imaging (BLI) of injected animals at the 11th postinjection week revealed significantly higher transgene activity in animals receiving compacted DNA plasmids when compared to animals receiving naked DNA. There was minimal evidence of brain inflammation. Intrastriatal injections of a compacted plasmid encoding for glial cell line-derived neurotrophic factor (pGDNF) resulted in a significant overexpression of GDNF protein in the striatum 1-3 weeks after injection.


Asunto(s)
Encéfalo/metabolismo , ADN/administración & dosificación , Nanopartículas , Transgenes , Animales , Secuencia de Bases , ADN/genética , Cartilla de ADN , Inmunohistoquímica , Hibridación in Situ , Luciferasas/genética , Plásmidos , Ratas , Transducción Genética
11.
J Pharmacol Toxicol Methods ; 54(1): 42-55, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16326118

RESUMEN

INTRODUCTION: The hERG (human ether-a-go-go related gene) potassium channel is required for normal cardiac repolarization, is susceptible to inhibition by a wide variety of compounds, and its blockage can lead to cardiac QT interval prolongation and life threatening arrhythmias. The present report examines the ability of hERG binding and functional assays to identify compounds with potential cardiovascular liabilities at the earliest stages of drug discovery. METHODS: Competitive binding assays were developed using (3)H-dofetilide and membranes from HEK293EBNA cells stably expressing recombinant hERG (HEK293-hERG) and IMR-32 cells expressing hERG endogenously. hERG functional assays were also developed using membrane potential indicator dye and rubidium efflux. The ability of these assays to identify compounds with potential adverse cardiac effects was examined using drugs with known cardiac effects ranging from those with no known adverse effects to drugs that were withdrawn from the market due to increased risk of sudden death associated with Torsades de Points. RESULTS: Binding assays using HEK293-hERG membranes and (3)H-dofetilide were robust (Z'=0.69+/-0.015, mean+/-S.E.M.), highly reproducible (test-retest slope=1.04, r(2)=0.98), and correlated well with IC(50) values obtained by patch clamp (slope=0.98, r(2)=0.89). Binding assays using IMR-32 membranes were less sensitive (Z'=0.4+/-0.03, mean+/-S.E.M., false negative rate=0.4) but still correlated well with patch clamp data (slope=1.06, r(2)=0.83). The hERG membrane potential assay could detect potent hERG inhibitors (defined by hERG patch clamp IC(50)<0.1 muM) using HEK293-hERG cells, but were prone to generate false-negative results with less potent inhibitors (false negative rate=0.5). Finally, the rubidium efflux assay gave highly reproducible results (Z'=0.80+/-0.02, mean+/-S.E.M.) that correlated with patch clamp IC(50) values (slope=0.87, r(2)=0.73). DISCUSSION: The hERG binding and rubidium efflux assays are robust, predictive of patch clamp results, and can be used at the earliest stages of drug discovery.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/metabolismo , Ensayo de Unión Radioligante/métodos , Proteínas Recombinantes/metabolismo , Animales , Células CHO , Línea Celular , Cricetinae , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/biosíntesis , Humanos , Unión Proteica/fisiología , Proteínas Recombinantes/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA