Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Mol Cell ; 44(3): 361-72, 2011 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-22055183

RESUMEN

The maintenance of H3K9 and DNA methylation at imprinting control regions (ICRs) during early embryogenesis is key to the regulation of imprinted genes. Here, we reveal that ZFP57, its cofactor KAP1, and associated effectors bind selectively to the H3K9me3-bearing, DNA-methylated allele of ICRs in ES cells. KAP1 deletion induces a loss of heterochromatin marks at ICRs, whereas deleting ZFP57 or DNMTs leads to ICR DNA demethylation. Accordingly, we find that ZFP57 and KAP1 associated with DNMTs and hemimethylated DNA-binding NP95. Finally, we identify the methylated TGCCGC hexanucleotide as the motif that is recognized by ZFP57 in all ICRs and in several tens of additional loci, several of which are at least ZFP57-dependently methylated in ES cells. These results significantly advance our understanding of imprinting and suggest a general mechanism for the protection of specific loci against the wave of DNA demethylation that affects the mammalian genome during early embryogenesis.


Asunto(s)
Ensamble y Desensamble de Cromatina , Metilación de ADN , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Impresión Genómica , Proteínas Nucleares/metabolismo , Motivos de Nucleótidos , Proteínas Represoras/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT , Línea Celular , Proteínas Cromosómicas no Histona/metabolismo , Metilasas de Modificación del ADN/metabolismo , Técnicas de Inactivación de Genes , N-Metiltransferasa de Histona-Lisina , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Proteína Metiltransferasas/metabolismo , Proteínas Represoras/genética , Proteína 28 que Contiene Motivos Tripartito , Ubiquitina-Proteína Ligasas
2.
Genome Res ; 24(8): 1260-70, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24879559

RESUMEN

Reverse transcription-derived sequences account for at least half of the human genome. Although these retroelements are formidable motors of evolution, they can occasionally cause disease, and accordingly are inactivated during early embryogenesis through epigenetic mechanisms. In the mouse, at least for endogenous retroviruses, important mediators of this process are the tetrapod-specific KRAB-containing zinc finger proteins (KRAB-ZFPs) and their cofactor TRIM28. The present study demonstrates that KRAB/TRIM28-mediated regulation is responsible for controlling a very broad range of human-specific endogenous retroelements (EREs) in human embryonic stem (ES) cells and that it exerts, as a consequence, a marked effect on the transcriptional dynamics of these cells. It further reveals reciprocal dependence between TRIM28 recruitment at specific families of EREs and DNA methylation. It finally points to the importance of persistent TRIM28-mediated control of ERE transcriptional impact beyond their presumed inactivation by DNA methylation.


Asunto(s)
Metilación de ADN , Proteínas Represoras/fisiología , Elementos Alu , Animales , Línea Celular , Células Madre Embrionarias , Retrovirus Endógenos/genética , Regulación de la Expresión Génica , Humanos , Ratones , Transcripción Genética , Proteína 28 que Contiene Motivos Tripartito
3.
Hepatology ; 56(4): 1279-90, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22684873

RESUMEN

UNLABELLED: The liver is characterized by sexually dimorphic gene expression translating into sex-specific differences in lipid, drug, steroid hormone, and xenobiotic metabolism, with distinct responses of males and females to environmental challenges. Here, we investigated the role of the Krüppel-associated box (KRAB)-associated protein 1 (KAP1) epigenetic regulator in this process. Liver-specific KAP1 knockout (KO) led to strikingly sexually dimorphic phenotypic disturbances, including male-predominant steatosis and hepatic tumors with up-regulation of protein kinase B and extracellular signal-related kinases 1/2 mitogen-activated protein kinase signaling. This correlated with the sex-specific transcriptional dysregulation of a wide range of metabolic genes, notably those involved in retinol and sex hormone processing as well as in detoxification. Furthermore, chromatin immunoprecipitation followed by deep sequencing indicated that a number of dysregulated genes are direct targets of the KRAB/KAP1 repression system. Those genes include sexually dimorphic cytochrome P 450 Cyp2d9, glutathione S-transferase π, Cyp2a, Cyp2b, and Cyp3a gene clusters. Additionally, we identified a male-restricted KAP1-binding site in the fat-specific protein 27 gene, correlating with its male-predominant up-regulation upon Kap1 deletion, suggesting that the latter might be an important trigger in the development of male-specific hepatosteatosis and secondary tumorigenesis. CONCLUSION: This work reveals KRAB/KAP1-mediated transcriptional regulation as a central event in metabolic control hormones, drugs, and xenobiotics in the liver and further links disturbances in these processes with hepatic carcinogenesis.


Asunto(s)
Adenoma/genética , Transformación Celular Neoplásica/genética , Hígado Graso/genética , Predisposición Genética a la Enfermedad , Neoplasias Hepáticas/genética , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Adenoma/patología , Animales , Biopsia con Aguja , Transformación Celular Neoplásica/patología , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Hígado Graso/patología , Femenino , Regulación de la Expresión Génica , Inmunohistoquímica , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria , Sensibilidad y Especificidad , Factores Sexuales , Proteína 28 que Contiene Motivos Tripartito , Dedos de Zinc/genética
4.
J Comp Neurol ; 530(11): 1773-1949, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35303367

RESUMEN

The thalamic paraventricular nucleus (PVT) is a structure highly interconnected with several nuclei ranging from forebrain to hypothalamus and brainstem. Numerous rodent studies have examined afferent and efferent connections of the PVT and their contribution to behavior, revealing its important role in the integration of arousal cues. However, the majority of these studies used a region-oriented approach, without considering the neuronal subtype diversity of the nucleus. In the present study, we provide the anatomical and transcriptomic characterization of a subpopulation of PVT neurons molecularly defined by the expression of glucokinase (Gck). Combining a genetically modified mouse model with viral tracing approaches, we mapped both the anterograde and the retrograde projections of Gck-positive neurons of the anterior PVT (GckaPVT ). Our results demonstrated that GckaPVT neurons innervate several nuclei throughout the brain axis. The strongest connections are with forebrain areas associated with reward and stress and with hypothalamic structures involved in energy balance and feeding regulation. Furthermore, transcriptomic analysis of the Gck-expressing neurons revealed that they are enriched in receptors for hypothalamic-derived neuropeptides, adhesion molecules, and obesity and diabetes susceptibility transcription factors. Using retrograde labeling combined with immunohistochemistry and in situ hybridization, we identify that GckaPVT neurons receive direct inputs from well-defined hypothalamic populations, including arginine-vasopressin-, melanin-concentrating hormone-, orexin-, and proopiomelanocortin-expressing neurons. This detailed anatomical and transcriptomic characterization of GckaPVT neurons provides a basis for functional studies of the integration of homeostatic and hedonic aspects of energy homeostasis, and for deciphering the potential role of these neurons in obesity and diabetes development.


Asunto(s)
Glucoquinasa , Núcleos Talámicos de la Línea Media , Animales , Glucoquinasa/genética , Glucoquinasa/metabolismo , Ratones , Núcleos Talámicos de la Línea Media/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Tálamo/metabolismo
5.
BMC Genomics ; 12: 378, 2011 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-21791101

RESUMEN

BACKGROUND: KRAB-ZFPs (Krüppel-associated box domain-zinc finger proteins) are vertebrate-restricted transcriptional repressors encoded in the hundreds by the mouse and human genomes. They act via an essential cofactor, KAP1, which recruits effectors responsible for the formation of facultative heterochromatin. We have recently shown that KRAB/KAP1 can mediate long-range transcriptional repression through heterochromatin spreading, but also demonstrated that this process is at times countered by endogenous influences. METHOD: To investigate this issue further we used an ectopic KRAB-based repressor. This system allowed us to tether KRAB/KAP1 to hundreds of euchromatic sites within genes, and to record its impact on gene expression. We then correlated this KRAB/KAP1-mediated transcriptional effect to pre-existing genomic and chromatin structures to identify specific characteristics making a gene susceptible to repression. RESULTS: We found that genes that were susceptible to KRAB/KAP1-mediated silencing carried higher levels of repressive histone marks both at the promoter and over the transcribed region than genes that were insensitive. In parallel, we found a high enrichment in euchromatic marks within both the close and more distant environment of these genes. CONCLUSION: Together, these data indicate that high levels of gene activity in the genomic environment and the pre-deposition of repressive histone marks within a gene increase its susceptibility to KRAB/KAP1-mediated repression.


Asunto(s)
Silenciador del Gen , Genómica , Proteínas Represoras/metabolismo , Transcripción Genética/genética , Cromatina/genética , Células HeLa , Histonas/genética , Humanos , Proteína 28 que Contiene Motivos Tripartito
6.
Stem Cells ; 28(8): 1390-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20641037

RESUMEN

Hematopoietic stem cells (HSC) are probably the best understood somatic stem cells and often serve as a paradigm for other stem cells. Nevertheless, most current techniques to genetically manipulate them in vivo are either constitutive and/or induced in settings of hematopoietic stress such as after irradiation. Here, we present a conditional expression system that allows for externally controllable transgenesis and knockdown in resident HSCs, based on a lentiviral vector containing a tet-O sequence and a transgenic mouse line expressing a doxycyclin-regulated tTR-KRAB repressor protein. HSCs harvested from tTR-KRAB mice are transduced with the lentiviral vector containing a cDNA (i.e., Green Fluorescent Protein (GFP)) and/or shRNA (i.e., p53) of interest and then transplanted into lethally irradiated recipients. While the vector is effectively repressed by tTR-KRAB during homing and engraftment, robust GFP/shp53 expression is induced on doxycyclin treatment in HSCs and their progeny. Doxycylin-controllable transcription is maintained on serial transplantation, indicating that repopulating HSCs are stably modified by this approach. In summary, this easy to implement conditional system provides inducible and reversible overexpression or knock down of genes in resident HSCs in vivo using a drug devoid of toxic or activating effects.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , ARN Interferente Pequeño/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proliferación Celular , Células Cultivadas , Doxiciclina/farmacología , Femenino , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Vectores Genéticos/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Lentivirus/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/genética
7.
Mol Ther ; 18(5): 1002-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20179674

RESUMEN

Duchenne muscular dystrophy (DMD) is characterized by the absence of dystrophin. Several previous studies demonstrated the feasibility of delivering microdystrophin complementary DNA (cDNA) into mouse and normal nonhuman primate muscles by ex vivo gene therapy. However, these animal models do not reproduce completely the human DMD phenotype, while the dystrophic dog model does. To progress toward the use of the best animal model of DMD, a dog microdystrophin was transduced into human and dystrophic dog muscle precursor cells (MPCs) with a lentivirus before their transplantation into mouse muscles. One month following MPC transplantation, myofibers expressing the dog microdystrophin were observed. We also used another approach to introduce this transgene into myofibers, i.e., the electrotransfer of a plasmid coding for the dog microdystrophin. The plasmid was injected into mouse and dog muscles, and brief electric pulses were applied in the region of injection. Two weeks later, the transgene was detected in both animals. Therefore, ex vivo gene therapy and electrotransfer are two possible methods to introduce a truncated version of dystrophin into myofibers of animal models and eventually into myofibers of DMD patients.


Asunto(s)
Distrofina/metabolismo , Animales , Western Blotting , Línea Celular , Perros , Distrofina/genética , Terapia Genética , Humanos , Lentivirus/genética , Ratones , Ratones Mutantes , Músculos/citología , Músculos/metabolismo , Distrofia Muscular de Duchenne/terapia , Plásmidos/genética , Células Madre/citología , Células Madre/metabolismo
8.
Diabetes ; 69(11): 2253-2266, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32839348

RESUMEN

The ventromedial nucleus of the hypothalamus (VMN) is involved in the counterregulatory response to hypoglycemia. VMN neurons activated by hypoglycemia (glucose-inhibited [GI] neurons) have been assumed to play a critical although untested role in this response. Here, we show that expression of a dominant negative form of AMPK or inactivation of AMPK α1 and α2 subunit genes in Sf1 neurons of the VMN selectively suppressed GI neuron activity. We found that Txn2, encoding a mitochondrial redox enzyme, was strongly downregulated in the absence of AMPK activity and that reexpression of Txn2 in Sf1 neurons restored GI neuron activity. In cell lines, Txn2 was required to limit glucopenia-induced reactive oxygen species production. In physiological studies, absence of GI neuron activity after AMPK suppression in the VMN had no impact on the counterregulatory hormone response to hypoglycemia or on feeding. Thus, AMPK is required for GI neuron activity by controlling the expression of the antioxidant enzyme Txn2. However, the glucose-sensing capacity of VMN GI neurons is not required for the normal counterregulatory response to hypoglycemia. Instead, it may represent a fail-safe system in case of impaired hypoglycemia sensing by peripherally located glucose detection systems that are connected to the VMN.


Asunto(s)
Glucosa/metabolismo , Hipoglucemia/sangre , Neuronas/fisiología , Tiorredoxinas/metabolismo , Núcleo Hipotalámico Ventromedial/citología , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Glucemia , Células Cultivadas , Humanos , Técnicas de Placa-Clamp , Tiorredoxinas/genética
9.
Curr Gene Ther ; 6(6): 625-32, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17168695

RESUMEN

Ex vivo gene therapy is a possible treatment for several muscular dystrophies. The best transgene to be expressed and the appropriate cell type to be used currently remain the subject of many investigations. The most adequate gene modification technique also remains to be established. Different transgenes have already been tested in animal models and transgenic mice. Several cell types were evaluated during the last decades and several vectors or transfection methods were analysed. From these essays, over time, several proofs of principles were made to demonstrate the feasibility of this type of therapy. For DMD, it is possible to express several truncated versions of dystrophin or exon skipping molecules. It is also possible to express other molecules that would mitigate the phenotype. Different cell types are also available. From the well documented myoblasts to the AC133 positive cells, the choice of cell types is exploding. Gene modification also evolved during the last decade. Efficient transfection technique and viral vectors are currently available. Given all these possibilities, the researcher has to make several choices. This review is trying to give clues of how to make those choices.


Asunto(s)
Terapia Genética/métodos , Animales , Expresión Génica , Terapia Genética/tendencias , Vectores Genéticos , Humanos , Técnicas In Vitro , Músculos/metabolismo , Distrofias Musculares/terapia
10.
Cell Rep ; 17(7): 1795-1806, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27829151

RESUMEN

The counterregulatory response to hypoglycemia, which restores normal blood glucose levels to ensure sufficient provision of glucose to the brain, is critical for survival. To discover underlying brain regulatory systems, we performed a genetic screen in recombinant inbred mice for quantitative trait loci (QTL) controlling glucagon secretion in response to neuroglucopenia. We identified a QTL on the distal part of chromosome 7 and combined this genetic information with transcriptomic analysis of hypothalami. This revealed Fgf15 as the strongest candidate to control the glucagon response. Fgf15 was expressed by neurons of the dorsomedial hypothalamus and the perifornical area. Intracerebroventricular injection of FGF19, the human ortholog of Fgf15, reduced activation by neuroglucopenia of dorsal vagal complex neurons, of the parasympathetic nerve, and lowered glucagon secretion. In contrast, silencing Fgf15 in the dorsomedial hypothalamus increased neuroglucopenia-induced glucagon secretion. These data identify hypothalamic Fgf15 as a regulator of glucagon secretion.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Pruebas Genéticas , Glucagón/metabolismo , Hipotálamo/metabolismo , Envejecimiento , Animales , Cromosomas de los Mamíferos/metabolismo , Desoxiglucosa/farmacología , Silenciador del Gen/efectos de los fármacos , Genoma , Hipotálamo/efectos de los fármacos , Ratones Endogámicos C57BL , Sistema Nervioso Parasimpático/efectos de los fármacos , Sistema Nervioso Parasimpático/metabolismo , Sitios de Carácter Cuantitativo/genética
11.
Cell Rep ; 2(4): 766-73, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-23041315

RESUMEN

De novo DNA methylation is an essential aspect of the epigenetic reprogramming that takes place during early development, yet factors responsible for its instatement at particular genomic loci are poorly defined. Here, we demonstrate that the KRAB-ZFP-mediated recruitment of KAP1 to DNA in embryonic stem cells (ESCs) induces cytosine methylation. This process is preceded by H3K9 trimethylation, and genome-wide analyses reveal that it spreads over short distances from KAP1-binding sites so as to involve nearby CpG islands. In sharp contrast, in differentiated cells, KRAB/KAP1-induced heterochromatin formation does not lead to DNA methylation. Correspondingly, the methylation status of CpG islands in the adult mouse liver correlates with their proximity to KAP1-binding sites in ESCs, not in hepatocytes. Therefore, KRAB-ZFPs and their cofactor KAP1 are in part responsible for the establishment during early embryogenesis of site-specific DNA methylation patterns that are maintained through development.


Asunto(s)
Proteínas Portadoras/metabolismo , Metilación de ADN , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Animales , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT , Línea Celular , Cromatina/metabolismo , Islas de CpG , Desarrollo Embrionario , Células Madre Embrionarias/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Ratones , Proteína 28 que Contiene Motivos Tripartito , Ubiquitina-Proteína Ligasas
12.
Cell Transplant ; 19(5): 589-96, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20650035

RESUMEN

Duchenne muscular dystrophy (DMD) is the most frequent muscular dystrophy. Currently, there is no cure for the disease. The transplantation of muscle precursor cells (MPCs) is one of the possible treatments, because it can restore the expression of dystrophin in DMD muscles. In this study, we investigated the effects of myoblasts injected with cardiotoxin on the contractile properties and resistance to eccentric contractions of transplanted and nontransplanted muscles. We used the extensor digitorum longus (EDL) as a model for our study. We conclude that the sole presence of dystrophin in a high percentage of muscle fibers is not sufficient by itself to increase the absolute or the specific force in the EDL of transplanted mdx muscle. This lack of strength increase may be due to the extensive damage that was produced by the cardiotoxin, which was coinjected with the myoblasts. However, the dystrophin presence is sufficient to protect muscle from eccentric damage as indicated by the force drop results.


Asunto(s)
Distrofina/biosíntesis , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Musculares/trasplante , Músculo Esquelético/lesiones , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/terapia , Animales , Distrofina/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Células Musculares/citología , Contracción Muscular/fisiología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología
13.
Mol Ther ; 15(2): 431-8, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17235323

RESUMEN

Duchenne muscular dystrophy (DMD) is characterized by the absence of dystrophin. We tested the ability of lentiviral vectors to deliver a transgene into myogenic cells before their transplantation. Enhanced green fluorescent protein (eGFP) transgene was efficiently transferred into cells and eGFP-positive fibers were generated following transplantation. An eGFP-micro-dystrophin transgene under the control of a cytomegalovirus promoter was then transferred with the same viral vector but caused some toxicity to the mono-nucleated cells. We then used instead a muscle creatine kinase promoter. Dystrophin expression was observed in the muscle fibers after the transplantation of such genetically modified cells into mdx and severe combined immunodeficient mice. Micro-dystrophin expression was also observed in monkey muscles a month after allogenic or autologous transplantation of genetically modified myoblasts. Therapeutic exon skipping was induced by infecting myoblasts of a DMD patient, deleted for dystrophin exons 49 and 50, with a lentivirus expressing a U7 small nuclear RNA containing antisense sequences against exon 51. The modification led to correct exon skipping and to the expression of a quasi-dystrophin in vitro and in vivo. These results demonstrate the feasibility of lentiviral-based ex vivo gene therapy for DMD.


Asunto(s)
Distrofina/genética , Lentivirus/genética , Células Musculares/trasplante , Distrofia Muscular Animal/terapia , Trasplante de Células Madre/métodos , Animales , Western Blotting , Línea Celular , Modelos Animales de Enfermedad , Distrofina/metabolismo , Terapia Genética/métodos , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Haplorrinos , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones SCID , Células Musculares/citología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Biochem Biophys Res Commun ; 319(1): 12-20, 2004 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-15158435

RESUMEN

Tat protein from human immunodeficiency virus can deliver biologically active proteins in vivo and is of considerable interest for protein therapeutics. The mechanism responsible for Tat-fusion protein internalization is still poorly understood and controversial. The punctuate distribution, timing, and temperature sensitivity observed in our experiments with Tat-fusion proteins are consistent with endocytosis. After a few hours, Tat-fusion proteins accumulated around the nucleus without any significant visible nuclear targeting. Using a Cre/Lox based functional assay, lysosomotropic agents known to disrupt endosome integrity, increased by up to 23-fold the nuclear delivery of functional Tat-Cre recombinase without increasing cell uptake in a similar fashion. This shows that endosome disruption can significantly increase Tat-fusion protein access to the cytosol and nucleus. In addition, we found that internalized Tat-fusion proteins persisted several hours and that inhibitors of lysosome acidification did not increase functional nuclear delivery of Tat-Cre. This suggests that Tat-fusion proteins enter via the endosomal pathway, circumvent lysosomal degradation, and are then sequestered in the periphery of the nucleus. Most importantly, our work indicates that an inadequate intracellular trafficking is the main factor limiting the efficiency of protein cargo delivery using Tat.


Asunto(s)
Núcleo Celular/metabolismo , Endosomas/fisiología , Productos del Gen tat/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Animales , Cloroquina/farmacología , Citosol/metabolismo , Relación Dosis-Respuesta a Droga , Endocitosis , Endosomas/metabolismo , Inhibidores Enzimáticos/farmacología , Técnicas de Transferencia de Gen , Immunoblotting , Integrasas/metabolismo , Cinética , Leupeptinas/farmacología , Lisosomas/metabolismo , Macrólidos/farmacología , Ratones , Células 3T3 NIH , Sacarosa/farmacología , Temperatura , Factores de Tiempo , Proteínas Virales/metabolismo
15.
Mol Ther ; 10(4): 679-87, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15451452

RESUMEN

Ex vivo gene therapy offers a potential treatment for Duchenne muscular dystrophy by transfection of the dystrophin gene into the patient's own myogenic precursor cells, followed by transplantation. We used nucleofection to introduce DNA plasmids coding for enhanced green fluorescent protein (eGFP) or eGFP-dystrophin fusion protein and the phage phiC31 integrase into myogenic cells and to integrate these genes into a limited number of sites in the genome. Using a plasmid expressing eGFP, we transfected 50% of a mouse muscle-derived stem cell line and 60% of normal human myoblasts. Co-nucleofection of a plasmid expressing the phiC31 integrase and an eGFP expression plasmid containing an attB sequence produced 15 times more frequent stable expression, because of site-specific integration of the transgene. Co-nucleofection of the phiC31 integrase plasmid and a large plasmid containing the attB sequence and the gene for an eGFP-full-length dystrophin fusion protein produced fluorescent human myoblasts that were able to form more intensely fluorescent myotubes after 1 month of culture. A nonviral approach combining nucleofection and the phiC31 integrase may eventually permit safe autotransplantation of genetically modified cells to patients.


Asunto(s)
Distrofina/genética , Electroporación/métodos , Integrasas/genética , Mioblastos/metabolismo , Transfección/métodos , Animales , Fusión Artificial Génica , Sitios de Ligazón Microbiológica/genética , Bacteriófagos/enzimología , Línea Celular , Núcleo Celular/metabolismo , Distrofina/análisis , Electroporación/instrumentación , Terapia Genética/métodos , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Humanos , Integrasas/metabolismo , Ratones , Distrofia Muscular de Duchenne/terapia , Mioblastos/química , Plásmidos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA