Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
ACS Chem Biol ; 17(2): 348-360, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35034446

RESUMEN

A major obstacle in the development of effective oligonucleotide therapeutics is a lack of understanding about their cytosolic and nuclear penetration. To address this problem, we have applied the chloroalkane penetration assay (CAPA) to oligonucleotide therapeutics. CAPA was used to quantitate cytosolic delivery of antisense oligonucleotides (ASOs) and siRNAs and to explore the effects of a wide variety of commonly used chemical modifications and their patterning. We evaluated potential artifacts by exploring the effects of serum, comparing activity data and CAPA data, and assessing the impact of the chloroalkane tag and its linker chemistry. We also used viral transduction to expand CAPA to the nuclear compartment in epithelial and neuronal cell lines. Using this enhanced method, we measured a 48-h time course of nuclear penetration for a panel of chemically diverse modified RNAs. Moving forward, CAPA will be a useful tool for deconvoluting the complex processes of endosomal uptake, escape into the cytosol, and subcellular trafficking of oligonucleotide therapeutics in therapeutically relevant cell types.


Asunto(s)
Oligonucleótidos Antisentido , Oligonucleótidos , Núcleo Celular , Citosol/metabolismo , Oligonucleótidos/metabolismo , Oligonucleótidos Antisentido/metabolismo , ARN Interferente Pequeño/metabolismo
2.
J Biol Chem ; 297(5): 101257, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34597669

RESUMEN

Healing of cutaneous wounds requires the collective migration of epithelial keratinocytes to seal the wound bed from the environment. However, the signaling events that coordinate this collective migration are unclear. In this report, we address the role of phosphorylation of eukaryotic initiation factor 2 (eIF2) and attendant gene expression during wound healing. Wounding of human keratinocyte monolayers in vitro led to the rapid activation of the eIF2 kinase GCN2. We determined that deletion or pharmacological inhibition of GCN2 significantly delayed collective cell migration and wound closure. Global transcriptomic, biochemical, and cellular analyses indicated that GCN2 is necessary for maintenance of intracellular free amino acids, particularly cysteine, as well as coordination of RAC1-GTP-driven reactive oxygen species (ROS) generation, lamellipodia formation, and focal adhesion dynamics following keratinocyte wounding. In vivo experiments using mice deficient for GCN2 validated the role of the eIF2 kinase during wound healing in intact skin. These results indicate that GCN2 is critical for appropriate induction of collective cell migration and plays a critical role in coordinating the re-epithelialization of cutaneous wounds.


Asunto(s)
Movimiento Celular , Queratinocitos/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Cicatrización de Heridas , Aminoácidos/metabolismo , Animales , Línea Celular Transformada , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Humanos , Queratinocitos/patología , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Seudópodos/genética , Seudópodos/metabolismo , Piel/enzimología , Piel/lesiones , Piel/patología
3.
Plant Biotechnol J ; 11(9): 1126-34, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23953646

RESUMEN

Modern agriculture demands crops carrying multiple traits. The current paradigm of randomly integrating and sorting independently segregating transgenes creates severe downstream breeding challenges. A versatile, generally applicable solution is hereby provided: the combination of high-efficiency targeted genome editing driven by engineered zinc finger nucleases (ZFNs) with modular 'trait landing pads' (TLPs) that allow 'mix-and-match', on-demand transgene integration and trait stacking in crop plants. We illustrate the utility of nuclease-driven TLP technology by applying it to the stacking of herbicide resistance traits. We first integrated into the maize genome an herbicide resistance gene, pat, flanked with a TLP (ZFN target sites and sequences homologous to incoming DNA) using WHISKERS™-mediated transformation of embryogenic suspension cultures. We established a method for targeted transgene integration based on microparticle bombardment of immature embryos and used it to deliver a second trait precisely into the TLP via cotransformation with a donor DNA containing a second herbicide resistance gene, aad1, flanked by sequences homologous to the integrated TLP along with a corresponding ZFN expression construct. Remarkably, up to 5% of the embryo-derived transgenic events integrated the aad1 transgene precisely at the TLP, that is, directly adjacent to the pat transgene. Importantly and consistent with the juxtaposition achieved via nuclease-driven TLP technology, both herbicide resistance traits cosegregated in subsequent generations, thereby demonstrating linkage of the two independently transformed transgenes. Because ZFN-mediated targeted transgene integration is becoming applicable across an increasing number of crop species, this work exemplifies a simple, facile and rapid approach to trait stacking.


Asunto(s)
Endonucleasas/genética , Marcación de Gen/métodos , Genoma de Planta/genética , Resistencia a los Herbicidas , Herbicidas/farmacología , Zea mays/genética , Productos Agrícolas , Endonucleasas/metabolismo , Ligamiento Genético , Fenotipo , Hojas de la Planta/genética , Hojas de la Planta/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Plantas Modificadas Genéticamente , Transgenes , Dedos de Zinc
4.
J Biol Chem ; 288(9): 6386-96, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23322769

RESUMEN

Control of plasma cholesterol levels is a major therapeutic strategy for management of coronary artery disease (CAD). Although reducing LDL cholesterol (LDL-c) levels decreases morbidity and mortality, this therapeutic intervention only translates into a 25-40% reduction in cardiovascular events. Epidemiological studies have shown that a high LDL-c level is not the only risk factor for CAD; low HDL cholesterol (HDL-c) is an independent risk factor for CAD. Apolipoprotein A-I (ApoA-I) is the major protein component of HDL-c that mediates reverse cholesterol transport from tissues to the liver for excretion. Therefore, increasing ApoA-I levels is an attractive strategy for HDL-c elevation. Using genome-wide siRNA screening, targets that regulate hepatocyte ApoA-I secretion were identified through transfection of 21,789 siRNAs into hepatocytes whereby cell supernatants were assayed for ApoA-I. Approximately 800 genes were identified and triaged using a convergence of information, including genetic associations with HDL-c levels, tissue-specific gene expression, druggability assessments, and pathway analysis. Fifty-nine genes were selected for reconfirmation; 40 genes were confirmed. Here we describe the siRNA screening strategy, assay implementation and validation, data triaging, and example genes of interest. The genes of interest include known and novel genes encoding secreted enzymes, proteases, G-protein-coupled receptors, metabolic enzymes, ion transporters, and proteins of unknown function. Repression of farnesyltransferase (FNTA) by siRNA and the enzyme inhibitor manumycin A caused elevation of ApoA-I secretion from hepatocytes and from transgenic mice expressing hApoA-I and cholesterol ester transfer protein transgenes. In total, this work underscores the power of functional genetic assessment to identify new therapeutic targets.


Asunto(s)
Apolipoproteína A-I/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , Animales , Apolipoproteína A-I/genética , HDL-Colesterol/genética , HDL-Colesterol/metabolismo , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa/antagonistas & inhibidores , Farnesiltransferasa/genética , Farnesiltransferasa/metabolismo , Estudio de Asociación del Genoma Completo , Células Hep G2 , Humanos , Hígado/citología , Ratones , Ratones Transgénicos , Polienos/farmacología , Alcamidas Poliinsaturadas/farmacología , ARN Interferente Pequeño/genética
5.
Hybridoma (Larchmt) ; 26(3): 155-61, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17600497

RESUMEN

Folypolyglutamate synthetase (FPGS) plays a critical role in the cellular retention of both folates and antifolates. Resistance to antifolates is in part related to changes in FPGS enzyme activity and levels of messenger RNA, or in some instances, protein as evaluated by Western blots using polyclonal antisera. The present study was designed to derive a series of monoclonal antibodies (MAb) against the native protein, to characterize them in terms of specificity and epitope mapping, and to determine kinetic constants by Biacore. We report on 3 IgG(1) kappa MAbs-namely, 4-2, 4-3, and 4-18-with epitopes localized to the carboxyl domain of the protein. These antibodies recognize a single band on Western blots of HeLa cell lysates, which is significantly reduced following RNAi knockdown. The recognition of both the native and denatured conformations of FPGS by these MAbs should provide useful reagents for FPGS quantitation in either tumor cell lysates or in tumor biopsies.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Péptido Sintasas/inmunología , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos , Mapeo Epitopo , Células HeLa , Humanos , Hibridomas/inmunología , Inmunoglobulina G/biosíntesis , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Neoplasias/enzimología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Péptido Sintasas/antagonistas & inhibidores , Péptido Sintasas/química , Péptido Sintasas/genética , Conformación Proteica , Desnaturalización Proteica , Interferencia de ARN , ARN Interferente Pequeño/genética , Resonancia por Plasmón de Superficie
6.
Mol Pharmacol ; 68(5): 1225-38, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16079270

RESUMEN

The pharmacological preservation of bone in the ovariectomized rat by estrogen, selective estrogen receptor modulators (SERMs), and bisphosphonates has been well described. However, comprehensive molecular analysis of the effects of these pharmacologically diverse antiresorptive agents on gene expression in bone has not been performed. This study used DNA microarrays to analyze RNA from the proximal femur metaphysis of sham and ovariectomized vehicle-treated rats, and ovariectomized rats treated for 35 days with maximally efficacious doses of 17-alpha ethinyl estradiol, the benzothiophene SERM, raloxifene, the benzopyran SERM, (S)-3-(4-hydroxyphenyl)-4-methyl-2-[4-[2-(1-piperidinyl)ethoxy]phenyl]-2H-1-benzopyran-7-ol (EM652), and the aminobisphosphonate, alendronate. Ovariectomy resulted in 644 significant probe set changes relative to sham control rats (p < 0.05), whereas E2, raloxifene, EM652, and alendronate regulated 613, 765, 652, and 737 probe sets, respectively, relative to ovariectomized control rats. An intersection of these data sets yielded 334 unique genes that were altered after ovariectomy and additionally changed by one or more antiresorptive treatment. Clustering analysis showed that the transcript profile was distinctly different for each pharmaceutical agent and that raloxifene maintained more genes at sham levels than any other treatment. In addition, E2 and alendronate suppressed a cluster of genes associated with bone formation activity below that of sham, whereas raloxifene had little effect on these genes. These data indicate stronger suppressive effects of E2 and alendronate on bone formation activity and that ovariectomy plus raloxifene resembles sham more closely than ovariectomized animals treated with E2, EM652, or alendronate.


Asunto(s)
Alendronato/farmacología , Estrógenos/farmacología , Fémur/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Clorhidrato de Raloxifeno/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Biología Computacional , Femenino , Perfilación de la Expresión Génica , Osteogénesis/genética , Ovariectomía , Hormona Paratiroidea/farmacología , Ratas , Ratas Sprague-Dawley
7.
J Cell Biochem ; 95(2): 403-18, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15779007

RESUMEN

Teriparatide, human PTH (1-34), a new therapy for osteoporosis, elicits markedly different skeletal responses depending on the treatment regimen. In order to understand potential mechanisms for this dichotomy, the present investigation utilized microarrays to delineate the genes and pathways that are regulated by intermittent (subcutaneous injection of 80 microg/kg/day) and continuous (subcutaneous infusion of 40 microg/kg/day by osmotic mini pump) PTH (1-34) for 1 week in 6-month-old female rats. The effect of each PTH regimen was confirmed by histomorphometric analysis of the proximal tibial metaphysis, and mRNA from the distal femoral metaphysis was analyzed using an Affymetrix microarray. Both PTH paradigms co-regulated 22 genes including known bone formation genes (i.e., collagens, osteocalcin, decorin, and osteonectin) and also uniquely modulated additional genes. Intermittent PTH regulated 19 additional genes while continuous treatment regulated 173 additional genes. This investigation details for the first time the broad profiling of the gene and pathway changes that occur in vivo following treatment of intermittent versus continuous PTH (1-34). These results extend previous observations of gene expression changes and reveal the in vivo regulation of BMP3 and multiple neuronal genes by PTH treatment.


Asunto(s)
Huesos/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Hormona Paratiroidea/farmacología , Fragmentos de Péptidos/farmacología , Animales , Femenino , Perfilación de la Expresión Génica , Neuronas/metabolismo , Ratas
8.
J Pharmacol Exp Ther ; 309(1): 369-79, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14718597

RESUMEN

Osteoprotegerin (OPG), a secreted member of the tumor necrosis factor receptor superfamily, is a potent inhibitor of osteoclast formation and bone resorption. Because OPG functions physiologically as a locally generated (paracrine) factor, we used high-throughput screening to identify small molecules that enhance the activity of the promoter of the human OPG gene. We found three structurally unrelated compounds that selectively increased OPG gene transcription, OPG mRNA levels, and OPG protein production and release by osteoblastic cells. Structural analysis of one compound, a benzamide derivative, led to the identification of four related molecules, which are also OPG inducers. The most potent of these compounds, Cmpd 5 inhibited osteoclast formation and parathyroid hormone-induced calvarial bone resorption. In vivo, Cmpd 5 completely blocked resorptive activity (serum calcium, osteoclast number) in parathyroid hormone-treated rats. Furthermore, Cmpd 5 reduced the ability of a rat breast cancer to metastasize to bone. Finally, the compound also prevented bone loss in a rat adjuvant arthritis model. These results provide proof of the concept that low molecular weight compounds can enhance OPG production in ways that can result in effective therapies.


Asunto(s)
Antiinflamatorios/uso terapéutico , Benzamidas/uso terapéutico , Resorción Ósea/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas/metabolismo , Piridinas/uso terapéutico , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Antiinflamatorios/farmacología , Benzamidas/farmacología , Diferenciación Celular , Modelos Animales de Enfermedad , Femenino , Glicoproteínas/genética , Humanos , Masculino , Ratones , Osteoclastos/efectos de los fármacos , Osteoprotegerina , Regiones Promotoras Genéticas/efectos de los fármacos , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/genética , Receptores del Factor de Necrosis Tumoral , Relación Estructura-Actividad , Células Tumorales Cultivadas
9.
J Cell Biochem ; 85(4): 837-50, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11968023

RESUMEN

Regulator of G-protein signaling-2 (RGS-2) belongs to a novel family of GTPase-activating proteins that rapidly turn-off G-protein coupled receptor signaling. RGS proteins contain a characteristic RGS domain by which they interact with the alpha-subunit of G-proteins and drive them into their inactive GDP-bound forms. Previously, we have reported that RGS-2 mRNA is rapidly and transiently increased by PTH in rat bone and in osteoblast cultures in vitro. In this study, we further explored the molecular basis for the regulation of RGS-2 by cloning and functionally characterizing the RGS-2 gene promoter. We cloned 2.3- and 2.8-kb fragments of the 5'-flanking regions of the rat and mouse RGS-2 genes, respectively, and generated a stable clone of UMR106 osteoblastic cells containing the rat RGS-2 promoter driving the beta-gal reporter gene (p2.3RGS-2-beta-gal). Treatment of the stable clone with PTH resulted in a maximal 2.2- to 3.6-fold increase in promoter activity at 8 h, reminiscent of the early response observed with endogenous RGS-2 mRNA regulation. Further, PTH (1-38), (1-31), PTHrP (1-34), and forskolin, which elevate cAMP levels, stimulated the promoter, while PTH (3-34) and (7-34), which do not readily stimulate cAMP accumulation, and PMA that directly activates protein kinase C, had no effect on promoter activity. Taken together, these results implicate the involvement of the Galpha(s)-adenylate cyclase-protein kinase A pathway in stimulating RGS-2 expression. Maintenance of a hyperphosphorylated state via the inhibition of type 2A protein phosphatases by okadaic acid, resulted in a strong dose-dependent increase in transcriptional activity of the RGS-2 promoter as well as that of the endogenous RGS-2 gene. Furthermore, overexpression of the osteoblast-specific transcription factor Runx2 also led to a stimulation of RGS-2 promoter activity. Functional analysis using RGS-2 overexpression suggests the potential negative regulatory effects of RGS-2 on PTH- and forskolin-induced cAMP production in osteoblastic cells. In summary, our data suggest that PTH treatment results in a direct transcriptional stimulation of RGS-2 that in turn may play a role in modulating the duration/intensity of PTH receptor signaling.


Asunto(s)
Proteínas de Neoplasias , Osteoblastos/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Animales , Secuencia de Bases , Sitios de Unión/genética , Línea Celular , Clonación Molecular , Subunidad alfa 1 del Factor de Unión al Sitio Principal , ADN Complementario/genética , Expresión Génica/efectos de los fármacos , Genes Reporteros , Ratones , Datos de Secuencia Molecular , Ácido Ocadaico/farmacología , Osteoblastos/efectos de los fármacos , Hormona Paratiroidea/farmacología , Fragmentos de Péptidos/farmacología , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Regiones Promotoras Genéticas/efectos de los fármacos , Ratas , Homología de Secuencia de Ácido Nucleico , Transducción de Señal , Factores de Transcripción/metabolismo , beta-Galactosidasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...