Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Gene Med ; 26(7): e3716, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38961849

RESUMEN

BACKGROUND: Differentiation of pluripotent stem cells into desired lineages is the key aspect of regenerative medicine and cell-based therapy. Although RNA interference (RNAi) technology is exploited extensively for this, methods for long term silencing of the target genes leading to differentiation remain a challenge. Sustained knockdown of the target gene by RNAi is often inefficient as a result of low delivery efficiencies, protocol induced toxicity and safety concerns related to viral vectors. Earlier, we established octa-arginine functionalized hydroxyapatite nano vehicles (R8HNPs) for delivery of small interfering RNA (siRNA) against a pluripotency marker gene in mouse embryonic stem cells. Although we demonstrated excellent knockdown efficiency of the target gene, sustained gene silencing leading to differentiation was yet to be achieved. METHODS: To establish a sustained non-viral gene silencing protocol using R8HNP, we investigated various methods of siRNA delivery: double delivery of adherent cells (Adh-D), suspension delivery followed by adherent delivery (Susp + Adh), single delivery in suspension (Susp-S) and multiple deliveries in suspension (Susp-R). Sustained knockdown of a pluripotent marker gene followed by differentiation was analysed by reverse transcriptase-PCR, fluoresence-activated cell sorting and immunofluorescence techniques. Impact on cell viability as a result of repeated exposure of the R8HNP was also tested. RESULTS: Amongst the protocols tested, the most efficient knockdown of the target gene for a prolonged period of time was obtained by repeated suspension delivery of the R8HNP-siRNA conjugate. The long-term silencing of a pluripotency marker gene resulted in differentiation of R1 ESCs predominantly towards the extra embryonic and ectodermal lineages. Cells displayed excellent tolerance to repeated exposures of R8HNPs. CONCLUSIONS: The results demonstrate that R8HNPs are promising, biocompatible, non-viral alternatives for prolonged gene silencing and obtaining differentiated cells for therapeutics.


Asunto(s)
Diferenciación Celular , Durapatita , Células Madre Embrionarias de Ratones , ARN Interferente Pequeño , Animales , Ratones , Durapatita/química , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/efectos de los fármacos , ARN Interferente Pequeño/genética , Silenciador del Gen , Materiales Biocompatibles/química , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Nanopartículas/química , Transducción Genética , Interferencia de ARN , Técnicas de Silenciamiento del Gen
2.
Biochem Biophys Res Commun ; 627: 21-29, 2022 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-36029534

RESUMEN

Deciphering the endocytosis mechanisms of nanoparticle entry in cells is crucial to understand the fate of nanoparticles and the biological activity of the transported cargo. Such studies require the use of reporter agents such as fluorescent markers. Previously, we have reported the synthesis of self-fluorescent HAp nanoparticles as efficient nucleic acid delivery agents in prokaryotic and eukaryotic cells. Here, we show the application of biocompatible self-fluorescent nano delivery vehicle based on HAp and CPP- octa-arginine as an efficient system to study the mechanisms of intracellular fate of a gene delivery agent. The pathway of octa-arginine functionalized HAp NP (R8HNP) and HAp NP uptake in R1 ESCs was elucidated using confocal microscopy with the help of endocytic inhibitors. The NPs mainly enter R1 ESCs by clathrin mediated and macropinocytosis pathways. It was established that the NPs escape endosomal degradation by proton sponge effect owing to their ability to buffer the pH and trigger osmotic rupture. The functionalization of CPP, effectively enhanced the internalization and endosomal escape in R1 ESCs. The detailed results of these studies are outlined in this manuscript.


Asunto(s)
Durapatita , Nanopartículas , Arginina/farmacología , Clatrina/metabolismo , Durapatita/farmacología , Endocitosis , Nanopartículas/química
3.
BBA Adv ; 2: 100037, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37082594

RESUMEN

Metabolic syndrome (MetS) is associated with a group of conditions, which enhances the risk of diabetes, heart diseases and stroke in the affected individuals. Earlier reports from our lab have shown that Tumor necrosis factor-α (TNF-α) significantly modulates the expression of 56 genes at the alternative splicing level which are involved in various signaling and metabolic pathways (MetS genes) connected to MetS. These MetS genes were predicted to interact with various RNA-binding proteins (RBPs) when exposed to TNF-α, resulting changes in their alternative splicing patterns. Here we are presenting data of an RNA-Seq analysis, which identified 1218 unique, and significantly regulated genes by TNF-α, 15% of which are RBPs . Among the 1218 genes, 204 genes have been identified as MetS genes by the ingenuity pathway analysis, and 10% of the MetS genes are found as RBPs. Our results also show that TNF-α changes the phosphorylation status of certain RBPs such as SR proteins, crucial players in alternative splicing, possibly via changing the activation status of certain upstream signaling molecules which also act as upstream kinases for these proteins. Taken together, these findings suggest that TNF-α influences the regulation of the RBPs at the various levels for their expression, which may lead to the alteration of the splicing pattern of the MetS genes. MetS genes acting as RBPs and are modulated by TNF-α, predict the existence of highly interconnected mechanisms which require further analysis to understand their dual roles on the onset of these diseases.

4.
Mol Pharm ; 18(3): 796-806, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33464088

RESUMEN

The small interference RNA (siRNA)-assisted RNA interference approach in stem cells for differentiating into cell-specific lineages is gaining importance for its therapeutic potential. An effective gene delivery platform is crucial to achieve this goal. In this context, self-fluorescent, cell-penetrating peptide (CPP)-functionalized hydroxyapatite nanoparticles (R8HNPs) were synthesized by a modified sol gel technique. R8HNPs were crystalline, displayed characteristic bands, and exhibited broad emission spectra from 350 to 750 nm corresponding to green and red fluorescence. The biocompatible R8HNPs displayed robust binding with siRNA and excellent uptake in R1 ESCs. This was attributed to functionalization with CPP. Moreover, the R8HNP-complexed siRNA exhibited excellent serum and room temperature stability. The NPs protected the siRNA from sonication, pH, and temperature-induced stress and efficiently delivered siRNA to trigger 80% silencing of a pluripotency marker gene, Oct4, in R1 ESCs at 48 h. The transient downregulation was also observed at the protein level. Our findings demonstrate R8HNPs as a promising delivery agent for siRNA therapeutics with the potential for lineage-specific differentiation and future applications in regenerative medicine.


Asunto(s)
Durapatita/química , Células Madre Embrionarias de Ratones/efectos de los fármacos , Nanopartículas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Péptidos de Penetración Celular/administración & dosificación , Péptidos de Penetración Celular/química , Regulación hacia Abajo/efectos de los fármacos , Técnicas de Transferencia de Gen , Ratones , Nanopartículas/química , Interferencia de ARN/efectos de los fármacos , ARN Interferente Pequeño/química
5.
FEBS Lett ; 595(4): 476-490, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33417721

RESUMEN

In this report, using the database of RNA-binding protein specificities (RBPDB) and our previously published RNA-seq data, we analyzed the interactions between RNA and RNA-binding proteins to decipher the role of alternative splicing in metabolic disorders induced by TNF-α. We identified 13 395 unique RNA-RBP interactions, including 385 unique RNA motifs and 35 RBPs, some of which (including MBNL-1 and 3, ZFP36, ZRANB2, and SNRPA) are transcriptionally regulated by TNF-α. In addition to some previously reported RBPs, such as RBMX and HuR/ELAVL1, we found a few novel RBPs, such as ZRANB2 and SNRPA, to be involved in the regulation of metabolic syndrome-associated genes that contain an enrichment of tetrameric RNA sequences (AUUU). Taken together, this study paves the way for novel RNA-protein interaction-based therapeutics for treating metabolic syndromes.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Síndrome Metabólico/genética , Proteínas de Unión al ARN/genética , Ribonucleoproteína Nuclear Pequeña U1/genética , Factor de Necrosis Tumoral alfa/farmacología , Secuencia de Bases , Biología Computacional/métodos , Proteína 2 Similar a ELAV/genética , Proteína 2 Similar a ELAV/metabolismo , Estudio de Asociación del Genoma Completo , Células HEK293 , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Humanos , Síndrome Metabólico/metabolismo , Síndrome Metabólico/patología , Unión Proteica , ARN/genética , ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Tristetraprolina/genética , Tristetraprolina/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
6.
Cytokine ; 125: 154815, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31476685

RESUMEN

BACKGROUND: TNF-α, a pro-inflammatory cytokine is one of the major contributors for metabolic syndromes including insulin resistance, obesity, type II diabetes etc. The role of alternative splicing, a post-transcriptional regulation of gene expression on the onset of these syndromes is poorly understood. However, the role of alternative splicing, which more than 95% of all exons in eukaryotic cells undergo in several other diseases including cancer and muscle dystrophy, has been elucidated. In this study we aim to investigate the role of alternative splicing in pathways leading to metabolic syndromes mediated by TNF-α. METHODS: A genome wide transcriptome analysis was carried out using Illumina platform. Results were validated using RT-PCR analysis. Various bioinformatics tools and databases (for example IPA, KEGG, STRING etc) were used for the pathway and interactome analysis. CURRENT FINDINGS: Transcriptome wide analysis revealed that TNF-α treatment in vitro causes a significant change in expression of 228 genes at the level of alternative splicing. Regulation of some of these genes was validated in different cell lines. Pathway analysis showed at least 15% of the alternatively spliced genes fall under the contributory pathways leading to different metabolic syndromes, among which the maximally interconnected genes were transcription regulators. CONCLUSION: These findings suggest that TNF-α.-mediated alternative splicing plays a crucial role in regulating various genes involved in pathways connected to metabolic syndromes.


Asunto(s)
Empalme Alternativo/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Síndrome Metabólico/metabolismo , Transcriptoma/genética , Factor de Necrosis Tumoral alfa/farmacología , Animales , Línea Celular , Biología Computacional , Bases de Datos Genéticas , Exones , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/genética , Síndrome Metabólico/genética , Ratones , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Curr Stem Cell Res Ther ; 15(2): 155-172, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31789134

RESUMEN

Stem cell-based regenerative medicine holds exceptional therapeutic potential and hence the development of efficient techniques to enhance control over the rate of differentiation has been the focus of active research. One of the strategies to achieve this involves delivering siRNA into stem cells and exploiting the RNA interference (RNAi) mechanism. Transport of siRNA across the cell membrane is a challenge due to its anionic property, especially in primary human cells and stem cells. Moreover, naked siRNA incites immune responses, may cause off-target effects, exhibits low stability and is easily degraded by endonucleases in the bloodstream. Although siRNA delivery using viral vectors and electroporation has been used in stem cells, these methods demonstrate low transfection efficiency, cytotoxicity, immunogenicity, events of integration and may involve laborious customization. With the advent of nanotechnology, nanocarriers which act as novel gene delivery vehicles designed to overcome the problems associated with safety and practicality are being developed. The various nanomaterials that are currently being explored and discussed in this review include liposomes, carbon nanotubes, quantum dots, protein and peptide nanocarriers, magnetic nanoparticles, polymeric nanoparticles, etc. These nanodelivery agents exhibit advantages such as low immunogenic response, biocompatibility, design flexibility allowing for surface modification and functionalization, and control over the surface topography for achieving the desired rate of siRNA delivery and improved gene knockdown efficiency. This review also includes discussion on siRNA co-delivery with imaging agents, plasmid DNA, drugs etc. to achieve combined diagnostic and enhanced therapeutic functionality, both for in vitro and in vivo applications.


Asunto(s)
Diferenciación Celular/genética , Técnicas de Transferencia de Gen , Nanopartículas/uso terapéutico , ARN Interferente Pequeño/administración & dosificación , Células Madre/fisiología , Animales , Sistemas de Liberación de Medicamentos , Terapia Genética/instrumentación , Terapia Genética/métodos , Humanos , Nanopartículas/química , Nanotecnología/métodos , Interferencia de ARN/fisiología , Transfección/instrumentación , Transfección/métodos
8.
J Biosci ; 44(6)2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31894131

RESUMEN

Type II diabetes mellitus (T2DM) and obesity are two common pathophysiological conditions of metabolic syndrome (MetS), a collection of similar metabolic dysfunctions due to sedentary lifestyle and overnutrition. Obesity arises from improper adipogenesis which otherwise has a crucial role in maintaining proper metabolic functions. Downstream events arising from obesity have been linked to T2DM. The nuclear receptor peroxisome proliferator activator gamma (PPAR-γ), responsible for maintaining lipid and glucose homeostasis, is down-regulated under obesity leading to a weakened insulin sensitivity of the human body. In course of our review we will outline details of the down-regulation mechanism, provide an overview of the current clinical therapeutics and their shortcomings. Toxicity studies on the seminal drug troglitazone, belonging to the most effective glitazone anti-diabetic category, is also discussed. This will lead to an overview about structural adaptations on the existing glitazones to alleviate their side effects and toxicity. Finally, we forward a concept of novel therapeutics mimicking the glitazone framework, based on some design concepts and preliminary in silico studies. These could be later developed into dual acting drugs towards alleviating the deleterious effects of obesity on normal glucose metabolism, and address obesity in itself.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Glucosa/metabolismo , Síndrome Metabólico/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Adipogénesis/efectos de los fármacos , Diabetes Mellitus Tipo 2/complicaciones , Humanos , Hipoglucemiantes/uso terapéutico , Resistencia a la Insulina/genética , Síndrome Metabólico/complicaciones , Síndrome Metabólico/genética , Obesidad/complicaciones , Obesidad/genética , PPAR gamma/genética , Tiazolidinedionas/uso terapéutico , Troglitazona/uso terapéutico
9.
Sci Rep ; 8(1): 17989, 2018 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-30559394

RESUMEN

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

10.
Sci Rep ; 7(1): 7555, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28790387

RESUMEN

Striatin and SG2NA are essential constituents of the multi-protein STRIPAK assembly harbouring protein phosphatase PP2A and several kinases. SG2NA has several isoforms generated by mRNA splicing and editing. While the expression of striatin is largely restricted to the striatum in brain, that of SG2NAs is ubiquitous. In NIH3T3 cells, only the 78 kDa isoform is expressed. When cells enter into the S phase, the level of SG2NA increases; reaches maximum at the G2/M phase and declines thereafter. Downregulation of SG2NA extends G1 phase and its overexpression extends G2. Ectopic expression of the 35 kDa has no effects on the cell cycle. Relative abundance of phospho-SG2NA is high in the microsome and cytosol and the nucleus but low in the mitochondria. Okadoic acid, an inhibitor of PP2A, increases the level of SG2NA which is further enhanced upon inhibition of proteasomal activity. Phospho-SG2NA is thus more stable than the dephosphorylated form. Inhibition of GSK3ß by LiCl reduces its level, but the inhibition of ERK by PD98059 increases it. Thus, ERK decreases the level of phospho-SG2NA by inhibiting GSK3ß. In cells depleted from SG2NA by shRNA, the levels of pGSK3ß and pERK are reduced, suggesting that these kinases and SG2NA regulate each other's expression.


Asunto(s)
Autoantígenos/genética , Proteínas de Unión a Calmodulina/genética , Ciclo Celular/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Glucógeno Sintasa Quinasa 3 beta/genética , Animales , Autoantígenos/química , Autoantígenos/metabolismo , Proteínas de Unión a Calmodulina/química , Proteínas de Unión a Calmodulina/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HEK293 , Humanos , Ratones , Peso Molecular , Células 3T3 NIH , Fosforilación , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN
11.
Cell Stem Cell ; 14(4): 523-34, 2014 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-24702998

RESUMEN

The creation of induced pluripotent stem cells (iPSCs) from somatic cells by ectopic expression of transcription factors has galvanized the fields of regenerative medicine and developmental biology. Here, we report a kinome-wide RNAi-based analysis to identify kinases that regulate somatic cell reprogramming to iPSCs. We prepared 3,686 small hairpin RNA (shRNA) lentiviruses targeting 734 kinase genes covering the entire mouse kinome and individually examined their effects on iPSC generation. We identified 59 kinases as barriers to iPSC generation and characterized seven of them further. We found that shRNA-mediated knockdown of the serine/threonine kinases TESK1 or LIMK2 promoted mesenchymal-to-epithelial transition, decreased COFILIN phosphorylation, and disrupted Actin filament structures during reprogramming of mouse embryonic fibroblasts. Similarly, knockdown of TESK1 in human fibroblasts also promoted reprogramming to iPSCs. Our study reveals the breadth of kinase networks regulating pluripotency and identifies a role for cytoskeletal remodeling in modulating the somatic cell reprogramming process.


Asunto(s)
Diferenciación Celular , Reprogramación Celular/genética , Citoesqueleto/metabolismo , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Proteínas Serina-Treonina Quinasas/genética , Factores Despolimerizantes de la Actina/genética , Factores Despolimerizantes de la Actina/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Redes Reguladoras de Genes , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Quinasas Lim/antagonistas & inhibidores , Quinasas Lim/genética , Quinasas Lim/metabolismo , Ratones , Microscopía Confocal , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Teratoma/metabolismo , Teratoma/patología
12.
PLoS One ; 6(11): e27869, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22132154

RESUMEN

Exon 11 of the insulin receptor gene (INSR) is alternatively spliced in a developmentally and tissue-specific manner. Linker scanning mutations in a 5' GA-rich enhancer in intron 10 identified AGGGA sequences that are important for enhancer function. Using RNA-affinity purification and mass spectrometry, we identified hnRNP F and hnRNP A1 binding to these AGGGA sites and also to similar motifs at the 3' end of the intron. The hnRNPs have opposite functional effects with hnRNP F promoting and hnRNP A1 inhibiting exon 11 inclusion, and deletion of the GA-rich elements eliminates both effects. We also observed specific binding of hnRNP A1 to the 5' splice site of intron 11. The SR protein SRSF1 (SF2/ASF) co-purified on the GA-rich enhancer and, interestingly, also competes with hnRNP A1 for binding to the splice site. A point mutation -3U→C decreases hnRNP A1 binding, increases SRSF1 binding and renders the exon constitutive. Lastly, our data point to a functional interaction between hnRNP F and SRSF1 as a mutant that eliminates SRSF1 binding to exon 11, or a SRSF1 knockdown, which prevents the stimulatory effect of hnRNP F over expression.


Asunto(s)
Empalme Alternativo/genética , Exones/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/metabolismo , Receptor de Insulina/genética , Secuencia de Bases , Sitios de Unión , Secuencia de Consenso , Técnicas de Silenciamiento del Gen , Células HEK293 , Células Hep G2 , Ribonucleoproteína Nuclear Heterogénea A1 , Humanos , Intrones/genética , Modelos Biológicos , Datos de Secuencia Molecular , Mutagénesis/genética , Mutación/genética , Proteínas Nucleares/metabolismo , Motivos de Nucleótidos/genética , Unión Proteica , Proteínas de Unión al ARN/metabolismo , Factores de Empalme Serina-Arginina
13.
J Biol Chem ; 285(33): 25426-37, 2010 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-20519504

RESUMEN

The insulin receptor exists as two isoforms, IR-A and IR-B, which result from alternative splicing of exon 11 in the primary transcript. These two isoforms show a cell-specific distribution, and their relative proportions also vary during development, aging, and in different disease states. We have previously demonstrated that both intron 10 and the alternatively spliced exon 11 contain regulatory sequences that affect insulin receptor splicing both positively and negatively and that these sequences bind the serine/arginine-rich (SR) proteins SRp20 and SF2/ASF and the CELF protein CUG-BP1. In this study, we describe a new intronic splicing element within intron 11 that is highly conserved across species. Using minigenes carrying deletion mutations within intron 11, we demonstrated that this sequence functions as an intronic splicing enhancer. We subsequently used RNA affinity chromatography to identify Mbnl1 as a splicing factor that recognizes this enhancer. By ribonucleoprotein immunoprecipitation, we also established that Mbnl1 binds specifically to the INSR (insulin receptor gene) RNA. Overexpression or knockdown of Mbnl1 in hepatoma and embryonic kidney cells altered the levels of exon 11 inclusion. Finally, we showed that deletion of the intronic enhancer eliminates the ability of Mbnl1 to promote exon inclusion. Collectively, these findings demonstrate a role for Mbnl1 in controlling insulin receptor exon 11 inclusion via binding to a downstream intronic enhancer element.


Asunto(s)
Exones/genética , Intrones/genética , Proteínas de Unión al ARN/metabolismo , Receptor de Insulina/genética , Animales , Western Blotting , Línea Celular , Elementos de Facilitación Genéticos , Evolución Molecular , Células Hep G2 , Humanos , Unión Proteica/genética , Unión Proteica/fisiología , Empalme del ARN/genética , Empalme del ARN/fisiología , ARN Interferente Pequeño , Proteínas de Unión al ARN/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Biochem Biophys Res Commun ; 388(1): 117-21, 2009 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-19646964

RESUMEN

Both polyunsaturated fatty acids and AMPK promote energy partitioning away from energy consuming processes, such as fatty acid synthesis, towards energy generating processes, such as beta-oxidation. In this report, we demonstrate that arachidonic acid activates AMPK in primary rat hepatocytes, and that this effect is p38 MAPK-dependent. Activation of AMPK mimics the inhibition by arachidonic acid of the insulin-mediated induction of G6PD. Similar to intracellular signaling by arachidonic acid, AMPK decreases insulin signal transduction, increasing Ser(307) phosphorylation of IRS-1 and a subsequent decrease in AKT phosphorylation. Overexpression of dominant-negative AMPK abolishes the effect of arachidonic acid on G6PD expression. These data suggest a role for AMPK in the inhibition of G6PD by polyunsaturated fatty acids.


Asunto(s)
Ácidos Grasos Insaturados/biosíntesis , Glucosafosfato Deshidrogenasa/antagonistas & inhibidores , Hepatocitos/enzimología , Proteínas Quinasas/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Ácido Araquidónico/biosíntesis , Ácido Araquidónico/farmacología , Ácidos Grasos Insaturados/farmacología , Glucosafosfato Deshidrogenasa/biosíntesis , Hepatocitos/efectos de los fármacos , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Masculino , Fosforilación , Ratas , Ratas Sprague-Dawley , Serina/metabolismo , Transducción de Señal
15.
Mol Cell Biol ; 29(3): 871-80, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19047369

RESUMEN

The insulin receptor (IR) exists as two isoforms, IR-A and IR-B, which result from alternative splicing of exon 11 in the primary transcript. This alternative splicing is cell specific, and the relative proportions of exon 11 isoforms also vary during development, aging, and different disease states. We have previously demonstrated that both intron 10 and exon 11 contain regulatory sequences that affect IR splicing both positively and negatively. In this study, we sought to define the precise sequence elements within exon 11 that control exon recognition and cellular factors that recognize these elements. Using minigenes carrying linker-scanning mutations within exon 11, we detected both exonic splicing enhancer and exonic splicing silencer elements. We identified binding of SRp20 and SF2/ASF to the exonic enhancers and CUG-BP1 to the exonic silencer by RNA affinity chromatography. Overexpression and knockdown studies with hepatoma and embryonic kidney cells demonstrated that SRp20 and SF2/ASF increase exon inclusion but that CUG-BP1 causes exon skipping. We found that CUG-BP1 also binds to an additional intronic splicing silencer, located at the 3' end of intron 10, to promote exon 11 skipping. Thus, we propose that SRp20, SF2/ASF, and CUG-BP1 act antagonistically to regulate IR alternative splicing in vivo and that the relative ratios of SRp20 and SF2/ASF to CUG-BP1 in different cells determine the degree of exon inclusion.


Asunto(s)
Empalme Alternativo/genética , Exones/genética , Proteínas de Unión al ARN/metabolismo , Receptor de Insulina/genética , Secuencia de Bases , Proteínas CELF1 , Línea Celular , Elementos de Facilitación Genéticos/genética , Humanos , Modelos Genéticos , Datos de Secuencia Molecular , Mutagénesis , Conformación de Ácido Nucleico , Unión Proteica , Proteínas Represoras/metabolismo , Factores de Empalme Serina-Arginina , Elementos Silenciadores Transcripcionales/genética
16.
Mol Endocrinol ; 21(5): 1175-91, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17299135

RESUMEN

The hypothalamic-pituitary-gonadal endocrine axis regulates reproduction through estrous phase-dependent release of the heterodimeric gonadotropic glycoprotein hormones, LH and FSH, from the gonadotropes of the anterior pituitary. Gonadotropin synthesis and release is dependent upon pulsatile stimulation by the hypothalamic neuropeptide GnRH. Alterations in pulse frequency and amplitude alter the relative levels of gonadotropin synthesis and release. The mechanism of interpretation of GnRH pulse frequency and amplitude by gonadotropes is not understood. We have examined gene expression in LbetaT2 gonadotropes under various pulse regimes in a cell perifusion system by microarray and identified 1127 genes activated by tonic or pulsatile GnRH. Distinct patterns of expression are associated with each pulse frequency, but the greatest changes occur at a 60-min or less interpulse interval. The immediate early gene mRNAs encoding early growth response (Egr)1 and Egr2, which activate the gonadotropin LH beta-subunit gene promoter, are stably induced at high pulse frequency. In contrast, mRNAs for the Egr corepressor genes Ngfi-A binding protein Nab1 and Nab2 are stably induced at low pulse frequency. We show that Ngfi-A binding protein members inhibit Egr-mediated frequency-dependent induction of the LH beta-subunit promoter. This pattern of expression suggests a model of pulse frequency detection that acts by suppressing activation by Egr family members at low frequency and allowing activation at sustained high-frequency pulses.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Hormona Luteinizante de Subunidad beta/genética , Regiones Promotoras Genéticas , Proteínas Represoras/genética , Animales , División Celular , Células Cultivadas , Retroalimentación , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Cinética , Células L , Ratones , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
17.
J Biol Chem ; 281(45): 34146-58, 2006 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-16980303

RESUMEN

The inhibition of glucose-6-phosphate dehydrogenase (G6PD) expression by arachidonic acid occurs by changes in the rate of pre-mRNA splicing. Here, we have identified a cis-acting RNA element required for regulated splicing of G6PD mRNA. Using transfection of G6PD RNA reporter constructs into rat hepatocytes, the cis-acting RNA element involved in this regulation was localized to nucleotides 43-72 of exon 12 in the G6PD mRNA. In in vitro splicing assays, RNA substrates containing exon 12 were not spliced. In contrast, RNA substrates containing other regions (exons 8 and 9 or exons 10 and 11) of the G6PD mRNA were efficiently spliced. Furthermore, exon 12 can inhibit splicing when substituted for other exons in RNA substrates that are readily spliced. This activity of the exon 12 regulatory element suggests that it is an exonic splicing silencer. Consistent with its activity as a splicing silencer, spliceosome assembly was inhibited on RNA substrates containing exon 12 compared with RNAs representing other regions of the G6PD transcript. Elimination of nucleotides 43-72 of exon 12 did not restore splicing of exon 12-containing RNA; thus, the 30-nucleotide element may not be exclusively a silencer. The binding of heterogeneous nuclear ribonucleoproteins K, L, and A2/B1 from both HeLa and hepatocyte nuclear extracts to the element further supports its activity as a silencer. In addition, SR proteins bind to the element, consistent with the presence of enhancer activity within this sequence. Thus, an exonic splicing silencer is involved in the inhibition of splicing of a constitutively spliced exon in the G6PD mRNA.


Asunto(s)
Exones/genética , Regulación de la Expresión Génica , Glucosafosfato Deshidrogenasa/genética , Empalme del ARN , ARN Mensajero/genética , Elementos Silenciadores Transcripcionales , Animales , Sitios de Unión , Western Blotting , Células Cultivadas , Cromatografía de Afinidad , Cromatografía Liquida , Glucosafosfato Deshidrogenasa/metabolismo , Células HeLa , Hepatocitos/citología , Hepatocitos/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Intrones/genética , Masculino , Espectrometría de Masas , Plásmidos/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ribonucleasas/metabolismo , Ribonucleoproteínas/metabolismo , Empalmosomas/metabolismo , Transcripción Genética
18.
J Biol Chem ; 280(49): 40660-7, 2005 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-16210322

RESUMEN

Polyunsaturated fatty acids are potent inhibitors of lipogenic gene expression in liver. The lipogenic enzyme glucose-6-phosphate dehydrogenase (G6PD) is unique in this gene family, in that fatty acids inhibit at a post-transcriptional step. In this study, we have provided evidence for a signaling pathway for the arachidonic acid inhibition of G6PD mRNA abundance. Arachidonic acid decreases the insulin induction of G6PD expression; by itself, arachidonic acid does not inhibit basal G6PD mRNA accumulation. The insulin stimulation of G6PD involves the phosphoinositide 3-kinase (PI 3-kinase) pathway (Wagle, A., Jivraj, S., Garlock, G. L., and Stapleton, S. R. (1998) J. Biol. Chem. 273, 14968-14974). Incubation of hepatocytes with arachidonic acid blocks the activation of PI 3-kinase by insulin as observed by a decrease in Ser(473) phosphorylation of Akt, the downstream effector of PI 3-kinase. The decrease in PI 3-kinase activity was associated with an increase in Ser(307) phosphorylation of IRS-1. Western analysis demonstrated increased phosphorylation of p38 mitogen-activated protein kinase (MAPK) in arachidonic acid-treated cells, whereas extracellular signal-regulated kinase and c-Jun NH(2)-terminal kinase activity was not changed. Incubating the hepatocytes with the p38 MAPK inhibitor, SB203580, blocked the arachidonic acid inhibition of G6PD mRNA accumulation. Furthermore, SB203580 decreased the arachidonic acid-mediated Ser(307) phosphorylation of IRS-1 and rescued Akt activation that was otherwise decreased by arachidonic acid. Thus, arachidonic acid inhibits the insulin stimulation of G6PD mRNA accumulation by stimulating the p38 MAPK pathway, thereby inhibiting insulin signal transduction.


Asunto(s)
Ácido Araquidónico/farmacología , Glucosafosfato Deshidrogenasa/biosíntesis , Insulina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Ácidos Grasos Insaturados/farmacología , Expresión Génica/efectos de los fármacos , Glucosafosfato Deshidrogenasa/antagonistas & inhibidores , Glucosafosfato Deshidrogenasa/genética , Hepatocitos/enzimología , Proteínas Sustrato del Receptor de Insulina , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Serina/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA