Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Int J Mol Sci ; 21(7)2020 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-32283676

RESUMEN

DDX6 helicase is an RNA-binding protein involved in different aspects of gene expression regulation. The roles played by DDX6 depend on the complexes associated with it. Here, for the first time, we characterize the protein complexes associated with DDX6 in human adipose tissue-derived stem cells (hASCs) and analyze the dynamics of this helicase under different conditions of translational activity and differentiation. The results obtained demonstrated that the DDX6 helicase is associated with proteins involved in the control of mRNA localization, translation and metabolism in hASCs. DDX6 complexes may also assemble into more complex structures, such as RNA-dependent granules, the abundance and composition of which change upon inhibited translational activity. This finding supports the supposition that DDX6 is possibly involved in the regulation of the mRNA life cycle in hASCs. Although there was no significant variation in the protein composition of these complexes during early adipogenic or osteogenic induction, there was a change in the distribution pattern of DDX6: the number of DDX6 granules per cell was reduced during adipogenesis and was enhanced during osteogenesis.


Asunto(s)
Adipogénesis , Tejido Adiposo/citología , Proteínas Portadoras/metabolismo , ARN Helicasas DEAD-box/metabolismo , Osteogénesis , Proteínas Proto-Oncogénicas/metabolismo , Células Madre/citología , Células Madre/metabolismo , Adipogénesis/genética , Adolescente , Adulto , Proteínas Portadoras/genética , Biología Computacional/métodos , Gránulos Citoplasmáticos/metabolismo , ARN Helicasas DEAD-box/genética , Femenino , Técnica del Anticuerpo Fluorescente , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Persona de Mediana Edad , Osteogénesis/genética , Unión Proteica , Transporte de Proteínas , Proteómica , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Adulto Joven
2.
Exp Mol Pathol ; 90(2): 149-56, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21111728

RESUMEN

Stem cell therapy has been considered a promise for damaged myocardial tissue. We have previously shown that S-nitroso-N-acetyl-D,L-penicillamine (SNAP) increases the expression of several muscular markers and VEGF in mesenchymal stem cells, indicating that transplantation of SNAP-treated cells could provide better functional outcomes. Here, we transplanted SNAP-treated adipose tissue-derived stem cells (ADSCs) in rat infarcted myocardium. After 30days, we observed a significant improvement of the ejection fraction in rats that received SNAP-treated ADSCs, compared with those that received untreated cells (p=0.008). Immunohistochemical reactions showed an increased expression of troponin T-C and von Willebrand factor, and organized vascular units in the infarcted area of tissue transplanted with treated ADSCs. SNAP exposure induced intracellular S-nitrosation, a decreased GSH/GSSG ratio, but did not increase cGMP levels. Collectively, these results indicate that SNAP alters the redox environment of ADSCs, possibly associated with a pre-differentiation state, which may improve cardiac function after transplantation.


Asunto(s)
Tejido Adiposo/citología , Corazón/fisiopatología , Infarto del Miocardio/terapia , Neovascularización Fisiológica/efectos de los fármacos , S-Nitroso-N-Acetilpenicilamina/farmacología , Trasplante de Células Madre , Células Madre/citología , Animales , Glutatión/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Corazón/efectos de los fármacos , Pruebas de Función Cardíaca/efectos de los fármacos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Masculino , Infarto del Miocardio/fisiopatología , Nitrosación/efectos de los fármacos , Ratas , Ratas Wistar , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Volumen Sistólico/efectos de los fármacos , Troponina/metabolismo , Factor de von Willebrand/metabolismo
3.
Biochem Biophys Res Commun ; 378(3): 456-61, 2009 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-19032948

RESUMEN

Mesenchymal stem cells (MSCs) have received special attention for cardiomyoplasty because several studies have shown that they differentiate into cardiomyocytes both in vitro and in vivo. Nitric oxide (NO) is a free radical signaling molecule that regulates several differentiation processes including cardiomyogenesis. Here, we report an investigation of the effects of two NO agents (SNAP and DEA/NO), able to activate both cGMP-dependent and -independent pathways, on the cardiomyogenic potential of bone marrow-derived mesenchymal stem cells (BM-MSCs) and adipose tissue-derived stem cells (ADSCs). The cells were isolated, cultured and treated with NO agents. Cardiac- and muscle-specific gene expression was analyzed by indirect immunofluorescence, flow cytometry, RT-PCR and real-time PCR. We found that untreated (control) ADSCs and BM-MSCs expressed some muscle markers and NO-derived intermediates induce an increased expression of some cardiac function genes in BM-MSCs and ADSCs. Moreover, NO agents considerably increased the pro-angiogenic potential mostly of BM-MSCs as determined by VEGF mRNA levels.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Hidrazinas/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Miocitos Cardíacos/citología , Donantes de Óxido Nítrico/farmacología , Penicilamina/análogos & derivados , Adulto , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Anciano , Antígenos CD/genética , Cardiomioplastia , Diferenciación Celular/genética , Células Cultivadas , Conexina 43/genética , Expresión Génica , Marcadores Genéticos , Corazón/fisiología , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Células Madre Multipotentes/citología , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/metabolismo , Proteínas Musculares/genética , Óxido Nítrico/metabolismo , Penicilamina/farmacología , Factor A de Crecimiento Endotelial Vascular/genética
4.
Sci Rep ; 9(1): 5611, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30948750

RESUMEN

The adipogenic process is characterized by the expression of adipocyte differentiation markers that lead to changes in cell metabolism and to the accumulation of lipid droplets. Moreover, during early adipogenesis, cells undergo a strong downregulation of translational activity with a decrease in cell size, proliferation and migration. In the present study, we identified that after 24 hours of adipogenic induction, human adipose tissue-derived stem cells (hASCs) undergo a G1-cell cycle arrest consistent with reduced proliferation, and this effect was correlated with a shift in polysome profile with an enrichment of the monosomal fraction and a reduction of the polysomal fraction. Polysome profiling analysis also revealed that this change in the monosomal/polysomal ratio was related to a strong downregulation of cell cycle and proliferation genes, such as cyclins and cyclin-dependent kinases (CDKs). Comparing total and polysome-associated mRNA sequencing, we also observed that this downregulation was mostly due to a reduction of cell cycle and proliferation transcripts via control of total mRNA abundance, rather than by translational control.


Asunto(s)
Adipogénesis/genética , Proteínas de Ciclo Celular/genética , Células Madre Mesenquimatosas/metabolismo , Adipocitos/metabolismo , Adipogénesis/fisiología , Tejido Adiposo/metabolismo , Adolescente , Adulto , Ciclo Celular , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Células Cultivadas , Femenino , Regulación de la Expresión Génica/genética , Humanos , Gotas Lipídicas/metabolismo , Masculino , Células Madre Mesenquimatosas/patología , Persona de Mediana Edad , ARN Mensajero/genética , Células Madre/citología , Transcriptoma/genética
5.
Stem Cell Res ; 25: 191-201, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29156375

RESUMEN

Commitment of adult stem cells involves the activation of specific gene networks regulated from transcription to protein synthesis. Here, we used ribosome profiling to identify mRNAs regulated at the translational level, through both differential association to polysomes and modulation of their translational rates. We observed that translational regulation during the differentiation of human adipose-derived stromal cells (hASCs, also known as adipose-derived mesenchymal stem cells), a subset of which are stem cells, to adipocytes was a major regulatory event. hASCs showed a significant reduction of whole protein synthesis after adipogenic induction and a downregulation of the expression and translational efficiency of ribosomal proteins. Additionally, focal adhesion and cytoskeletal proteins were downregulated at the translational level. This negative regulation of the essential biological functions of hASCs resulted in a reduction in cell size and the potential of hASCs to migrate. We analyzed whether the inactivation of key translation initiation factors was involved in this observed major repression of translation. We showed that there was an increase in the hypo phosphorylated forms of 4E-BP1, a negative regulator of translation, during early adipogenesis. Our results showed that extensive translational regulation occurred during the early stage of the adipogenic differentiation of hASCs.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis , Células Madre Mesenquimatosas/metabolismo , Biosíntesis de Proteínas , Células del Estroma/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adipocitos/citología , Proteínas de Ciclo Celular , Regulación hacia Abajo , Regulación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/citología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Células del Estroma/citología
6.
Stem Cell Res ; 17(2): 413-421, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27653462

RESUMEN

The understanding of metabolism during cell proliferation and commitment provides a greater insight into the basic biology of cells, allowing future applications. Here we evaluated the energy and oxidative changes during the early adipogenic differentiation of human adipose tissue-derived stromal cells (hASCs). hASCs were maintained under differentiation conditions during 3 and 7days. Oxygen consumption, mitochondrial mass and membrane potential, reactive oxygen species (ROS) generation, superoxide dismutase (SOD) and catalase activities, non-protein thiols (NPT) concentration and lipid peroxidation were analyzed. We observed that 7days of adipogenic induction are required to stimulate cells to consume more oxygen and increase mitochondrial activity, indicating organelle maturation and a transition from glycolytic to oxidative energy metabolism. ROS production was only increased after 3days and may be involved in the differentiation commitment. ROS source was not only the mitochondria and we suggest that NOX proteins are related to ROS generation and therefore adipogenic commitment. ROS production did not change after 7days, but an increased activity of catalase and NPT concentration as well as a decreased lipid peroxidation were observed. Thus, a short period of differentiation induction is able to change the energetic and oxidative metabolic profile of hASCs and stimulate cytoprotection processes.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/metabolismo , Adipogénesis , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Catalasa/metabolismo , Células Cultivadas , Glucólisis , Humanos , Peroxidación de Lípido , Potencial de la Membrana Mitocondrial , Células Madre Mesenquimatosas/citología , Microscopía Fluorescente , Mitocondrias/metabolismo , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Fosforilación Oxidativa , Consumo de Oxígeno , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
7.
Stem Cell Res ; 11(2): 902-12, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23845413

RESUMEN

Adipocyte stem cells (hASCs) can proliferate and self-renew and, due to their multipotent nature, they can differentiate into several tissue-specific lineages, making them ideal candidates for use in cell therapy. Most attempts to determine the mRNA profile of self-renewing or differentiating stem cells have made use of total RNA for gene expression analysis. Several lines of evidence suggest that self-renewal and differentiation are also dependent on the control of protein synthesis by posttranscriptional mechanisms. We used adipogenic differentiation as a model, to investigate the extent to which posttranscriptional regulation controlled gene expression in hASCs. We focused on the initial steps of differentiation and isolated both the total mRNA fraction and the subpopulation of mRNAs associated with translating ribosomes. We observed that adipogenesis is committed in the first days of induction and three days appears as the minimum time of induction necessary for efficient differentiation. RNA-seq analysis showed that a significant percentage of regulated mRNAs were posttranscriptionally controlled. Part of this regulation involves massive changes in transcript untranslated regions (UTR) length, with differential extension/reduction of the 3'UTR after induction. A slight correlation can be observed between the expression levels of differentially expressed genes and the 3'UTR length. When we considered association to polysomes, this correlation values increased. Changes in the half lives were related to the extension of the 3'UTR, with longer UTRs mainly stabilizing the transcripts. Thus, changes in the length of these extensions may be associated with changes in the ability to associate with polysomes or in half-life.


Asunto(s)
Adipocitos/fisiología , Polirribosomas/fisiología , Células Madre/fisiología , Regiones no Traducidas 3' , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis , Adulto , Diferenciación Celular/fisiología , Femenino , Regulación de la Expresión Génica , Glutatión/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Polirribosomas/genética , Polirribosomas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Madre/citología , Células Madre/metabolismo , Transcripción Genética , Adulto Joven
8.
Stem Cells Dev ; 21(2): 217-27, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-21649561

RESUMEN

Stem cells can either differentiate into more specialized cells or undergo self-renewal. Several lines of evidence from different organisms suggest that these processes depend on the post-transcriptional regulation of gene expression. The presence of the PUF [Pumilio/FBF (fem-3 binding factor)] domain defines a conserved family of RNA binding proteins involved in repressing gene expression. It has been suggested that a conserved function of PUF proteins is to repress differentiation and sustain the mitotic proliferation of stem cells. In humans, Pumilio-2 (PUM2) is expressed in embryonic stem cells and adult germ cells. Here we show that PUM2 is expressed in a subpopulation of adipose-derived stem cell (ASC) cultures, with a granular pattern of staining in the cytoplasm. Protein levels of PUM2 showed no changes during the differentiation of ASCs into adipocytes. Moreover, RNAi knockdown of pum2 did not alter the rate of adipogenic differentiation compared with wild-type control cells. A ribonomic approach was used to identify PUM2-associated mRNAs. Microarray analysis showed that PUM2-bound mRNAs are part of gene networks involved in cell proliferation and gene expression control. We studied pum2 expression in cell cultures with low or very high levels of proliferation and found that changes in pum2 production were dependent on the proliferation status of the cell. Transient knockdown of pum2 expression by RNAi impaired proliferation of ASCs in vitro. Our results suggest that PUM2 does not repress differentiation of ASCs but rather is involved in the positive control of ASCs division and proliferation.


Asunto(s)
Adipocitos/metabolismo , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Mitosis/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Adipocitos/citología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Células Madre Mesenquimatosas/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA