Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Microsc Microanal ; 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39027929

RESUMEN

Zinc is a critical ion for a large number of cellular functions. In the central nervous system, zinc ions are involved in synaptic transmission. Therefore, zinc homeostasis is essential, and cells have developed a variety of mechanisms to control cellular zinc concentration, including the zincosome formation. Alterations of free zinc levels have been associated with brain dysfunction and are present in many illnesses and syndromes. Astrocytes are implicated in the maintenance of the neuronal milleu and brain homeostasis. In this work, we have analyzed the combination of direct (TSQ) and indirect (autometallography) zinc detection methods to increase sensitivity for studying zinc uptake by rat astrocytes in vitro. Zincosome formation was visualized with the zinc fluorochrome TSQ by light microscopy. Additionally, we improved both zinc precipitation and cellular fixation methods to preserve zinc ions and make them suitable for autometallography development. Our tests pinpointed paraformaldehyde and sodium sulfide as the more adequate methods for cellular fixation and zinc precipitation, respectively. TSQ incubation and pH of the fixative were shown to be crucial for autometallography. Using this improved method, we visualized the zinc content of zincosomes at the ultrastructural level both as silver autometallographic precipitates and as electrodense sulfide-osmium zinc precipitates.

2.
Cell Death Discov ; 9(1): 217, 2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-37393339

RESUMEN

Charcot-Marie-Tooth disease is a chronic hereditary motor and sensory polyneuropathy targeting Schwann cells and/or motor neurons. Its multifactorial and polygenic origin portrays a complex clinical phenotype of the disease with a wide range of genetic inheritance patterns. The disease-associated gene GDAP1 encodes for a mitochondrial outer membrane protein. Mouse and insect models with mutations in Gdap1 have reproduced several traits of the human disease. However, the precise function in the cell types affected by the disease remains unknown. Here, we use induced-pluripotent stem cells derived from a Gdap1 knockout mouse model to better understand the molecular and cellular phenotypes of the disease caused by the loss-of-function of this gene. Gdap1-null motor neurons display a fragile cell phenotype prone to early degeneration showing (1) altered mitochondrial morphology, with an increase in the fragmentation of these organelles, (2) activation of autophagy and mitophagy, (3) abnormal metabolism, characterized by a downregulation of Hexokinase 2 and ATP5b proteins, (4) increased reactive oxygen species and elevated mitochondrial membrane potential, and (5) increased innate immune response and p38 MAP kinase activation. Our data reveals the existence of an underlying Redox-inflammatory axis fueled by altered mitochondrial metabolism in the absence of Gdap1. As this biochemical axis encompasses a wide variety of druggable targets, our results may have implications for developing therapies using combinatorial pharmacological approaches and improving therefore human welfare. A Redox-immune axis underlying motor neuron degeneration caused by the absence of Gdap1. Our results show that Gdap1-/- motor neurons have a fragile cellular phenotype that is prone to degeneration. Gdap1-/- iPSCs differentiated into motor neurons showed an altered metabolic state: decreased glycolysis and increased OXPHOS. These alterations may lead to hyperpolarization of mitochondria and increased ROS levels. Excessive amounts of ROS might be the cause of increased mitophagy, p38 activation and inflammation as a cellular response to oxidative stress. The p38 MAPK pathway and the immune response may, in turn, have feedback mechanisms, leading to the induction of apoptosis and senescence, respectively. CAC, citric acid cycle; ETC, electronic transport chain; Glc, glucose; Lac, lactate; Pyr, pyruvate.

3.
Neurotherapeutics ; 18(4): 2565-2578, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34697772

RESUMEN

Dopamine replacement represents the standard therapy for Parkinson's disease (PD), a common, chronic, and incurable neurological disorder; however, this approach only treats the symptoms of this devastating disease. In the search for novel disease-modifying therapies that target other relevant molecular and cellular mechanisms, Drosophila has emerged as a valuable tool to study neurodegenerative diseases due to the presence of a complex central nervous system, the blood-brain barrier, and a similar neurotransmitter profile to humans. Human PD-related genes also display conservation in flies; DJ-1ß is the fly ortholog of DJ-1, a gene for which mutations prompt early-onset recessive PD. Interestingly, flies mutant for DJ-1ß exhibit PD-related phenotypes, including motor defects, high oxidative stress (OS) levels and metabolic alterations. To identify novel therapies for PD, we performed an in vivo high-throughput screening assay using DJ-1ß mutant flies and compounds from the Prestwick® chemical library. Drugs that improved motor performance in DJ-1ß mutant flies were validated in DJ-1-deficient human neural-like cells, revealing that zaprinast displayed the most significant ability to suppress OS-induced cell death. Zaprinast inhibits phosphodiesterases and activates GPR35, an orphan G-protein-coupled receptor not previously associated with PD. We found that zaprinast exerts its beneficial effect in both fly and human PD models through several disease-modifying mechanisms, including reduced OS levels, attenuated apoptosis, increased mitochondrial viability, and enhanced glycolysis. Therefore, our results support zaprinast as a potential therapeutic for PD in future clinical trials.


Asunto(s)
Enfermedad de Parkinson , Animales , Drosophila/genética , Drosophila/metabolismo , Estrés Oxidativo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Proteína Desglicasa DJ-1/genética , Proteína Desglicasa DJ-1/metabolismo , Purinonas/metabolismo , Purinonas/farmacología , Purinonas/uso terapéutico
4.
Stem Cell Rev Rep ; 17(6): 2245-2261, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34476741

RESUMEN

Metabolic rewiring and mitochondrial dynamics remodelling are hallmarks of cell reprogramming, but the roles of the reprogramming factors in these changes are not fully understood. Here we show that c-MYC induces biosynthesis of fatty acids and increases the rate of pentose phosphate pathway. Time-course profiling of fatty acids and complex lipids during cell reprogramming using lipidomics revealed a profound remodelling of the lipid content, as well as the saturation and length of their acyl chains, in a c-MYC-dependent manner. Pluripotent cells displayed abundant cardiolipins and scarce phosphatidylcholines, with a prevalence of monounsaturated acyl chains. Cells undergoing cell reprogramming showed an increase in mitochondrial membrane potential that paralleled that of mitochondrial-specific cardiolipins. We conclude that c-MYC controls the rewiring of somatic cell metabolism early in cell reprogramming by orchestrating cell proliferation, synthesis of macromolecular components and lipid remodelling, all necessary processes for a successful phenotypic transition to pluripotency. c-MYC promotes anabolic metabolism, mitochondrial fitness and lipid remodelling early in cell reprogramming. A high rate of aerobic glycolysis is crucial to provide intermediaries for biosynthetic pathways. To ensure the availability of nucleotides, amino acids and lipids for cell proliferation, cells must provide with a constant flux of the elemental building blocks for macromolecule assembly and fulfil the anabolic demands to reach the critical cellular mass levels to satisfactorily undergo cell division. A high rate of aerobic glycolysis is induced by c-MYC, increasing the amounts of intracellular Glucose-6-phosphate (G6P), fructose-6-phosphate (F6P), and glyceraldehyde-3-phosphate (GA3P), which can all enter pentose phosphate pathway (PPP) to produce Ribose-5-Phosphate (R5P) and NADPH, which are necessary for the biosynthesis of biomolecules such as proteins, nucleic acids, or lipids. C-MYC-dependent activation of glucose-6-phosphate dehydrogenase (G6PD) may play a critical role in the shunting of G6P to PPP and generation of NADPH. High glycolytic flux increases the amounts of dihydroxyacetone phosphate (DHAP), which is crucial for biosynthesis of phospholipids and triacylglycerols, and pyruvate (Pyr), which can be converted to citrate (Cit) in the mitochondria and enter the biosynthesis of fatty acids (FA). During cell reprogramming, c-MYC-dependent lipid remodelling leads to Polyunsaturated Fatty Acid (PUFA) downregulation and Monounsaturated Fatty Acid (MUFA) upregulation, which may play critical roles in cytoarchitectural remodelling of cell membrane or non-canonical autophagy, respectively. Cardiolipin (pink dots) rise early in cell reprogramming correlates with an increase in mitochondrial fitness, suggesting that c-MYC may restore proper levels of cardiolipins and antioxidant proteins, such as UCP2, to guarantee an optimal mitochondrial function while upholding ROS levels, reinforcing the idea of cell rejuvenation early in cell reprogramming.


Asunto(s)
Reprogramación Celular , Vía de Pentosa Fosfato , Reprogramación Celular/genética , Glucólisis , Lípidos , Dinámicas Mitocondriales
5.
Free Radic Biol Med ; 158: 137-148, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32726690

RESUMEN

Parkinson's disease (PD) is a neurodegenerative debilitating disorder characterized by progressive disturbances in motor, autonomic and psychiatric functions. One of the genes involved in familial forms of the disease is DJ-1, whose mutations cause early-onset PD. Besides, it has been shown that an over-oxidized and inactive form of the DJ-1 protein is found in brains of sporadic PD patients. Interestingly, the DJ-1 protein plays an important role in cellular defense against oxidative stress and also participates in mitochondrial homeostasis. Valuable insights into potential PD pathogenic mechanisms involving DJ-1 have been obtained from studies in cell and animal PD models based on DJ-1 deficiency such as Drosophila. Flies mutant for the DJ-1ß gene, the Drosophila ortholog of human DJ-1, exhibited disease-related phenotypes such as motor defects, increased reactive oxygen species production and high levels of protein carbonylation. In the present study, we demonstrate that DJ-1ß mutants also show a significant increase in the activity of several regulatory glycolytic enzymes. Similar results were obtained in DJ-1-deficient SH-SY5Y neuroblastoma cells, thus suggesting that loss of DJ-1 function leads to an increase in the glycolytic rate. In such a scenario, an enhancement of the glycolytic pathway could be a protective mechanism to decrease ROS production by restoring ATP levels, which are decreased due to mitochondrial dysfunction. Our results also show that meclizine and dimethyl fumarate, two FDA-approved compounds with different clinical applications, are able to attenuate PD-related phenotypes in both models. Moreover, we found that they may exert their beneficial effect by increasing glycolysis through the activation of key glycolytic enzymes. Taken together, these results are consistent with the idea that increasing glycolysis could be a potential disease-modifying strategy for PD, as recently suggested. Besides, they also support further evaluation and potential repurposing of meclizine and dimethyl fumarate as modulators of energy metabolism for neuroprotection in PD.


Asunto(s)
Proteínas de Drosophila , Enfermedad de Parkinson , Animales , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Glucólisis , Humanos , Proteínas del Tejido Nervioso/metabolismo , Estrés Oxidativo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , Proteína Desglicasa DJ-1/genética
6.
Exp Gerontol ; 133: 110870, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32045634

RESUMEN

Somatic cells can be reprogrammed to pluripotency by either ectopic expression of defined factors or exposure to chemical cocktails. During reprogramming, somatic cells undergo dramatic changes in a wide range of cellular processes, such as metabolism, mitochondrial morphology and function, cell signaling pathways or immortalization. Regulation of these processes during cell reprograming lead to the acquisition of a pluripotent state, which enables indefinite propagation by symmetrical self-renewal without losing the ability of reprogrammed cells to differentiate into all cell types of the adult. In this review, recent data from different laboratories showing how these processes are controlled during the phenotypic transformation of a somatic cell into a pluripotent stem cell will be discussed.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas , Adulto , Diferenciación Celular , Humanos , Dinámicas Mitocondriales , Transducción de Señal
7.
Stem Cell Reports ; 11(6): 1479-1492, 2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30472011

RESUMEN

Cell reprogramming is thought to be associated with a full metabolic switch from an oxidative- to a glycolytic-based metabolism. However, neither the dynamics nor the factors controlling this metabolic switch are fully understood. By using cellular, biochemical, protein array, metabolomic, and respirometry analyses, we found that c-MYC establishes a robust bivalent energetics program early in cell reprogramming. Cells prone to undergo reprogramming exhibit high mitochondrial membrane potential and display a hybrid metabolism. We conclude that MYC proteins orchestrate a rewiring of somatic cell metabolism early in cell reprogramming, whereby somatic cells acquire the phenotypic plasticity necessary for their transition to pluripotency in response to either intrinsic or external cues.


Asunto(s)
Reprogramación Celular , Células Híbridas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Proteína Quinasa CDC2/metabolismo , Glucólisis , Humanos , Potencial de la Membrana Mitocondrial , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Fosforilación Oxidativa , Fosforilación
8.
Stem Cells Int ; 2017: 8073721, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28484497

RESUMEN

Somatic cells can be reprogrammed into a pluripotent cellular state similar to that of embryonic stem cells. Given the significant physiological differences between the somatic and pluripotent cells, cell reprogramming is associated with a profound reorganization of the somatic phenotype at all levels. The remodeling of mitochondrial morphology is one of these dramatic changes that somatic cells have to undertake during cell reprogramming. Somatic cells transform their tubular and interconnected mitochondrial network to the fragmented and isolated organelles found in pluripotent stem cells early during cell reprogramming. Accordingly, mitochondrial fission, the process whereby the mitochondria divide, plays an important role in the cell reprogramming process. Here, we present an overview of the importance of mitochondrial fission in both cell reprogramming and cellular transformation.

9.
Stem Cell Res ; 18: 1-4, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28395795

RESUMEN

Human CMT2-FiPS4F1 cell line was generated from fibroblasts of a patient with Charcot-Marie-Tooth disease harbouring the following mutations in the GDAP1 gene in heterozygosis: p.Q163X/p.T288NfsX3. This patient did not present mutations in the PM22, MPZ or GJB genes. Human reprogramming factors OCT3/4, KLF4, SOX2 and C-MYC were delivered using a non-integrative methodology that involves the use of Sendai virus.


Asunto(s)
Reprogramación Celular , Enfermedad de Charcot-Marie-Tooth/patología , Células Madre Pluripotentes Inducidas/citología , Proteínas del Tejido Nervioso/genética , Secuencia de Bases , Diferenciación Celular , Línea Celular , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/metabolismo , Análisis Mutacional de ADN , Fibroblastos/citología , Fibroblastos/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Heterocigoto , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipo , Factor 4 Similar a Kruppel , Masculino , Microscopía Fluorescente , Polimorfismo de Nucleótido Simple , Virus Sendai/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Cell Cycle ; 15(23): 3240-3250, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27753531

RESUMEN

We have recently shown that mitochondrial fission is induced early in reprogramming in a Drp1-dependent manner; however, the identity of the factors controlling Drp1 recruitment to mitochondria was unexplored. To investigate this, we used a panel of RNAi targeting factors involved in the regulation of mitochondrial dynamics and we observed that MiD51, Gdap1 and, to a lesser extent, Mff were found to play key roles in this process. Cells derived from Gdap1-null mice were used to further explore the role of this factor in cell reprogramming. Microarray data revealed a prominent down-regulation of cell cycle pathways in Gdap1-null cells early in reprogramming and cell cycle profiling uncovered a G2/M growth arrest in Gdap1-null cells undergoing reprogramming. High-Content analysis showed that this growth arrest was DNA damage-independent. We propose that lack of efficient mitochondrial fission impairs cell reprogramming by interfering with cell cycle progression in a DNA damage-independent manner.


Asunto(s)
Reprogramación Celular , Dinámicas Mitocondriales , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Daño del ADN , Fase G2/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Ratones , Dinámicas Mitocondriales/efectos de los fármacos , Mitosis/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/farmacología
11.
Stem Cell Res Ther ; 7(1): 96, 2016 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-27460218

RESUMEN

BACKGROUND: Several studies have reported the direct conversion of mouse fibroblasts to hepatocyte-like cells with different degrees of maturation by expression of hepatic fate-conversion factors. METHODS: We have used a combination of lentiviral vectors expressing hepatic fate-conversion factors with Oct4, Sox2, Klf4, and Myc to convert mouse embryonic fibroblasts into hepatic cells. RESULTS: We have generated hepatic cells with progenitor-like features (iHepL cells). iHepL cells displayed basic hepatocyte functions but failed to perform functions characteristic of mature hepatocytes such as significant Cyp450 or urea cycle activities. iHepL cells expressed multiple hepatic-specific transcription factors and functional genes characteristic of immature hepatocytes and cholangiocytes, as well as high levels of Foxl1, Cd24a, and Lgr5, specific markers of hepatic progenitor cells. When transplanted into partial hepatectomized and hepatic irradiated mice, they differentiated into hepatocytes and cholangiocytes. However, iHepL cells formed malignant non-teratoma cell aggregations in one out of five engrafted livers and five out of five xenografts assays. All the cells in these tumors had silenced key hepatic fate-conversion factors, and lost hepatic features. CONCLUSIONS: This study highlights the dangers of using pluripotency factors in reprogramming strategies when fate-conversion factors are silenced in vivo, and urges us to perform extensive tumorigenic tests in reprogrammed cells.


Asunto(s)
Carcinogénesis/genética , Reprogramación Celular , Fibroblastos/metabolismo , Silenciador del Gen , Teratoma/genética , Animales , Biomarcadores/metabolismo , Antígeno CD24/genética , Antígeno CD24/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Diferenciación Celular , Embrión de Mamíferos , Fibroblastos/citología , Fibroblastos/trasplante , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Hepatectomía , Hepatocitos/metabolismo , Hepatocitos/patología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Células Madre/metabolismo , Células Madre/patología , Teratoma/metabolismo , Teratoma/patología , Transgenes
12.
Nat Commun ; 7: 11124, 2016 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-27030341

RESUMEN

During the process of reprogramming to induced pluripotent stem (iPS) cells, somatic cells switch from oxidative to glycolytic metabolism, a transition associated with profound mitochondrial reorganization. Neither the importance of mitochondrial remodelling for cell reprogramming, nor the molecular mechanisms controlling this process are well understood. Here, we show that an early wave of mitochondrial fragmentation occurs upon expression of reprogramming factors. Reprogramming-induced mitochondrial fission is associated with a minor decrease in mitochondrial mass but not with mitophagy. The pro-fission factor Drp1 is phosphorylated early in reprogramming, and its knockdown and inhibition impairs both mitochondrial fragmentation and generation of iPS cell colonies. Drp1 phosphorylation depends on Erk activation in early reprogramming, which occurs, at least in part, due to downregulation of the MAP kinase phosphatase Dusp6. Taken together, our data indicate that mitochondrial fission controlled by an Erk-Drp1 axis constitutes an early and necessary step in the reprogramming process to pluripotency.


Asunto(s)
Reprogramación Celular , Dinaminas/fisiología , Células Madre Pluripotentes Inducidas/citología , Sistema de Señalización de MAP Quinasas , Dinámicas Mitocondriales , Animales , Línea Celular , Dinaminas/genética , Dinaminas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/ultraestructura , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
13.
Cell Stem Cell ; 12(1): 88-100, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23260487

RESUMEN

In the adult brain, continual neurogenesis of olfactory neurons is sustained by the existence of neural stem cells (NSCs) in the subependymal niche. Elimination of the cyclin-dependent kinase inhibitor 1A (p21) leads to premature exhaustion of the subependymal NSC pool, suggesting a relationship between cell cycle control and long-term self-renewal, but the molecular mechanisms underlying NSC maintenance by p21 remain unexplored. Here we identify a function of p21 in the direct regulation of the expression of pluripotency factor Sox2, a key regulator of the specification and maintenance of neural progenitors. We observe that p21 directly binds a Sox2 enhancer and negatively regulates Sox2 expression in NSCs. Augmented levels of Sox2 in p21 null cells induce replicative stress and a DNA damage response that leads to cell growth arrest mediated by increased levels of p19(Arf) and p53. Our results show a regulation of NSC expansion driven by a p21/Sox2/p53 axis.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Mutantes , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1/genética
14.
Stem Cells ; 31(1): 71-82, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23132827

RESUMEN

Somatic cells can be reprogrammed to induced pluripotent stem (iPS) cells by ectopic expression of the four factors Oct4, Klf4, Sox2, and Myc. Here, we investigated the role of Gata4 in the reprogramming process and present evidence for a negative role of this family of transcription factors in the induction of pluripotency. Coexpression of Gata4 with Oct4, Klf4, and Sox2 with or without Myc in mouse embryonic fibroblasts greatly impaired reprogramming and endogenous Nanog expression. The lack of Nanog upregulation was associated with a blockade in the transition from the initiation phase of reprogramming to the full pluripotent state characteristic of iPS cells. Addition of Nanog to the reprogramming cocktail blocked the deleterious effects observed with Gata4 expression. Downregulation of endogenous Gata4 by short hairpin RNAs during reprogramming both accelerated and increased the efficiency of the process and augmented the mRNA levels of endogenous Nanog. Using comparative genomics, we identified a consensus binding site for Gata factors in an evolutionary conserved region located 9 kb upstream of the Nanog gene. Using chromatin immunoprecipitation, gel retardation, and luciferase assays, we found that Gata4 bound to this region and inhibited Nanog transcription in mouse embryonic stem cells. Overall, our results describe for first time the negative effect of Gata4 in the reprogramming of somatic cells and highlight the role of Gata factors in the transcriptional networks that control cell lineage choices in the early embryo.


Asunto(s)
Reprogramación Celular , Células Madre Embrionarias/metabolismo , Factor de Transcripción GATA4/metabolismo , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Inmunoprecipitación de Cromatina , Regulación hacia Abajo , Ensayo de Cambio de Movilidad Electroforética , Factor de Transcripción GATA4/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Proteínas de Homeodominio/genética , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño , Factores de Transcripción SOXB1/metabolismo , Transcripción Genética , Activación Transcripcional
15.
PLoS One ; 7(4): e36405, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22558462

RESUMEN

The ability to direct differentiation of mouse embryonic stem (ES) cells into specific lineages not only provides new insights into the pathways that regulate lineage selection but also has translational applications, for example in drug discovery. We set out to develop a method of differentiating ES cells into mesodermal cells at high efficiency without first having to induce embryoid body formation. ES cells were plated on a feeder layer of PA6 cells, which have membrane-associated stromal-derived inducing activity (SDIA), the molecular basis of which is currently unknown. Stimulation of ES/PA6 co-cultures with Bone Morphogenetic Protein 4 (BMP4) both favoured self-renewal of ES cells and induced differentiation into a Desmin and Nestin double positive cell population. Combined stimulation with BMP4 and all-trans Retinoic Acid (RA) inhibited self-renewal and resulted in 90% of cells expressing Desmin and Nestin. Quantitative reverse transcription-polymerase chain reaction (qPCR) analysis confirmed that the cells were of mesodermal origin and expressed markers of mesenchymal and smooth muscle cells. BMP4 activation of a MAD-homolog (Smad)-dependent reporter in undifferentiated ES cells was attenuated by co-stimulation with RA and co-culture with PA6 cells. The Notch ligand Jag1 was expressed in PA6 cells and inhibition of Notch signalling blocked the differentiation inducing activity of PA6 cells. Our data suggest that mesodermal differentiation is regulated by the level of Smad activity as a result of inputs from BMP4, RA and the Notch pathway.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Mesodermo/citología , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología , Animales , Línea Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Mesodermo/efectos de los fármacos , Ratones , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Transcriptoma/efectos de los fármacos
16.
Nat Cell Biol ; 10(2): 194-201, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18223644

RESUMEN

Embryonic stem (ES) cells are pluripotent cells derived from the inner cell mass of blastocysts. Self-renewal of mouse ES cells depends on activation of Stat3 by leukaemia inhibitory factor (LIF) in collaboration with bone morphogenetic protein signalling. The transcription factor Nanog is essential in maintaining pluripotency but the mechanisms involved are poorly understood. Here we examine the functional interactions of Nanog with the Stat3 and NFkappaB pathways. Nanog and Stat3 were found to bind to and synergistically activate Stat3-dependent promoters. We also found that Nanog binds to NFkappaB proteins; however, Nanog binding inhibited transcriptional activity of NFkappaB proteins. Endogenous NFkappaB activity and target-gene expression increased during differentiation of ES cells. Overexpression of NFkappaB proteins promoted differentiation, whereas inhibition of NFkappaB signalling, either by genetic ablation of the Ikbkg gene or overexpression of the IkappaBalpha super-repressor, increased expression of pluripotency markers. We conclude that Nanog represses the pro-differentiation activities of NFkappaB and cooperates with Stat3 to maintain pluripotency.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Células Madre Embrionarias/fisiología , Proteínas de Homeodominio/fisiología , FN-kappa B/antagonistas & inhibidores , Células Madre Pluripotentes/fisiología , Factor de Transcripción STAT3/metabolismo , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Activación Enzimática , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Ratones , FN-kappa B/genética , Proteína Homeótica Nanog , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Factor de Transcripción STAT3/genética , Transducción de Señal/fisiología
17.
Mol Biol Cell ; 17(9): 4002-13, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16807353

RESUMEN

The targeting of the tumor suppressor PTEN protein to distinct subcellular compartments is a major regulatory mechanism of PTEN function, by controlling its access to substrates and effector proteins. Here, we investigated the molecular basis and functional consequences of PTEN nuclear/cytoplasmic distribution. PTEN accumulated in the nucleus of cells treated with apoptotic stimuli. Nuclear accumulation of PTEN was enhanced by mutations targeting motifs in distinct PTEN domains, and it was dependent on an N-terminal nuclear localization domain. Coexpression of a dominant negative Ran GTPase protein blocked PTEN accumulation in the nucleus, which was also affected by coexpression of importin alpha proteins. The lipid- and protein-phosphatase activity of PTEN differentially modulated PTEN nuclear accumulation. Furthermore, catalytically active nuclear PTEN enhanced cell apoptotic responses. Our findings indicate that multiple nuclear exclusion motifs and a nuclear localization domain control PTEN nuclear localization by a Ran-dependent mechanism and suggest a proapoptotic role for PTEN in the cell nucleus.


Asunto(s)
Apoptosis , Núcleo Celular/metabolismo , Señales de Localización Nuclear/metabolismo , Fosfohidrolasa PTEN/química , Fosfohidrolasa PTEN/metabolismo , Proteína de Unión al GTP ran/metabolismo , Células 3T3 , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Células COS , Catálisis , Células Cultivadas , Chlorocebus aethiops , Células HeLa , Humanos , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Eliminación de Secuencia
18.
J Biol Chem ; 280(32): 28936-43, 2005 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-15951562

RESUMEN

The tumor suppressor phosphatase PTEN is a key regulator of cell growth and apoptosis that interacts with PDZ domains from regulatory proteins, including MAGI-1/2/3, hDlg, and MAST205. Here we identified novel PTEN-binding PDZ domains within the MAST205-related proteins, syntrophin-associated serine/threonine kinase and MAST3, characterized the regions of PTEN involved in its interaction with distinctive PDZ domains, and analyzed the functional consequences on PTEN of PDZ domain binding. Using a panel of PTEN mutations, as well as PTEN chimeras containing distinct domains of the related protein TPTE, we found that the PTP and C2 domains of PTEN do not affect PDZ domain binding and that the C-terminal tail of PTEN (residues 350-403) provides selectivity to recognize specific PDZ domains from MAGI-2, hDlg, and MAST205. Binding of PTEN to the PDZ-2 domain from MAGI-2 increased PTEN protein stability. Furthermore, binding of PTEN to the PDZ domains from microtubule-associated serine/threonine kinases facilitated PTEN phosphorylation at its C terminus by these kinases. Our results suggest an important role for the C-terminal region of PTEN in the selective association with scaffolding and/or regulatory molecules and provide evidence that PDZ domain binding stabilizes PTEN and targets this tumor suppressor for phosphorylation by microtubule-associated serine/threonine kinases.


Asunto(s)
Proteínas Asociadas a la Distrofina/química , Microtúbulos/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secuencias de Aminoácidos , Animales , Células COS , Proteínas Portadoras , Homólogo 1 de la Proteína Discs Large , Glutatión Transferasa/metabolismo , Guanilato-Quinasas , Humanos , Inmunoprecipitación , Proteínas de la Membrana , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Mutación , Nucleósido-Fosfato Quinasa/metabolismo , Fosfohidrolasa PTEN , Monoéster Fosfórico Hidrolasas/química , Fosforilación , Plásmidos/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas/metabolismo , Proteínas Recombinantes de Fusión/química , Saccharomyces cerevisiae/metabolismo , Factores de Tiempo , Transfección , Proteínas Supresoras de Tumor/química , Técnicas del Sistema de Dos Híbridos
19.
Cancer Lett ; 223(2): 303-12, 2005 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-15896465

RESUMEN

The binding of PTEN to PDZ-domain-containing proteins appears to play an important role in the control of cell growth, motility and apoptosis. In turn, this binding can be abrogated by cleavage of the PTEN C-terminal region by caspase-3. We have generated and characterized monoclonal antibodies (mAb) directed against distinct epitopes at the C-terminal region of PTEN, and used them to define protein-binding epitopes on PTEN and to study its cleavage by caspase-3. mAb directed against epitopes at the far C-terminus of PTEN blocked binding to PTEN cognate PDZ domains and did not recognize the caspase-3 cleaved PTEN fragments. On the other hand, mAb that recognized an epitope within the C2 domain of PTEN did not prevent binding to PDZ domains, but could detect the caspase-3 cleaved PTEN fragments. The analysis of PTEN cleavage by caspase-3 revealed that the lipid phosphatase activity of PTEN controls its own degradation by interfering with the PI3-K anti-apoptotic activity. Our results define protein-binding sites on the PTEN tumor suppressor at the immunochemical level, and suggest a regulatory link between PTEN phosphatase activity, caspase-3 sensitivity and PTEN-protein interactions.


Asunto(s)
Sitios de Unión , Monoéster Fosfórico Hidrolasas/fisiología , Proteínas Supresoras de Tumor/fisiología , Caspasa 3 , Caspasas/metabolismo , Epítopos , Genes Supresores de Tumor , Humanos , Inmunohistoquímica , Proteínas de Neoplasias , Neoplasias/fisiopatología , Fosfohidrolasa PTEN , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA