Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Sci Adv ; 9(13): eade1792, 2023 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-36989351

RESUMEN

The blueprints of developing organs are preset at the early stages of embryogenesis. Transcriptional and epigenetic mechanisms are proposed to preset developmental trajectories. However, we reveal that the competence for the future cardiac fate of human embryonic stem cells (hESCs) is preset in pluripotency by a specialized mRNA translation circuit controlled by RBPMS. RBPMS is recruited to active ribosomes in hESCs to control the translation of essential factors needed for cardiac commitment program, including Wingless/Integrated (WNT) signaling. Consequently, RBPMS loss specifically and severely impedes cardiac mesoderm specification, leading to patterning and morphogenetic defects in human cardiac organoids. Mechanistically, RBPMS specializes mRNA translation, selectively via 3'UTR binding and globally by promoting translation initiation. Accordingly, RBPMS loss causes translation initiation defects highlighted by aberrant retention of the EIF3 complex and depletion of EIF5A from mRNAs, thereby abrogating ribosome recruitment. We demonstrate how future fate trajectories are programmed during embryogenesis by specialized mRNA translation.


Asunto(s)
Células Madre Embrionarias Humanas , Humanos , Células Madre Embrionarias Humanas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribosomas/metabolismo , Corazón , Transducción de Señal , Proteínas de Unión al ARN/metabolismo
3.
RNA ; 28(11): 1481-1495, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35973723

RESUMEN

Circular RNAs are an endogenous long-lived and abundant noncoding species. Despite their prevalence, only a few circRNAs have been dissected mechanistically to date. Here, we cataloged nascent RNA-enriched circRNAs from primary human cells and functionally assigned a role to circRAB3IP in sustaining cellular homeostasis. We combined "omics" and functional experiments to show how circRAB3IP depletion deregulates hundreds of genes, suppresses cell cycle progression, and induces senescence-associated gene expression changes. Conversely, excess circRAB3IP delivered to endothelial cells via extracellular vesicles suffices for accelerating their division. We attribute these effects to an interplay between circRAB3IP and the general splicing factor SF3B1, which can affect transcript variant expression levels of cell cycle-related genes. Together, our findings link the maintenance of cell homeostasis to the presence of a single circRNA.


Asunto(s)
MicroARNs , ARN Circular , Humanos , ARN Circular/genética , Células Endoteliales/metabolismo , Proliferación Celular/genética , ARN Mensajero/genética , Expresión Génica , MicroARNs/genética
4.
Int J Mol Sci ; 23(6)2022 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-35328717

RESUMEN

The evaluation of substances for their potency to induce embryotoxicity is controlled by safety regulations. Test guidelines for reproductive and developmental toxicity rely mainly on animal studies, which make up the majority of animal usage in regulatory toxicology. Therefore, there is an urgent need for alternative in vitro methods to follow the 3R principles. To improve human safety, cell models based on human cells are of great interest to overcome species differences. Here, human induced pluripotent stem cells (hiPSCs) are an ideal cell source as they largely recapitulate embryonic stem cells without bearing ethical concerns and they are able to differentiate into most cell types of the human body. Here, we set up and characterized a fetal bovine serum (FBS)-free hiPSC-based in vitro test method, called the human induced pluripotent stem cell test (hiPS Test), to evaluate the embryotoxic potential of substances. After 10 days in culture, hiPSCs develop into beating cardiomyocytes. As terminal endpoint evaluations, cell viability, qPCR analyses as well as beating frequency and area of beating cardiomyocytes by video analyses are measured. The embryotoxic positive and non-embryotoxic negative controls, 5-Fluorouracil (5-FU) and Penicillin G (PenG), respectively, were correctly assessed in the hiPS Test. More compounds need to be screened in the future for defining the assay's applicability domain, which will inform us of the suitability of the hiPS Test for detecting adverse effects of substances on embryonic development.


Asunto(s)
Células Madre Pluripotentes Inducidas , Animales , Diferenciación Celular , Células Madre Embrionarias , Fluorouracilo/farmacología , Humanos , Miocitos Cardíacos , Teratógenos/toxicidad , Pruebas de Toxicidad/métodos
5.
Nat Commun ; 12(1): 3014, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-34021162

RESUMEN

Members of the chromodomain-helicase-DNA binding (CHD) protein family are chromatin remodelers implicated in human pathologies, with CHD6 being one of its least studied members. We discovered a de novo CHD6 missense mutation in a patient clinically presenting the rare Hallermann-Streiff syndrome (HSS). We used genome editing to generate isogenic iPSC lines and model HSS in relevant cell types. By combining genomics with functional in vivo and in vitro assays, we show that CHD6 binds a cohort of autophagy and stress response genes across cell types. The HSS mutation affects CHD6 protein folding and impairs its ability to recruit co-remodelers in response to DNA damage or autophagy stimulation. This leads to accumulation of DNA damage burden and senescence-like phenotypes. We therefore uncovered a molecular mechanism explaining HSS onset via chromatin control of autophagic flux and genotoxic stress surveillance.


Asunto(s)
Autofagia/fisiología , Daño del ADN , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Autofagia/genética , Cromatina , Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión al ADN/metabolismo , Epigenómica , Edición Génica , Expresión Génica , Síndrome de Hallermann/genética , Humanos , Mutación , Fenotipo
6.
Cancer Res ; 81(6): 1457-1471, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33372039

RESUMEN

Neuroblastoma has a low mutation rate for the p53 gene. Alternative ways of p53 inactivation have been proposed in neuroblastoma, such as abnormal cytoplasmic accumulation of wild-type p53. However, mechanisms leading to p53 inactivation via cytoplasmic accumulation are not well investigated. Here we show that the neuroblastoma risk-associated locus 6p22.3-derived tumor suppressor NBAT1 is a p53-responsive lncRNA that regulates p53 subcellular levels. Low expression of NBAT1 provided resistance to genotoxic drugs by promoting p53 accumulation in cytoplasm and loss from mitochondrial and nuclear compartments. Depletion of NBAT1 altered CRM1 function and contributed to the loss of p53-dependent nuclear gene expression during genotoxic drug treatment. CRM1 inhibition rescued p53-dependent nuclear functions and sensitized NBAT1-depleted cells to genotoxic drugs. Combined inhibition of CRM1 and MDM2 was even more effective in sensitizing aggressive neuroblastoma cells with p53 cytoplasmic accumulation. Thus, our mechanistic studies uncover an NBAT1-dependent CRM1/MDM2-based potential combination therapy for patients with high-risk neuroblastoma. SIGNIFICANCE: This study shows how a p53-responsive lncRNA mediates chemotherapeutic response by modulating nuclear p53 pathways and identifies a potential treatment strategy for patients with high-risk neuroblastoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos/genética , Neuroblastoma/tratamiento farmacológico , ARN Largo no Codificante/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis , Fraccionamiento Celular , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Daño del ADN/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Carioferinas/antagonistas & inhibidores , Carioferinas/metabolismo , Masculino , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Neuroblastoma/genética , Neuroblastoma/patología , Neuroblastoma/cirugía , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , ARN Largo no Codificante/genética , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Exportina 1
7.
Commun Biol ; 3(1): 265, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32457386

RESUMEN

Blood vessels are constantly exposed to shear stress, a biomechanical force generated by blood flow. Normal shear stress sensing and barrier function are crucial for vascular homeostasis and are controlled by adherens junctions (AJs). Here we show that AJs are stabilized by the shear stress-induced long non-coding RNA LASSIE (linc00520). Silencing of LASSIE in endothelial cells impairs cell survival, cell-cell contacts and cell alignment in the direction of flow. LASSIE associates with junction proteins (e.g. PECAM-1) and the intermediate filament protein nestin, as identified by RNA affinity purification. The AJs component VE-cadherin showed decreased stabilization, due to reduced interaction with nestin and the microtubule cytoskeleton in the absence of LASSIE. This study identifies LASSIE as link between nestin and VE-cadherin, and describes nestin as crucial component in the endothelial response to shear stress. Furthermore, this study indicates that LASSIE regulates barrier function by connecting AJs to the cytoskeleton.


Asunto(s)
Células Endoteliales/metabolismo , ARN Largo no Codificante/metabolismo , Fenómenos Biomecánicos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Estrés Mecánico
9.
Hum Mol Genet ; 28(17): 2885-2899, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31127934

RESUMEN

Sulfite oxidase (SO) is encoded by the nuclear SUOX gene and catalyzes the final step in cysteine catabolism thereby oxidizing sulfite to sulfate. Oxidation of sulfite is dependent on two cofactors within SO, a heme and the molybdenum cofactor (Moco), the latter forming the catalytic site of sulfite oxidation. SO localizes to the intermembrane space of mitochondria where both-pre-SO processing and cofactor insertion-are essential steps during SO maturation. Isolated SO deficiency (iSOD) is a rare inborn error of metabolism caused by mutations in the SUOX gene that lead to non-functional SO. ISOD is characterized by rapidly progressive neurodegeneration and death in early infancy. We diagnosed an iSOD patient with homozygous mutation of SUOX at c.1084G>A replacing Gly362 to serine. To understand the mechanism of disease, we expressed patient-derived G362S SO in Escherichia coli and surprisingly found full catalytic activity, while in patient fibroblasts no SO activity was detected, suggesting differences between bacterial and human expression. Moco reconstitution of apo-G362S SO was found to be approximately 90-fold reduced in comparison to apo-WT SO in vitro. In line, levels of SO-bound Moco in cells overexpressing G362S SO were significantly reduced compared to cells expressing WT SO providing evidence for compromised maturation of G362S SO in cellulo. Addition of molybdate to culture medium partially rescued impaired Moco binding of G362S SO and restored SO activity in patient fibroblasts. Thus, this study demonstrates the importance of the orchestrated maturation of SO and provides a first case of Moco-responsive iSOD.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Mitocondrias/metabolismo , Sulfito-Oxidasa/deficiencia , Sulfito-Oxidasa/metabolismo , Alelos , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/genética , Secuencia de Aminoácidos , Biomarcadores , Catálisis , Activación Enzimática , Fibroblastos/metabolismo , Genotipo , Humanos , Lactante , Recién Nacido , Imagen por Resonancia Magnética , Masculino , Modelos Moleculares , Mutación , Oxidación-Reducción , Conformación Proteica , Proteínas Recombinantes , Índice de Severidad de la Enfermedad , Sulfito-Oxidasa/química , Sulfito-Oxidasa/genética
10.
EMBO Rep ; 20(4)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30886000

RESUMEN

Cardiac dysfunctions dramatically increase with age. Revealing a currently unknown contributor to cardiac ageing, we report the age-dependent, cardiac-specific accumulation of the lysosphingolipid sphinganine (dihydrosphingosine, DHS) as an evolutionarily conserved hallmark of the aged vertebrate heart. Mechanistically, the DHS-derivative sphinganine-1-phosphate (DHS1P) directly inhibits HDAC1, causing an aberrant elevation in histone acetylation and transcription levels, leading to DNA damage. Accordingly, the pharmacological interventions, preventing (i) the accumulation of DHS1P using SPHK2 inhibitors, (ii) the aberrant increase in histone acetylation using histone acetyltransferase (HAT) inhibitors, (iii) the DHS1P-dependent increase in transcription using an RNA polymerase II inhibitor, block DHS-induced DNA damage in human cardiomyocytes. Importantly, an increase in DHS levels in the hearts of healthy young adult mice leads to an impairment in cardiac functionality indicated by a significant reduction in left ventricular fractional shortening and ejection fraction, mimicking the functional deterioration of aged hearts. These molecular and functional defects can be partially prevented in vivo using HAT inhibitors. Together, we report an evolutionarily conserved mechanism by which increased DHS levels drive the decline in cardiac health.


Asunto(s)
Envejecimiento/genética , Envejecimiento/metabolismo , Variación Genética , Inestabilidad Genómica , Miocardio/metabolismo , Esfingolípidos/metabolismo , Animales , Curcumina/química , Curcumina/farmacología , Daño del ADN/efectos de los fármacos , Metabolismo Energético , Epigénesis Genética , Evolución Molecular , Fundulidae , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genómica/métodos , Histona Acetiltransferasas/química , Histona Acetiltransferasas/metabolismo , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Humanos , Modelos Moleculares , Miocitos Cardíacos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Relación Estructura-Actividad , Vertebrados/genética , Vertebrados/metabolismo
11.
Nucleic Acids Res ; 47(6): e32, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30698727

RESUMEN

Long non-coding RNAs (lncRNAs) can act as scaffolds that promote the interaction of proteins, RNA, and DNA. There is increasing evidence of sequence-specific interactions of lncRNAs with DNA via triple-helix (triplex) formation. This process allows lncRNAs to recruit protein complexes to specific genomic regions and regulate gene expression. Here we propose a computational method called Triplex Domain Finder (TDF) to detect triplexes and characterize DNA-binding domains and DNA targets statistically. Case studies showed that this approach can detect the known domains of lncRNAs Fendrr, HOTAIR and MEG3. Moreover, we validated a novel DNA-binding domain in MEG3 by a genome-wide sequencing method. We used TDF to perform a systematic analysis of the triplex-forming potential of lncRNAs relevant to human cardiac differentiation. We demonstrated that the lncRNA with the highest triplex-forming potential, GATA6-AS, forms triple helices in the promoter of genes relevant to cardiac development. Moreover, down-regulation of GATA6-AS impairs GATA6 expression and cardiac development. These data indicate the unique ability of our computational tool to identify novel triplex-forming lncRNAs and their target genes.


Asunto(s)
Biología Computacional/métodos , ADN/metabolismo , ARN Largo no Codificante/química , ARN Largo no Codificante/metabolismo , Algoritmos , Secuencia de Bases , Sitios de Unión/genética , ADN/química , Expresión Génica , Humanos , Conformación de Ácido Nucleico , Unión Proteica , Factores de Transcripción/metabolismo
12.
Cytometry B Clin Cytom ; 96(6): 480-489, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30479054

RESUMEN

BACKGROUND: Human breast milk could be an important stem cell source for the development of newborn and preterm infants, but quantitative data on the stem cell content in breast milk at various gestational stages are needed to determine the clinical value of breast milk as a source of stem cells. Breast milk also contains milk fat globules, lipid droplets of different sizes, debris and dead cells and these components hamper flow cytometry analysis of human breast milk samples. METHODS: Here, we originally used standard protocols for flow cytometry to characterize cell populations in human breast milk but failed to discriminate between cells and noncellular components. We then applied a centrifugation protocol to separate cream and skim milk from the cell-containing pellet and used a novel staining protocol with DRAQ5™ and SYTOX® blue dye as well as antibodies to characterize cells within the pellet fraction. RESULTS: Flow cytometry analysis identified viable DRAQ5™+ /SYTOX® Blue- cells and determined the content of CD11b+ monocytes and TRA-1-81+ putative stem cells in human breast milk samples. CONCLUSIONS: Hence, we developed a novel and reliable flow cytometry based-approach to quantify subpopulation of cells in human breast milk with a high content of milk fat globules, lipid droplets, and particles. This approach will improve the identification and quantification of breast milk cells and allow standardizing the flow cytometry-based evaluation of the stem cell content. © 2018 International Clinical Cytometry Society.


Asunto(s)
Citometría de Flujo/métodos , Leche Humana/citología , Células Madre/citología , Recuento de Células , Células Cultivadas , Colorantes/química , Citometría de Flujo/normas , Glucolípidos/análisis , Glicoproteínas/análisis , Humanos , Gotas Lipídicas/química , Leche Humana/química
13.
Cell Stem Cell ; 24(2): 318-327.e8, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30554961

RESUMEN

Human protein-coding genes are often accompanied by divergently transcribed non-coding RNAs whose functions, especially in cell fate decisions, are poorly understood. Using an hESC-based cardiac differentiation model, we define a class of divergent lncRNAs, termed yin yang lncRNAs (yylncRNAs), that mirror the cell-type-specific expression pattern of their protein-coding counterparts. yylncRNAs are preferentially encoded from the genomic loci of key developmental cell fate regulators. Most yylncRNAs are spliced polyadenylated transcripts showing comparable expression patterns in vivo in mouse and in human embryos. Signifying their developmental function, the key mesoderm specifier BRACHYURY (T) is accompanied by yylncT, which localizes to the active T locus during mesoderm commitment. yylncT binds the de novo DNA methyltransferase DNMT3B, and its transcript is required for activation of the T locus, with yylncT depletion specifically abolishing mesodermal commitment. Collectively, we report a lncRNA-mediated regulatory layer safeguarding embryonic cell fate transitions.


Asunto(s)
Linaje de la Célula/genética , Proteínas Fetales/metabolismo , Mesodermo/metabolismo , Células Madre Pluripotentes/metabolismo , ARN Largo no Codificante/genética , Proteínas de Dominio T Box/metabolismo , Transcripción Genética , Animales , Diferenciación Celular , Línea Celular , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Sitios Genéticos , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ratones , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ADN Metiltransferasa 3B
14.
Methods Mol Biol ; 1694: E1, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30187448

RESUMEN

Correction to: Chapter 26 in: Rubén Alcázar and Antonio F. Tiburcio (eds.), Polyamines: Methods and Protocols, Methods in Molecular Biology, vol. 1694, https://doi.org/10.1007/978-1-4939-7398-9_26.

15.
Cancer Cell ; 33(3): 417-434.e7, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29533783

RESUMEN

Trait-associated loci often map to genomic regions encoding long noncoding RNAs (lncRNAs), but the role of these lncRNAs in disease etiology is largely unexplored. We show that a pair of sense/antisense lncRNA (6p22lncRNAs) encoded by CASC15 and NBAT1 located at the neuroblastoma (NB) risk-associated 6p22.3 locus are tumor suppressors and show reduced expression in high-risk NBs. Loss of functional synergy between 6p22lncRNAs results in an undifferentiated state that is maintained by a gene-regulatory network, including SOX9 located on 17q, a region frequently gained in NB. 6p22lncRNAs regulate SOX9 expression by controlling CHD7 stability via modulating the cellular localization of USP36, encoded by another 17q gene. This regulatory nexus between 6p22.3 and 17q regions may lead to potential NB treatment strategies.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , ARN Largo no Codificante/genética , Factor de Transcripción SOX9/genética , Animales , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Ratones , Neuroblastoma/genética , Neuroblastoma/patología , Ubiquitina Tiolesterasa/genética
16.
Methods Mol Biol ; 1724: 159-166, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29322448

RESUMEN

A substantial proportion of the currently annotated genes in eukaryotes are proposed to function as RNA molecules (>200 bp) with no significant protein coding potential, currently classified as long noncoding RNAs (lncRNA). A distinct subgroup of lncRNAs is circular RNAs (circRNAs), which can be easily identified by unique junction reads, resulting from their biogenesis. CircRNAs are largely cytosolic and thought to either code for micro-peptides or facilitate gene regulation by sequestering microRNAs (miRNAs) or RNA-binding proteins (RBPs) from their targets. Interrogation of the interaction of circRNAs with cellular macromolecular machineries could indicate their mode of action. Here, we detail a sucrose gradient-based method to pinpoint association of a given circRNA (or any transcript of interest) with distinct ribosomal fractions. This method can evaluate the coding potential of candidate circRNAs (or any transcript of interest) and its association with the translation machinery.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Humanos , ARN/genética , ARN Circular , ARN Mensajero/genética , Proteínas de Unión al ARN/genética
17.
Methods Mol Biol ; 1694: 309-323, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29080176

RESUMEN

Polyamines are essential poly-cations with vital functions in all cellular systems. Their levels are controlled by intricate regulatory feedback mechanisms. Abnormally high levels of polyamines have been linked to cancer. A rate-limiting enzyme in the biosynthesis of polyamines in fungi and higher eukaryotes is ornithine-decarboxylase (ODC). Its levels are largely controlled posttranslationally via ubiquitin-independent degradation mediated by ODC antizyme (OAZ). The latter is a critical polyamine sensor in a feedback control mechanism that adjusts cellular polyamine levels. Here, we describe an approach employing quantitative western blot analyses that provides in vivo evidence for cotranslational polyamine-sensing by nascent OAZ in yeast. In addition, we describe an in vitro method to detect polyamine binding by antizyme.


Asunto(s)
Poliaminas/metabolismo , Biosíntesis de Proteínas , Proteínas/genética , ARN Mensajero/genética , Secuencia de Bases , Western Blotting , Unión Proteica , Proteínas/química , Ribosomas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
18.
Nat Commun ; 8(1): 1456, 2017 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-29129916

RESUMEN

While the transcriptional network of human embryonic stem cells (hESCs) has been extensively studied, relatively little is known about how post-transcriptional modulations determine hESC function. RNA-binding proteins play central roles in RNA regulation, including translation and turnover. Here we show that the RNA-binding protein CSDE1 (cold shock domain containing E1) is highly expressed in hESCs to maintain their undifferentiated state and prevent default neural fate. Notably, loss of CSDE1 accelerates neural differentiation and potentiates neurogenesis. Conversely, ectopic expression of CSDE1 impairs neural differentiation. We find that CSDE1 post-transcriptionally modulates core components of multiple regulatory nodes of hESC identity, neuroectoderm commitment and neurogenesis. Among these key pro-neural/neuronal factors, CSDE1 binds fatty acid binding protein 7 (FABP7) and vimentin (VIM) mRNAs, as well as transcripts involved in neuron projection development regulating their stability and translation. Thus, our results uncover CSDE1 as a central post-transcriptional regulator of hESC identity and neurogenesis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteína de Unión a los Ácidos Grasos 7/metabolismo , Células Madre Embrionarias Humanas/citología , Placa Neural/embriología , Neurogénesis/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Vimentina/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Proteína de Unión a los Ácidos Grasos 7/genética , Regulación de la Expresión Génica/genética , Humanos , Ratones , Sistema Nervioso/embriología , Placa Neural/citología , Células-Madre Neurales/citología , Neurogénesis/fisiología , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Proteínas Supresoras de Tumor/genética , Vimentina/genética
20.
Front Cell Dev Biol ; 4: 128, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27882316

RESUMEN

Our conception of the human genome, long focused on the 2% that codes for proteins, has profoundly changed since its first draft assembly in 2001. Since then, an unanticipatedly expansive functionality and convolution has been attributed to the majority of the genome that is transcribed in a cell-type/context-specific manner into transcripts with no apparent protein coding ability. While the majority of these transcripts, currently annotated as long non-coding RNAs (lncRNAs), are functionally uncharacterized, their prominent role in embryonic development and tissue homeostasis, especially in the context of the heart, is emerging. In this review, we summarize and discuss the latest advances in understanding the relevance of lncRNAs in (re)building the heart.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...