Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 148(1-2): 259-72, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22225612

RESUMEN

Identification of the factors critical to the tumor-initiating cell (TIC) state may open new avenues in cancer therapy. Here we show that the metabolic enzyme glycine decarboxylase (GLDC) is critical for TICs in non-small cell lung cancer (NSCLC). TICs from primary NSCLC tumors express high levels of the oncogenic stem cell factor LIN28B and GLDC, which are both required for TIC growth and tumorigenesis. Overexpression of GLDC and other glycine/serine enzymes, but not catalytically inactive GLDC, promotes cellular transformation and tumorigenesis. We found that GLDC induces dramatic changes in glycolysis and glycine/serine metabolism, leading to changes in pyrimidine metabolism to regulate cancer cell proliferation. In the clinic, aberrant activation of GLDC correlates with poorer survival in lung cancer patients, and aberrant GLDC expression is observed in multiple cancer types. This link between glycine metabolism and tumorigenesis may provide novel targets for advancing anticancer therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Transformación Celular Neoplásica , Glicina-Deshidrogenasa (Descarboxilante)/metabolismo , Neoplasias Pulmonares/metabolismo , Secuencia de Aminoácidos , Antígenos CD/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Moléculas de Adhesión Celular Neuronal/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Proteínas Fetales/metabolismo , Glicina/metabolismo , Humanos , Datos de Secuencia Molecular , Neoplasias/enzimología , Neoplasias/genética , Proteínas de Unión al ARN , Alineación de Secuencia , Serina/metabolismo , Thermus thermophilus/enzimología , Trasplante Heterólogo
2.
Cell ; 147(3): 525-38, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-22036562

RESUMEN

The extent of lung regeneration following catastrophic damage and the potential role of adult stem cells in such a process remains obscure. Sublethal infection of mice with an H1N1 influenza virus related to that of the 1918 pandemic triggers massive airway damage followed by apparent regeneration. We show here that p63-expressing stem cells in the bronchiolar epithelium undergo rapid proliferation after infection and radiate to interbronchiolar regions of alveolar ablation. Once there, these cells assemble into discrete, Krt5+ pods and initiate expression of markers typical of alveoli. Gene expression profiles of these pods suggest that they are intermediates in the reconstitution of the alveolar-capillary network eradicated by viral infection. The dynamics of this p63-expressing stem cell in lung regeneration mirrors our parallel finding that defined pedigrees of human distal airway stem cells assemble alveoli-like structures in vitro and suggests new therapeutic avenues to acute and chronic airway disease.


Asunto(s)
Bronquios/citología , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana/patología , Pulmón/fisiología , Alveolos Pulmonares/citología , Síndrome de Dificultad Respiratoria/patología , Células Madre/citología , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Pulmón/citología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Alveolos Pulmonares/virología , Ratas , Factores de Transcripción/genética , Cicatrización de Heridas
3.
Physiol Rev ; 95(1): 245-95, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25540144

RESUMEN

Pluripotent cells in embryos are situated near the apex of the hierarchy of developmental potential. They are capable of generating all cell types of the mammalian body proper. Therefore, they are the exemplar of stem cells. In vivo, pluripotent cells exist transiently and become expended within a few days of their establishment. Yet, when explanted into artificial culture conditions, they can be indefinitely propagated in vitro as pluripotent stem cell lines. A host of transcription factors and regulatory genes are now known to underpin the pluripotent state. Nonetheless, how pluripotent cells are equipped with their vast multilineage differentiation potential remains elusive. Consensus holds that pluripotency transcription factors prevent differentiation by inhibiting the expression of differentiation genes. However, this does not explain the developmental potential of pluripotent cells. We have presented another emergent perspective, namely, that pluripotency factors function as lineage specifiers that enable pluripotent cells to differentiate into specific lineages, therefore endowing pluripotent cells with their multilineage potential. Here we provide a comprehensive overview of the developmental biology, transcription factors, and extrinsic signaling associated with pluripotent cells, and their accompanying subtypes, in vitro heterogeneity and chromatin states. Although much has been learned since the appreciation of mammalian pluripotency in the 1950s and the derivation of embryonic stem cell lines in 1981, we will specifically emphasize what currently remains unclear. However, the view that pluripotency factors capacitate differentiation, recently corroborated by experimental evidence, might perhaps address the long-standing question of how pluripotent cells are endowed with their multilineage differentiation potential.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Mamíferos/embriología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Animales , Desarrollo Embrionario , Humanos
4.
Nat Methods ; 14(12): 1205-1212, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29106405

RESUMEN

Multiple adult tissues are maintained by stem cells of restricted developmental potential which can only form a subset of lineages within the tissue. For instance, the two adult lung epithelial compartments (airways and alveoli) are separately maintained by distinct lineage-restricted stem cells. A challenge has been to obtain multipotent stem cells and/or progenitors that can generate all epithelial cell types of a given tissue. Here we show that mouse Sox9+ multipotent embryonic lung progenitors can be isolated and expanded long term in 3D culture. Cultured Sox9+ progenitors transcriptionally resemble their in vivo counterparts and generate both airway and alveolar cell types in vitro. Sox9+ progenitors that were transplanted into injured adult mouse lungs differentiated into all major airway and alveolar lineages in vivo in a region-appropriate fashion. We propose that a single expandable embryonic lung progenitor population with broader developmental competence may eventually be used as an alternative for region-restricted adult tissue stem cells in regenerative medicine.


Asunto(s)
Pulmón/citología , Células Madre Multipotentes/citología , Factor de Transcripción SOX9/genética , Animales , Diferenciación Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Técnicas de Sustitución del Gen , Pulmón/embriología , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Ratones Transgénicos , Células Madre Multipotentes/metabolismo , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/metabolismo , Factor de Transcripción SOX9/metabolismo , Ingeniería de Tejidos
5.
J Am Chem Soc ; 141(37): 14673-14686, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31436967

RESUMEN

Tumor initiating cells (TIC) are resistant to conventional anticancer therapy and associated with metastasis and relapse in cancer. Although various TIC markers and their antibodies have been proposed, it is limited to the use of antibodies for in vivo imaging or treatment of TIC. In this study, we discovered heme oxygenase 2 (HMOX2) as a novel biomarker for TIC and developed a selective small molecule probe TiNIR (tumor initiating cell probe with near infrared). TiNIR detects and enriches the functionally active TIC in human lung tumors, and through the photoacoustic property, TiNIR also visualizes lung TIC in the patient-derived xenograft (PDX) model. Furthermore, we demonstrate that TiNIR inhibits tumor growth by blocking the function of HMOX2, resulting in significantly increased survival rates of the cancer model mice. The novel therapeutic target HMOX2 and its fluorescent ligand TiNIR will open a new path for the molecular level of lung TIC diagnosis and treatment.


Asunto(s)
Colorantes Fluorescentes/farmacología , Hemo Oxigenasa (Desciclizante)/metabolismo , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/efectos de los fármacos , Espectroscopía Infrarroja Corta/métodos , Animales , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/terapia , Ratones , Células Madre Neoplásicas/enzimología , Tasa de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Nature ; 499(7456): 92-6, 2013 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-23748442

RESUMEN

Stem cells and progenitors in many lineages undergo self-renewing divisions, but the extracellular and intracellular proteins that regulate this process are largely unknown. Glucocorticoids stimulate red blood cell formation by promoting self-renewal of early burst-forming unit-erythroid (BFU-E) progenitors. Here we show that the RNA-binding protein ZFP36L2 is a transcriptional target of the glucocorticoid receptor (GR) in BFU-Es and is required for BFU-E self-renewal. ZFP36L2 is normally downregulated during erythroid differentiation from the BFU-E stage, but its expression is maintained by all tested GR agonists that stimulate BFU-E self-renewal, and the GR binds to several potential enhancer regions of ZFP36L2. Knockdown of ZFP36L2 in cultured BFU-E cells did not affect the rate of cell division but disrupted glucocorticoid-induced BFU-E self-renewal, and knockdown of ZFP36L2 in transplanted erythroid progenitors prevented expansion of erythroid lineage progenitors normally seen following induction of anaemia by phenylhydrazine treatment. ZFP36L2 preferentially binds to messenger RNAs that are induced or maintained at high expression levels during terminal erythroid differentiation and negatively regulates their expression levels. ZFP36L2 therefore functions as part of a molecular switch promoting BFU-E self-renewal and a subsequent increase in the total numbers of colony-forming unit-erythroid (CFU-E) progenitors and erythroid cells that are generated.


Asunto(s)
División Celular , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Tristetraprolina/metabolismo , Animales , Recuento de Células , División Celular/efectos de los fármacos , Linaje de la Célula , Regulación hacia Abajo , Eritropoyesis/genética , Técnicas de Silenciamiento del Gen , Glucocorticoides/farmacología , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/metabolismo , Estrés Fisiológico , Tristetraprolina/deficiencia , Tristetraprolina/genética
7.
Genes Dev ; 25(2): 119-24, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21196494

RESUMEN

Using RNA-seq technology, we found that the majority of microRNAs (miRNAs) present in CFU-E erythroid progenitors are down-regulated during terminal erythroid differentiation. Of the developmentally down-regulated miRNAs, ectopic overexpression of miR-191 blocks erythroid enucleation but has minor effects on proliferation and differentiation. We identified two erythroid-enriched and developmentally up-regulated genes, Riok3 and Mxi1, as direct targets of miR-191. Knockdown of either Riok3 or Mxi1 blocks enucleation, and either physiological overexpression of miR-191 or knockdown of Riok3 or Mxi1 blocks chromatin condensation. Thus, down-regulation of miR-191 is essential for erythroid chromatin condensation and enucleation by allowing up-regulation of Riok3 and Mxi1.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación hacia Abajo , Eritroblastos/citología , MicroARNs/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Línea Celular , Núcleo Celular/metabolismo , Células Cultivadas , Eritroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Ratones , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba
8.
J Immunol ; 196(12): 4935-46, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27183569

RESUMEN

T cells undergo homeostatic expansion and acquire an activated phenotype in lymphopenic microenvironments. Restoration of normal lymphocyte numbers typically re-establishes normal homeostasis, and proinflammatory cytokine production returns to baseline. Mice deficient in guanine nucleotide exchange factor RasGRP1 exhibit dysregulated homeostatic expansion, which manifests as lymphoproliferative disease with autoantibody production. Our previous work revealed that autoreactive B cells lacking RasGRP1 break tolerance early during development, as well as during germinal center responses, suggesting that T cell-independent and T cell-dependent mechanisms are responsible. Examination of whether a particular T cell subset is involved in the breach of B cell tolerance revealed increased Th17 cells in Rasgrp1-deficient mice relative to control mice. Rasgrp1-deficient mice lacking IL-17R had fewer germinal centers, and germinal centers that formed contained fewer autoreactive B cells, suggesting that IL-17 signaling is required for a break in B cell tolerance in germinal centers. Interestingly, a fraction of Th17 cells from Rasgrp1-deficient mice were CXCR5(+) and upregulated levels of CD278 coordinate with their appearance in germinal centers, all attributes of T follicular helper cells (Tfh17). To determine whether CD278-CD275 interactions were required for the development of Tfh17 cells and for autoantibody, Rasgrp1-deficient mice were crossed with CD275-deficient mice. Surprisingly, mice deficient in RasGRP1 and CD275 formed Tfh17 cells and germinal centers and produced similar titers of autoantibodies as mice deficient in only RasGRP1. Therefore, these studies suggest that requirements for Tfh cell development change in lymphopenia-associated autoimmune settings.


Asunto(s)
Autoinmunidad , Centro Germinal/inmunología , Ligando Coestimulador de Linfocitos T Inducibles/inmunología , Interleucina-17/inmunología , Linfopenia/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Linfocitos B/inmunología , Centro Germinal/citología , Factores de Intercambio de Guanina Nucleótido/deficiencia , Homeostasis , Tolerancia Inmunológica/genética , Ligando Coestimulador de Linfocitos T Inducibles/deficiencia , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Interleucina-17/biosíntesis , Ratones , Receptores CXCR5/genética , Receptores de Interleucina-17/deficiencia , Transducción de Señal , Células Th17/inmunología
9.
Angew Chem Int Ed Engl ; 57(11): 2851-2854, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29377425

RESUMEN

Tumor initiating cells (TICs) have been implicated in clinical relapse and metastasis of a variety of epithelial cancers, including lung cancer. While efforts toward the development of specific probes for TIC detection and targeting are ongoing, a universal TIC probe has yet to be developed. We report the first TIC-specific fluorescent chemical probe, TiY, with identification of the molecular target as vimentin, a marker for epithelial-to-mesenchymal transition (EMT). TiY selectively stains TICs over differentiated tumor cells or normal cells, and facilitates the visualization and enrichment of functionally active TICs from patient tumors. At high concentration, TiY also shows anti-TIC activity with low toxicity to non-TICs. With the unexplored target vimentin, TiY shows potential as a first universal probe for TIC detection in different cancers.


Asunto(s)
Colorantes Fluorescentes/química , Células Madre Neoplásicas/patología , Bibliotecas de Moléculas Pequeñas/química , Vimentina/análisis , Animales , Biomarcadores de Tumor/análisis , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Humanos , Neoplasias Pulmonares/patología , Ratones
10.
Stem Cells ; 34(2): 277-87, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26676652

RESUMEN

In recent years, the highly conserved promyelocytic leukemia zinc finger (PLZF, also known as ZBTB16, ZNF145) has attracted attention as a multifunctional transcription factor involved in major biological processes during development. As a transcription factor, PLZF shows tight regulation in its cell-type-specific and stage-specific expression patterns. Emerging evidence shows that PLZF regulates the balance of self-renewal and differentiation in stem cells. However, the gene regulatory network of PLZF is only beginning to be understood. In this review, we discuss the diverse functions of PLZF, in particular its role in self-renewal versus differentiation of stem cells. We also discuss the current state of knowledge on the gene regulatory network of PLZF, in conjunction with its upstream factors, post-translational modifications and binding cofactors for multiprotein complexes. This review aims to provide the reader with an in-depth understanding of the molecular mechanisms underlying PLZF and the potential applications in tissue regeneration.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Regulación de la Expresión Génica/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Células Madre/metabolismo , Animales , Humanos , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Células Madre/citología
11.
Genes Dev ; 23(7): 862-76, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19293287

RESUMEN

The p53 transcription factor is a key tumor suppressor and a central regulator of the stress response. To ensure a robust and precise response to cellular signals, p53 gene expression must be tightly regulated from the transcriptional to the post-translational levels. Computational predictions suggest that several microRNAs are involved in the post-transcriptional regulation of p53. Here we demonstrate that miR-125b, a brain-enriched microRNA, is a bona fide negative regulator of p53 in both zebrafish and humans. miR-125b-mediated down-regulation of p53 is strictly dependent on the binding of miR-125b to a microRNA response element in the 3' untranslated region of p53 mRNA. Overexpression of miR-125b represses the endogenous level of p53 protein and suppresses apoptosis in human neuroblastoma cells and human lung fibroblast cells. In contrast, knockdown of miR-125b elevates the level of p53 protein and induces apoptosis in human lung fibroblasts and in the zebrafish brain. This phenotype can be rescued significantly by either an ablation of endogenous p53 function or ectopic expression of miR-125b in zebrafish. Interestingly, miR-125b is down-regulated when zebrafish embryos are treated with gamma-irradiation or camptothecin, corresponding to the rapid increase in p53 protein in response to DNA damage. Ectopic expression of miR-125b suppresses the increase of p53 and stress-induced apoptosis. Together, our study demonstrates that miR-125b is an important negative regulator of p53 and p53-induced apoptosis during development and during the stress response.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Regulación de la Expresión Génica , Genes p53/fisiología , MicroARNs/metabolismo , Regiones no Traducidas 3'/metabolismo , Animales , Apoptosis/fisiología , Secuencia de Bases , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Embrión no Mamífero/metabolismo , Prueba de Complementación Genética , Humanos , Datos de Secuencia Molecular , Unión Proteica , Estrés Fisiológico/fisiología , Pez Cebra
12.
Genes Dev ; 23(21): 2507-20, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19884257

RESUMEN

The histone H3 Lys 9 (H3K9) methyltransferase Eset is an epigenetic regulator critical for the development of the inner cell mass (ICM). Although ICM-derived embryonic stem (ES) cells are normally unable to contribute to the trophectoderm (TE) in blastocysts, we find that depletion of Eset by shRNAs leads to differentiation with the formation of trophoblast-like cells and induction of trophoblast-associated gene expression. Using chromatin immmunoprecipitation (ChIP) and sequencing (ChIP-seq) analyses, we identified Eset target genes with Eset-dependent H3K9 trimethylation. We confirmed that genes that are preferentially expressed in the TE (Tcfap2a and Cdx2) are bound and repressed by Eset. Single-cell PCR analysis shows that the expression of Cdx2 and Tcfap2a is also induced in Eset-depleted morula cells. Importantly, Eset-depleted cells can incorporate into the TE of a blastocyst and, subsequently, placental tissues. Coimmunoprecipitation and ChIP assays further demonstrate that Eset interacts with Oct4, which in turn recruits Eset to silence these trophoblast-associated genes. Our results suggest that Eset restricts the extraembryonic trophoblast lineage potential of pluripotent cells and links an epigenetic regulator to key cell fate decision through a pluripotency factor.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias/citología , Metiltransferasas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteína Metiltransferasas/metabolismo , Trofoblastos/citología , Trofoblastos/metabolismo , Animales , Inmunoprecipitación de Cromatina , Regulación del Desarrollo de la Expresión Génica , Genoma/fisiología , N-Metiltransferasa de Histona-Lisina , Proteínas de Homeodominio/metabolismo , Ratones , Mórula/citología , Factor de Transcripción AP-2/metabolismo
13.
Nature ; 521(7552): 299-300, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25993958
14.
Nature ; 463(7284): 1096-100, 2010 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-20139965

RESUMEN

Induced pluripotent stem (iPS) cells can be obtained by the introduction of defined factors into somatic cells. The combination of Oct4 (also known as Pou5f1), Sox2 and Klf4 (which we term OSK) constitutes the minimal requirement for generating iPS cells from mouse embryonic fibroblasts. These cells are thought to resemble embryonic stem cells (ESCs) on the basis of global gene expression analyses; however, few studies have tested the ability and efficiency of iPS cells to contribute to chimaerism, colonization of germ tissues, and most importantly, germ-line transmission and live birth from iPS cells produced by tetraploid complementation. Using genomic analyses of ESC genes that have roles in pluripotency and fusion-mediated somatic cell reprogramming, here we show that the transcription factor Tbx3 significantly improves the quality of iPS cells. iPS cells generated with OSK and Tbx3 (OSKT) are superior in both germ-cell contribution to the gonads and germ-line transmission frequency. However, global gene expression profiling could not distinguish between OSK and OSKT iPS cells. Genome-wide chromatin immunoprecipitation sequencing analysis of Tbx3-binding sites in ESCs suggests that Tbx3 regulates pluripotency-associated and reprogramming factors, in addition to sharing many common downstream regulatory targets with Oct4, Sox2, Nanog and Smad1. This study underscores the intrinsic qualitative differences between iPS cells generated by different methods, and highlights the need to rigorously characterize iPS cells beyond in vitro studies.


Asunto(s)
Quimera/metabolismo , Células Germinativas/citología , Células Germinativas/metabolismo , Gónadas/citología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas de Dominio T Box/metabolismo , Animales , Fusión Celular , Reprogramación Celular , Quimera/embriología , Inmunoprecipitación de Cromatina , Embrión de Mamíferos/citología , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Proteína Smad1/metabolismo , Proteínas de Dominio T Box/genética , Transcripción Genética/genética , Transducción Genética
15.
Nature ; 468(7321): 316-20, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-20953172

RESUMEN

The derivation of human ES cells (hESCs) from human blastocysts represents one of the milestones in stem cell biology. The full potential of hESCs in research and clinical applications requires a detailed understanding of the genetic network that governs the unique properties of hESCs. Here, we report a genome-wide RNA interference screen to identify genes which regulate self-renewal and pluripotency properties in hESCs. Interestingly, functionally distinct complexes involved in transcriptional regulation and chromatin remodelling are among the factors identified in the screen. To understand the roles of these potential regulators of hESCs, we studied transcription factor PRDM14 to gain new insights into its functional roles in the regulation of pluripotency. We showed that PRDM14 regulates directly the expression of key pluripotency gene POU5F1 through its proximal enhancer. Genome-wide location profiling experiments revealed that PRDM14 colocalized extensively with other key transcription factors such as OCT4, NANOG and SOX2, indicating that PRDM14 is integrated into the core transcriptional regulatory network. More importantly, in a gain-of-function assay, we showed that PRDM14 is able to enhance the efficiency of reprogramming of human fibroblasts in conjunction with OCT4, SOX2 and KLF4. Altogether, our study uncovers a wealth of novel hESC regulators wherein PRDM14 exemplifies a key transcription factor required for the maintenance of hESC identity and the reacquisition of pluripotency in human somatic cells.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Genoma Humano/genética , Interferencia de ARN , Proteínas Represoras/metabolismo , Animales , Secuencia de Bases , Línea Celular , Reprogramación Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas de Unión al ARN , Proteínas Represoras/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción
16.
Nucleic Acids Res ; 42(12): 7997-8007, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24860167

RESUMEN

LIN28 function is fundamental to the activity and behavior of human embryonic stem cells (hESCs) and induced pluripotent stem cells. Its main roles in these cell types are the regulation of translational efficiency and let-7 miRNA maturation. However, LIN28-associated mRNA cargo shifting and resultant regulation of translational efficiency upon the initiation of differentiation remain unknown. An RNA-immunoprecipitation and microarray analysis protocol, eRIP, that has high specificity and sensitivity was developed to test endogenous LIN28-associated mRNA cargo shifting. A combined eRIP and polysome analysis of early stage differentiation of hESCs with two distinct differentiation cues revealed close similarities between the dynamics of LIN28 association and translational modulation of genes involved in the Wnt signaling, cell cycle, RNA metabolism and proteasomal pathways. Our data demonstrate that change in translational efficiency is a major contributor to early stages of differentiation of hESCs, in which LIN28 plays a central role. This implies that eRIP analysis of LIN28-associated RNA cargoes may be used for rapid functional quality control of pluripotent stem cells under manufacture for therapeutic applications.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Células Cultivadas , Células Madre Embrionarias/citología , Humanos , Polirribosomas/metabolismo
17.
J Immunol ; 191(7): 3605-13, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23997211

RESUMEN

Lymphopenic hosts offer propitious microenvironments for expansion of autoreactive B and T cells. Despite this, many lymphopenic hosts do not develop autoimmune disease, suggesting that additional factors are required for breaching self-tolerance in the setting of lymphopenia. Mice deficient in guanine nucleotide exchange factor Rasgrp1 develop a lymphoproliferative disorder with features of human systemic lupus erythematosus. Early in life, Rasgrp1-deficient mice have normal B cell numbers but are T lymphopenic, leading to defective homeostatic expansion of CD4 T cells. To investigate whether B cell-intrinsic mechanisms also contribute to autoimmunity, Rasgrp1-deficient mice were bred to mice containing a knockin autoreactive BCR transgene (564Igi), thereby allowing the fate of autoreactive B cells to be assessed. During B cell development, the frequency of receptor-edited 564Igi B cells was reduced in Rasrp1-deficient mice compared with Rasgrp1-sufficient littermate control mice, suggesting that tolerance was impaired. In addition, the number of 564Igi transitional B cells was increased in Rasgrp1-deficient mice compared with control mice. Immature 564Igi B cells in bone marrow and spleen lacking RasGRP1 expressed lower levels of Bim mRNA and protein, suggesting that autoreactive B cells elude clonal deletion during development. Concomitant with increased serum autoantibodies, Rasgrp1-deficient mice developed spontaneous germinal centers at 8-10 wk of age. The frequency and number of 564Igi B cells within these germinal centers were significantly increased in Rasgrp1-deficient mice relative to control mice. Taken together, these studies suggest that autoreactive B cells lacking Rasgrp1 break central and peripheral tolerance through both T cell-independent and -dependent mechanisms.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Tolerancia Inmunológica/genética , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Autoinmunidad/genética , Autoinmunidad/inmunología , Proteína 11 Similar a Bcl2 , Médula Ósea/inmunología , Médula Ósea/metabolismo , Centro Germinal/inmunología , Centro Germinal/metabolismo , Factores de Intercambio de Guanina Nucleótido/deficiencia , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Bazo/inmunología , Bazo/metabolismo , Receptores Toll-Like/metabolismo
18.
J Immunol ; 191(6): 3192-9, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23935193

RESUMEN

Engraftment of human CD34⁺ hematopoietic stem/progenitor cells into immunodeficient mice leads to robust reconstitution of human T and B cells but not monocytes and macrophages. To identify the cause underlying the poor monocyte and macrophage reconstitution, we analyzed human myeloid cell development in humanized mice and found that it was blocked at the promonocyte stage in the bone marrow. Expression of human M-CSF or GM-CSF by hydrodynamic injection of cytokine-encoding plasmid completely abolished the accumulation of promonocytes in the bone marrow. M-CSF promoted the development of mature monocytes and tissue-resident macrophages whereas GM-CSF did not. Moreover, correlating with an increased human macrophages at the sites of infection, M-CSF-treated humanized mice exhibited an enhanced protection against influenza virus and Mycobacterium infection. Our study identifies the precise stage at which human monocyte/macrophage development is blocked in humanized mice and reveals overlapping and distinct functions of M-CSF and GM-CSF in human monocyte and macrophage development. The improved reconstitution and functionality of monocytes/macrophages in the humanized mice following M-CSF expression provide a superior in vivo system to investigate the role of macrophages in physiological and pathological processes.


Asunto(s)
Diferenciación Celular/inmunología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/citología , Células Precursoras de Monocitos y Macrófagos/citología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Diferenciación Celular/efectos de los fármacos , Separación Celular , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/inmunología , Humanos , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Precursoras de Monocitos y Macrófagos/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Nat Genet ; 38(4): 431-40, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16518401

RESUMEN

Oct4 and Nanog are transcription factors required to maintain the pluripotency and self-renewal of embryonic stem (ES) cells. Using the chromatin immunoprecipitation paired-end ditags method, we mapped the binding sites of these factors in the mouse ES cell genome. We identified 1,083 and 3,006 high-confidence binding sites for Oct4 and Nanog, respectively. Comparative location analyses indicated that Oct4 and Nanog overlap substantially in their targets, and they are bound to genes in different configurations. Using de novo motif discovery algorithms, we defined the cis-acting elements mediating their respective binding to genomic sites. By integrating RNA interference-mediated depletion of Oct4 and Nanog with microarray expression profiling, we demonstrated that these factors can activate or suppress transcription. We further showed that common core downstream targets are important to keep ES cells from differentiating. The emerging picture is one in which Oct4 and Nanog control a cascade of pathways that are intricately connected to govern pluripotency, self-renewal, genome surveillance and cell fate determination.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Embrión de Mamíferos/citología , Proteínas de Homeodominio/fisiología , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Células Madre/citología , Transcripción Genética/fisiología , Animales , Embrión de Mamíferos/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Ratones , Proteína Homeótica Nanog , Interferencia de ARN , Células Madre/metabolismo
20.
J Mol Cell Cardiol ; 67: 12-25, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24370890

RESUMEN

The ability of human pluripotent stem cells (hPSCs) to differentiate into any cell type of the three germ layers makes them a very promising cell source for multiple purposes, including regenerative medicine, drug discovery, and as a model to study disease mechanisms and progression. One of the first specialized cell types to be generated from hPSC was cardiomyocytes (CM), and differentiation protocols have evolved over the years and now allow for robust and large-scale production of hPSC-CM. Still, scientists are struggling to achieve the same, mainly ventricular, phenotype of the hPSC-CM in vitro as their adult counterpart in vivo. In vitro generated cardiomyocytes are generally described as fetal-like rather than adult. In this review, we compare the in vivo development of cardiomyocytes to the in vitro differentiation of hPSC into CM with focus on electrophysiology, structure and contractility. Furthermore, known epigenetic changes underlying the differences between adult human CM and CM differentiated from pluripotent stem cells are described. This should provide the reader with an extensive overview of the current status of human stem cell-derived cardiomyocyte phenotype and function. Additionally, the reader will gain insight into the underlying signaling pathways and mechanisms responsible for cardiomyocyte development.


Asunto(s)
Diferenciación Celular , Fenómenos Electrofisiológicos , Miocitos Cardíacos/citología , Técnicas de Cultivo , Epigenómica , Corazón/embriología , Corazón/crecimiento & desarrollo , Humanos , Células Madre Pluripotentes/citología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA