Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Development ; 147(12)2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32487737

RESUMEN

The intrinsic mechanisms that link extracellular signalling to the onset of neural differentiation are not well understood. In pluripotent mouse cells, BMP blocks entry into the neural lineage via transcriptional upregulation of inhibitor of differentiation (Id) factors. We have previously identified the major binding partner of Id proteins in pluripotent cells as the basic helix-loop-helix (bHLH) transcription factor (TF) E2A. Id1 can prevent E2A from forming heterodimers with bHLH TFs or from forming homodimers. Here, we show that overexpression of a forced E2A homodimer is sufficient to drive robust neural commitment in pluripotent cells, even under non-permissive conditions. Conversely, we find that E2A null cells display a defect in their neural differentiation capacity. E2A acts as an upstream activator of neural lineage genes, including Sox1 and Foxd4, and as a repressor of Nodal signalling. Our results suggest a crucial role for E2A in establishing neural lineage commitment in pluripotent cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Neuronas/metabolismo , Regiones no Traducidas 3' , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sistemas CRISPR-Cas/genética , Linaje de la Célula , Autorrenovación de las Células , Dimerización , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Neuronas/citología , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , ARN Guía de Kinetoplastida/metabolismo , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transcriptoma , Regulación hacia Arriba
2.
Nat Commun ; 10(1): 967, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30814500

RESUMEN

The stem cell pluripotency factor Oct4 serves a critical protective role during atherosclerotic plaque development by promoting smooth muscle cell (SMC) investment. Here, we show using Myh11-CreERT2 lineage-tracing with inducible SMC and pericyte (SMC-P) knockout of Oct4 that Oct4 regulates perivascular cell migration and recruitment during angiogenesis. Knockout of Oct4 in perivascular cells significantly impairs perivascular cell migration, increases perivascular cell death, delays endothelial cell migration, and promotes vascular leakage following corneal angiogenic stimulus. Knockout of Oct4 in perivascular cells also impairs perfusion recovery and decreases angiogenesis following hindlimb ischemia. Transcriptomic analyses demonstrate that expression of the migratory gene Slit3 is reduced following loss of Oct4 in cultured SMCs, and in Oct4-deficient perivascular cells in ischemic hindlimb muscle. Together, these results provide evidence that Oct4 plays an essential role within perivascular cells in injury- and hypoxia-induced angiogenesis.


Asunto(s)
Neovascularización Fisiológica , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Células Madre Pluripotentes/metabolismo , Animales , Muerte Celular , Linaje de la Célula , Movimiento Celular , Células Cultivadas , Neovascularización de la Córnea/metabolismo , Neovascularización de la Córnea/patología , Femenino , Miembro Posterior , Isquemia/metabolismo , Isquemia/patología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/metabolismo , Neovascularización Patológica , Factor 3 de Transcripción de Unión a Octámeros/genética , Pericitos/metabolismo , Pericitos/patología , Células Madre Pluripotentes/patología
3.
Med Sci Monit ; 25: 1214-1219, 2019 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-30763293

RESUMEN

BACKGROUND POU5F1B, serving as a carcinogen, participates in radiosensitivity of several tumors. However, in esophageal cancer, its potential mechanism and function in regulating radiosensitivity remain unclear. MATERIAL AND METHODS The expression level of POU5F1B was detected in plasma of esophageal tumor patients and cancer cell lines. The effect of POU5F1B knockdown on cell proliferation and colony formation was determined using CCK-8 assay and colony formation assay. Cell apoptosis rate was detected by flow cytometry. RESULTS POU5F1B expression level declined after radiotherapy in the plasma of esophageal cancer patients (p=0.025). Compared with HEEPIC, the level of POU5F1B was upregulated in ECA109 (p<0.01), ECA9706 (p<0.01), KYSE410 (p<0.01), and KYSE510 (p=0.036). The silencing of POU5F1B played a role in inhibiting colony formation. After radiotherapy, the apoptosis rates in the ECA109 with 4Gy si-POU5F1B group and 4Gy si-NC group were 39.1±0.1% and 35.3±0.1%, respectively (p=0.0193). The rate was 21.00±0.1 and 29.1±0.1% (p<0.0072) in the si-NC group and si-POU5F1B group, respectively. For proliferation rate, 4Gy si-POU5F1B ECA109 performed better than 4Gy si-NC. CONCLUSIONS Radiotherapy contributed to the decline in the expression level of POU5F1B in plasma, which was upregulated in ECA109, ECA9706, KYSE410, and KYSE510, but not in HEEPIC. The knockdown of POU5F1B increased the radiosensitivity of esophageal cancer cell lines.


Asunto(s)
Neoplasias Esofágicas/radioterapia , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Regulación hacia Abajo/efectos de la radiación , Neoplasias Esofágicas/sangre , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Técnicas de Silenciamiento del Gen/métodos , Genes myc , Humanos , Factor 3 de Transcripción de Unión a Octámeros/sangre , Factor 3 de Transcripción de Unión a Octámeros/genética , ARN Largo no Codificante/genética , Tolerancia a Radiación , Regulación hacia Arriba/efectos de la radiación
4.
Nature ; 550(7674): 67-73, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28953884

RESUMEN

Despite their fundamental biological and clinical importance, the molecular mechanisms that regulate the first cell fate decisions in the human embryo are not well understood. Here we use CRISPR-Cas9-mediated genome editing to investigate the function of the pluripotency transcription factor OCT4 during human embryogenesis. We identified an efficient OCT4-targeting guide RNA using an inducible human embryonic stem cell-based system and microinjection of mouse zygotes. Using these refined methods, we efficiently and specifically targeted the gene encoding OCT4 (POU5F1) in diploid human zygotes and found that blastocyst development was compromised. Transcriptomics analysis revealed that, in POU5F1-null cells, gene expression was downregulated not only for extra-embryonic trophectoderm genes, such as CDX2, but also for regulators of the pluripotent epiblast, including NANOG. By contrast, Pou5f1-null mouse embryos maintained the expression of orthologous genes, and blastocyst development was established, but maintenance was compromised. We conclude that CRISPR-Cas9-mediated genome editing is a powerful method for investigating gene function in the context of human development.


Asunto(s)
Desarrollo Embrionario/genética , Edición Génica , Regulación del Desarrollo de la Expresión Génica , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Animales , Blastocisto/metabolismo , Sistemas CRISPR-Cas/genética , Linaje de la Célula , Ectodermo/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Estratos Germinativos/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Masculino , Ratones , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Especificidad por Sustrato , Cigoto/metabolismo
5.
Sci Rep ; 7: 46312, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28406185

RESUMEN

Oct4A is a master regulator of self-renewal and pluripotency in embryonic stem cells. It is a well-established marker for cancer stem cell (CSC) in malignancies. Recently, using a loss of function studies, we have demonstrated key roles for Oct4A in tumor cell survival, metastasis and chemoresistance in in vitro and in vivo models of ovarian cancer. In an effort to understand the regulatory role of Oct4A in tumor biology, we employed the use of an ovarian cancer shRNA Oct4A knockdown cell line (HEY Oct4A KD) and a global mass spectrometry (MS)-based proteomic analysis to investigate novel biological targets of Oct4A in HEY samples (cell lysates, secretomes and mouse tumor xenografts). Based on significant differential expression, pathway and protein network analyses, and comprehensive literature search we identified key proteins involved with biologically relevant functions of Oct4A in tumor biology. Across all preparations of HEY Oct4A KD samples significant alterations in protein networks associated with cytoskeleton, extracellular matrix (ECM), proliferation, adhesion, metabolism, epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs) and drug resistance was observed. This comprehensive proteomics study for the first time presents the Oct4A associated proteome and expands our understanding on the biological role of this stem cell regulator in carcinomas.


Asunto(s)
Reprogramación Celular/genética , Células Madre Neoplásicas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Animales , Biomarcadores , Línea Celular Tumoral , Biología Computacional/métodos , Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Ováricas/patología , Proteoma/metabolismo , Proteómica/métodos , Reproducibilidad de los Resultados , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Neuropsychopharmacology ; 42(3): 757-765, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27604564

RESUMEN

The mechanisms by which stressful life events increase the risk of relapse in recovering cocaine addicts are not well understood. We previously reported that stress, via elevated corticosterone, potentiates cocaine-primed reinstatement of cocaine seeking following self-administration in rats and that this potentiation appears to involve corticosterone-induced blockade of dopamine clearance via the organic cation transporter 3 (OCT3). In the present study, we use a conditioned place preference/reinstatement paradigm in mice to directly test the hypothesis that corticosterone potentiates cocaine-primed reinstatement by blockade of OCT3. Consistent with our findings following self-administration in rats, pretreatment of male C57/BL6 mice with corticosterone (using a dose that reproduced stress-level plasma concentrations) potentiated cocaine-primed reinstatement of extinguished cocaine-induced conditioned place preference. Corticosterone failed to re-establish extinguished preference alone but produced a leftward shift in the dose-response curve for cocaine-primed reinstatement. A similar potentiating effect was observed upon pretreatment of mice with the non-glucocorticoid OCT3 blocker, normetanephrine. To determine the role of OCT3 blockade in these effects, we examined the abilities of corticosterone and normetanephrine to potentiate cocaine-primed reinstatement in OCT3-deficient and wild-type mice. Conditioned place preference, extinction and reinstatement of extinguished preference in response to low-dose cocaine administration did not differ between genotypes. However, corticosterone and normetanephrine failed to potentiate cocaine-primed reinstatement in OCT3-deficient mice. Together, these data provide the first direct evidence that the interaction of corticosterone with OCT3 mediates corticosterone effects on drug-seeking behavior and establish OCT3 function as an important determinant of susceptibility to cocaine use.


Asunto(s)
Cocaína/farmacología , Condicionamiento Clásico/efectos de los fármacos , Corticosterona/farmacología , Inhibidores de Captación de Dopamina/farmacología , Normetanefrina/farmacología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Animales , Cocaína/administración & dosificación , Corticosterona/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Normetanefrina/administración & dosificación , Factor 3 de Transcripción de Unión a Octámeros/antagonistas & inhibidores , Factor 3 de Transcripción de Unión a Octámeros/deficiencia
7.
Nature ; 540(7631): 114-118, 2016 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-27905446

RESUMEN

Germ-cell tumours (GCTs) are derived from germ cells and occur most frequently in the testes. GCTs are histologically heterogeneous and distinctly curable with chemotherapy. Gains of chromosome arm 12p and aneuploidy are nearly universal in GCTs, but specific somatic genomic features driving tumour initiation, chemosensitivity and progression are incompletely characterized. Here, using clinical whole-exome and transcriptome sequencing of precursor, primary (testicular and mediastinal) and chemoresistant metastatic human GCTs, we show that the primary somatic feature of GCTs is highly recurrent chromosome arm level amplifications and reciprocal deletions (reciprocal loss of heterozygosity), variations that are significantly enriched in GCTs compared to 19 other cancer types. These tumours also acquire KRAS mutations during the development from precursor to primary disease, and primary testicular GCTs (TGCTs) are uniformly wild type for TP53. In addition, by functional measurement of apoptotic signalling (BH3 profiling) of fresh tumour and adjacent tissue, we find that primary TGCTs have high mitochondrial priming that facilitates chemotherapy-induced apoptosis. Finally, by phylogenetic analysis of serial TGCTs that emerge with chemotherapy resistance, we show how TGCTs gain additional reciprocal loss of heterozygosity and that this is associated with loss of pluripotency markers (NANOG and POU5F1) in chemoresistant teratomas or transformed carcinomas. Our results demonstrate the distinct genomic features underlying the origins of this disease and associated with the chemosensitivity phenotype, as well as the rare progression to chemoresistance. These results identify the convergence of cancer genomics, mitochondrial priming and GCT evolution, and may provide insights into chemosensitivity and resistance in other cancers.


Asunto(s)
Resistencia a Antineoplásicos , Genoma Humano/genética , Neoplasias de Células Germinales y Embrionarias/tratamiento farmacológico , Neoplasias de Células Germinales y Embrionarias/genética , Apoptosis , Progresión de la Enfermedad , Evolución Molecular , Exoma/genética , Genómica , Humanos , Pérdida de Heterocigocidad , Masculino , Mitocondrias/metabolismo , Mutación , Proteína Homeótica Nanog/deficiencia , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Neoplasias de Células Germinales y Embrionarias/metabolismo , Neoplasias de Células Germinales y Embrionarias/patología , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Filogenia , Proteínas Proto-Oncogénicas p21(ras)/genética , Teratoma/genética , Neoplasias Testiculares/tratamiento farmacológico , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patología , Transcriptoma/genética , Proteína p53 Supresora de Tumor/genética
8.
Nat Commun ; 6: 6810, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25857206

RESUMEN

The actions of transcription factors, chromatin modifiers and noncoding RNAs are crucial for the programming of cell states. Although the importance of various epigenetic machineries for controlling pluripotency of embryonic stem (ES) cells has been previously studied, how chromatin modifiers cooperate with specific transcription factors still remains largely elusive. Here, we find that Pontin chromatin remodelling factor plays an essential role as a coactivator for Oct4 for maintenance of pluripotency in mouse ES cells. Genome-wide analyses reveal that Pontin and Oct4 share a substantial set of target genes involved in ES cell maintenance. Intriguingly, we find that the Oct4-dependent coactivator function of Pontin extends to the transcription of large intergenic noncoding RNAs (lincRNAs) and in particular linc1253, a lineage programme repressing lincRNA, is a Pontin-dependent Oct4 target lincRNA. Together, our findings demonstrate that the Oct4-Pontin module plays critical roles in the regulation of genes involved in ES cell fate determination.


Asunto(s)
ADN Helicasas/genética , Epigénesis Genética , Células Madre Embrionarias de Ratones/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , ARN Largo no Codificante/genética , Animales , Diferenciación Celular , Cromatina/química , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , ADN Helicasas/deficiencia , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/citología , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Receptores Patched , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , ARN Largo no Codificante/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Tropomiosina/genética , Tropomiosina/metabolismo
9.
Nat Cell Biol ; 15(9): 1089-97, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23934214

RESUMEN

Oct4A is a core component of the regulatory network of pluripotent cells, and by itself can reprogram neural stem cells into pluripotent cells in mice and humans. However, its role in defining totipotency and inducing pluripotency during embryonic development is still unclear. We genetically eliminated maternal Oct4A using a Cre/loxP approach in mouse and found that the establishment of totipotency was not affected, as shown by the generation of live pups. After complete inactivation of both maternal and zygotic Oct4A expression, the embryos still formed Oct4-GFP- and Nanog-expressing inner cell masses, albeit non-pluripotent, indicating that Oct4A is not a determinant for the pluripotent cell lineage separation. Interestingly, Oct4A-deficient oocytes were able to reprogram fibroblasts into pluripotent cells. Our results clearly demonstrate that, in contrast to its role in the maintenance of pluripotency, maternal Oct4A is not crucial for either the establishment of totipotency in embryos, or the induction of pluripotency in somatic cells using oocytes.


Asunto(s)
Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factor 3 de Transcripción de Unión a Octámeros/genética , Oocitos/metabolismo , Células Madre Totipotentes/metabolismo , Animales , Linaje de la Célula/genética , Células Cultivadas , Reprogramación Celular/genética , Embrión de Mamíferos , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Fibroblastos/citología , Genes Reporteros , Proteínas Fluorescentes Verdes , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Noqueados , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Oocitos/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Embarazo , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Transducción de Señal , Células Madre Totipotentes/citología
10.
PLoS One ; 7(8): e43628, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22928007

RESUMEN

The cancer stem cell (CSC) model posits the presence of a small number of CSCs in the heterogeneous cancer cell population that are ultimately responsible for tumor initiation, as well as cancer recurrence and metastasis. CSCs have been isolated from a variety of human cancers and are able to generate a hierarchical and heterogeneous cancer cell population. CSCs are also resistant to conventional chemo- and radio-therapies. Here we report that ionizing radiation can induce stem cell-like properties in heterogeneous cancer cells. Exposure of non-stem cancer cells to ionizing radiation enhanced spherogenesis, and this was accompanied by upregulation of the pluripotency genes Sox2 and Oct3/4. Knockdown of Sox2 or Oct3/4 inhibited radiation-induced spherogenesis and increased cellular sensitivity to radiation. These data demonstrate that ionizing radiation can activate stemness pathways in heterogeneous cancer cells, resulting in the enrichment of a CSC subpopulation with higher resistance to radiotherapy.


Asunto(s)
Rayos gamma , Células Madre Neoplásicas/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Células Madre Neoplásicas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Factor 3 de Transcripción de Unión a Octámeros/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción SOXB1/genética
11.
PLoS One ; 7(8): e43748, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22928027

RESUMEN

Mouse preimplantation development is characterized by three major transitions and two lineage segregations. Each transition or lineage segregation entails pronounced changes in the pattern of gene expression. Thus, research into the function of genes with obvious changes in expression pattern will shed light on the molecular basis of preimplantation development. We have described a simplified and effective method--electroporation--of introducing plasmid DNA and morpholinos into mouse preimplantation embryos and verified effectiveness of this approach by testing the procedure on the endogenous gene Oct4. Before electroporation, the zona pellucida was weakened by the treatment of acid Tyrode's solution. Then we optimized the parameters such as voltage, pulse duration, number of pulses and repeats, and applied these parameters to subsequent experiments. Compared with the control groups, the number of apoptotic cells and the expression and localization of OCT3/4 or CDX2 was not significantly changed in blastocysts developed from 1-cell embryos, which were electroporated with pIRES2-AcGFP1-Nuc eukaryotic expression vector or mismatched morpholino oligonucleotides. Furthermore, electroporated plasmid DNA and morpholinos targeting the endogenous gene Oct4 were able to sharply down regulate expression of OCT4 protein and actually cause expected phenotypes in mouse preimplantation embryos. In conclusion, plasmid DNA and morpholinos could be efficient delivered into mouse preimplantation embryos by electroporation and exert their functions, and normal development of preimplantation embryos was not affected.


Asunto(s)
Blastocisto/metabolismo , ADN/metabolismo , Electroporación/métodos , Morfolinos/metabolismo , Animales , Secuencia de Bases , Blastocisto/citología , ADN/genética , Electroporación/instrumentación , Femenino , Técnicas de Silenciamiento del Gen , Ratones , Morfolinos/genética , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Factor 3 de Transcripción de Unión a Octámeros/genética , Fenotipo , Plásmidos/genética , ARN Interferente Pequeño/genética , Zona Pelúcida/metabolismo
12.
Histochem Cell Biol ; 134(2): 197-204, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20532795

RESUMEN

The biological basis for manifestation of chemotherapy resistance in metastatic testicular germ cell tumors (GCT) remains obscure and is of particular clinical interest. In nonseminomatous GCT (NSGCT) the pluripotent embryonal carcinoma (EC) cells are the precursors of the manifold differentiated structures but also drive the malignant growth. They are known to be hypersensitive towards DNA-damaging agents and to express the embryonal transcription factor OCT4. We recently characterized EC cells that lack OCT4 expression and show cisplatin resistance. In the present, immunohistochemical study we analyzed the composition of NSGCT with the focus on such OCT4-negative EC cells using a NSGCT xenograft model as well as patient-derived NSGCT samples. In the xenograft model, the cisplatin-sensitive cell line H12.1 gives rise to xenografts where EC structures are mainly composed of OCT4-positive cells, whereas xenografts from the resistant cell line 1411HP exclusively comprise OCT4-negative EC areas. We found that post-chemotherapy residual metastatic tumors of patients can be comprised of exclusively OCT4-negative EC, whereas the matched testicular primary tumor harbors OCT4-positive EC. Thorough histological analyses revealed a few examples of such OCT4-negative EC cells also in the testicular primary tumor as well as in xenografts from the cisplatin-sensitive NSGCT-cell line. For these cells we propose an identity as early extraembryonal progenitor cells directly derived from OCT4-expressing EC cells. This challenges the use of the term EC cell. The data also support our hypothesis that malignant growth of resistant NSGCT may be driven by this cell type.


Asunto(s)
Cisplatino/farmacología , Resistencia a Antineoplásicos , Células Madre de Carcinoma Embrionario/patología , Neoplasias de Células Germinales y Embrionarias/patología , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Animales , Línea Celular Tumoral , Femenino , Histología , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Células Madre , Trasplante Heterólogo/patología
13.
Nature ; 460(7251): 128-32, 2009 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-19536159

RESUMEN

Pluripotency of embryonic stem (ES) cells is controlled by defined transcription factors. During differentiation, mouse ES cells undergo global epigenetic reprogramming, as exemplified by X-chromosome inactivation (XCI) in which one female X chromosome is silenced to achieve gene dosage parity between the sexes. Somatic XCI is regulated by homologous X-chromosome pairing and counting, and by the random choice of future active and inactive X chromosomes. XCI and cell differentiation are tightly coupled, as blocking one process compromises the other and dedifferentiation of somatic cells to induced pluripotent stem cells is accompanied by X chromosome reactivation. Recent evidence suggests coupling of Xist expression to pluripotency factors occurs, but how the two are interconnected remains unknown. Here we show that Oct4 (also known as Pou5f1) lies at the top of the XCI hierarchy, and regulates XCI by triggering X-chromosome pairing and counting. Oct4 directly binds Tsix and Xite, two regulatory noncoding RNA genes of the X-inactivation centre, and also complexes with XCI trans-factors, Ctcf and Yy1 (ref. 17), through protein-protein interactions. Depletion of Oct4 blocks homologous X-chromosome pairing and results in the inactivation of both X chromosomes in female cells. Thus, we have identified the first trans-factor that regulates counting, and ascribed new functions to Oct4 during X-chromosome reprogramming.


Asunto(s)
Emparejamiento Cromosómico , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Represoras/metabolismo , Inactivación del Cromosoma X/genética , Cromosoma X/genética , Cromosoma X/metabolismo , Animales , Factor de Unión a CCCTC , Línea Celular , Femenino , Humanos , Masculino , Ratones , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Factor 3 de Transcripción de Unión a Octámeros/genética , Unión Proteica , ARN Largo no Codificante , ARN no Traducido/genética , Factores de Transcripción SOXB1 , Activación Transcripcional , Factor de Transcripción YY1/metabolismo
14.
Blood ; 114(1): 60-3, 2009 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-19321862

RESUMEN

In embryonic stem cells, Oct-4 concentration is critical in determining the development of endoderm, mesoderm, and trophectoderm. Although Oct-4 expression is essential for mesoderm development, it is unclear whether it has a role in the development of specific mesodermal tissues. In this study, we have examined the importance of Oct-4 in the generation of hematopoietic cells using an inducible Oct-4 ESC line. We demonstrate that Oct-4 has a role in supporting hematopoiesis after specifying brachyury-positive mesoderm. When we suppressed Oct-4 expression before or after mesoderm specification, no hematopoietic cells are detected. However, hematopoiesis can be rescued in the absence of Oct-4 after mesoderm specification if the essential hematopoietic transcription factor stem cell leukemia is expressed. Our results suggest that, for hematopoiesis to occur, Oct-4 is required for the initial specification of mesoderm and subsequently is required for the development of hematopoietic cells from uncommitted mesoderm.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Hematopoyesis/fisiología , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Proteínas Proto-Oncogénicas/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Hematopoyesis/genética , Mesodermo/embriología , Mesodermo/fisiología , Ratones , Factor 3 de Transcripción de Unión a Octámeros/antagonistas & inhibidores , Factor 3 de Transcripción de Unión a Octámeros/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda , Transfección
15.
Dev Biol ; 315(1): 1-17, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18215655

RESUMEN

Pou5f1/Oct4 is a transcription factor required for pluripotency of embryonic stem cells in mammals. Zebrafish pou5f1 deficient maternal and zygotic spiel ohne grenzen (MZspg) mutant embryos develop severe gastrulation defects, are dorsalized, and defective in endoderm formation. Here we analyze in detail gastrulation defects, which are manifested by a severe delay in epiboly progression. All three embryonic lineages in MZspg embryos behave abnormally during epiboly: the yolk cell forms an altered array of cortical microtubules and F-Actin, with large patches of microtubule free areas; the enveloping layer (EVL) is delayed in the coordinated cell shape changes of marginal cells, that may be mediated by F-Actin; the deep layer cells (DEL), forming the embryo proper, are non-autonomously affected in their motility and do not enter the space opening by epiboly of the EVL. Analysis of adhesiveness as well as high resolution in vivo time lapse image analysis of DEL cells suggests changed adhesive properties and inability to migrate properly on EVL and yolk syncytial layer (YSL) surfaces. Our data further reveal that during epiboly the EVL may actively probe the YSL by filopodia formation, rather than just being passively pulled vegetalwards. Our findings on the effect of Pou5f1 on cell behavior may be relevant to understand stem cell behavior and tumorigenesis involving Pou5f1.


Asunto(s)
Linaje de la Célula , Citoesqueleto/fisiología , Mutación , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Pez Cebra/embriología , Actinas/metabolismo , Animales , Adhesión Celular/fisiología , Agregación Celular , Movimiento Celular , Tamaño de la Célula , Trasplante de Células , Embrión no Mamífero , Femenino , Gástrula/anomalías , Proteínas Fluorescentes Verdes/metabolismo , Microinyecciones , Microtúbulos/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , ARN Mensajero/genética , Transcripción Genética , Pez Cebra/genética , Pez Cebra/metabolismo
16.
FASEB J ; 21(9): 2020-32, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17356004

RESUMEN

Stem cells convert graded stimuli into all-or-nothing cell-fate responses. We investigated how embryonic stem cells (ESCs) convert leukemia inhibitory factor (LIF) concentration into an all-or-nothing cell-fate decision (self-renewal). Using a combined experimental/computational approach we demonstrate unexpected switch-like (on/off) signaling in response to LIF. This behavior emerges over time due to a positive feedback loop controlling transcriptional expression of LIF signaling pathway components. The autoregulatory loop maintains robust pathway responsiveness ("on") at sufficient concentrations of exogenous LIF, while autocrine signaling and low concentrations of exogenous LIF cause ESCs to adopt the weakly responsive ("off") state of differentiated cells. We demonstrate that loss of ligand responsiveness is reversible and precedes loss of the ESC transcription factors Oct4 and Nanog, suggesting an early step in the hierarchical control of differentiation. While endogenously produced ligands were insufficient to sustain the "on" state, they buffer it, influencing the timing of differentiation. These results demonstrate a novel switch-like behavior, which establishes the LIF threshold for ESC self-renewal.


Asunto(s)
Comunicación Autocrina/fisiología , Células Madre Embrionarias/citología , Retroalimentación Fisiológica/fisiología , Factor Inhibidor de Leucemia/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , División Celular/efectos de los fármacos , División Celular/fisiología , Receptor gp130 de Citocinas/biosíntesis , Receptor gp130 de Citocinas/genética , Proteínas de Unión al ADN/fisiología , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/fisiología , Factor Inhibidor de Leucemia/fisiología , Ratones , Modelos Biológicos , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Receptores OSM-LIF/biosíntesis , Receptores OSM-LIF/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología
17.
Stem Cells ; 25(2): 500-10, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17068183

RESUMEN

The POU domain transcription factor OCT4 is a key regulator of pluripotency in the early mammalian embryo and is highly expressed in the inner cell mass of the blastocyst. Consistent with its essential role in maintaining pluripotency, Oct4 expression is rapidly downregulated during formation of the trophoblast lineage. To enhance our understanding of the molecular basis of this differentiation event in humans, we used a functional genomics approach involving RNA interference-mediated suppression of OCT4 function in a human ESC line and analysis of the resulting transcriptional profiles to identify OCT4-dependent genes in human cells. We detected altered expression of >1,000 genes, including targets regulated directly by OCT4 either positively (NANOG, SOX2, REX1, LEFTB, LEFTA/EBAF DPPA4, THY1, and TDGF1) or negatively (CDX2, EOMES, BMP4, TBX18, Brachyury [T], DKK1, HLX1, GATA6, ID2, and DLX5), as well as targets for the OCT4-associated stem cell regulators SOX2 and NANOG. Our data set includes regulators of ACTIVIN, BMP, fibroblast growth factor, and WNT signaling. These pathways are implicated in regulating human ESC differentiation and therefore further validate the results of our analysis. In addition, we identified a number of differentially expressed genes that are involved in epigenetics, chromatin remodeling, apoptosis, and metabolism that may point to underlying molecular mechanisms that regulate pluripotency and trophoblast differentiation in humans. Significant concordance between this data set and previous comparisons between inner cell mass and trophectoderm in human embryos indicates that the study of human ESC differentiation in vitro represents a useful model of early embryonic differentiation in humans.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Redes Reguladoras de Genes/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Apoptosis/genética , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Linaje de la Célula , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Redes y Vías Metabólicas/genética , Factor 3 de Transcripción de Unión a Octámeros/deficiencia , Factor 3 de Transcripción de Unión a Octámeros/genética , Interferencia de ARN , ARN Interferente Pequeño , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal/genética , Supresión Genética , Transfección , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Trofoblastos/citología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...