Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
RNA ; 29(10): 1471-1480, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37433650

RESUMEN

RNA-directed transposon silencing operates in the mammalian soma and germline to safeguard genomic integrity. The piRNA pathway and the HUSH complex identify active transposons through recognition of their nascent transcripts, but mechanistic understanding of how these distinct pathways evolved is lacking. TASOR is an essential component of the HUSH complex. TASOR's DUF3715 domain adopts a pseudo-PARP structure and is required for transposon silencing in a manner independent of complex assembly. TEX15, an essential piRNA pathway factor, also contains the DUF3715 domain. Here, we show that TASOR's and TEX15's DUF3715 domain share extensive structural homology. We found that the DUF3715 domain arose in early eukaryotes and that in vertebrates it is restricted to TEX15, TASOR, and TASORB orthologs. While TASOR-like proteins are found throughout metazoa, TEX15 is vertebrate-specific. The branching of TEX15 and the TASOR-like DUF3715 domain likely occurred in early metazoan evolution. Remarkably, despite this vast evolutionary distance, the DUF3715 domain from divergent TEX15 sequences can functionally substitute the DUF3715 domain of TASOR and mediates transposon silencing. We have thus termed this domain of unknown function as the RNA-directed pseudo-PARP transposon silencing (RDTS) domain. In summary, we show an unexpected functional link between these critical transposon silencing pathways.


Asunto(s)
Proteínas de Drosophila , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Animales , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Interferencia de ARN , Genoma , Proteínas Argonautas/genética , ARN de Interacción con Piwi , Mamíferos/genética , Elementos Transponibles de ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética
2.
Genes Dev ; 31(20): 2085-2098, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-29138277

RESUMEN

Expression of the transcription factors OCT4, SOX2, KLF4, and cMYC (OSKM) reprograms somatic cells into induced pluripotent stem cells (iPSCs). Reprogramming is a slow and inefficient process, suggesting the presence of safeguarding mechanisms that counteract cell fate conversion. One such mechanism is senescence. To identify modulators of reprogramming-induced senescence, we performed a genome-wide shRNA screen in primary human fibroblasts expressing OSKM. In the screen, we identified novel mediators of OSKM-induced senescence and validated previously implicated genes such as CDKN1A We developed an innovative approach that integrates single-cell RNA sequencing (scRNA-seq) with the shRNA screen to investigate the mechanism of action of the identified candidates. Our data unveiled regulation of senescence as a novel way by which mechanistic target of rapamycin (mTOR) influences reprogramming. On one hand, mTOR inhibition blunts the induction of cyclin-dependent kinase (CDK) inhibitors (CDKIs), including p16INK4a, p21CIP1, and p15INK4b, preventing OSKM-induced senescence. On the other hand, inhibition of mTOR blunts the senescence-associated secretory phenotype (SASP), which itself favors reprogramming. These contrasting actions contribute to explain the complex effect that mTOR has on reprogramming. Overall, our study highlights the advantage of combining functional screens with scRNA-seq to accelerate the discovery of pathways controlling complex phenotypes.


Asunto(s)
Reprogramación Celular , Senescencia Celular , Perfilación de la Expresión Génica , ARN Interferente Pequeño , Análisis de Secuencia de ARN , Serina-Treonina Quinasas TOR/fisiología , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Factor 4 Similar a Kruppel , Ratones , Análisis de la Célula Individual , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
3.
Genome Res ; 28(4): 592-605, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29572359

RESUMEN

The identification of transcription factor (TF) binding sites in the genome is critical to understanding gene regulatory networks (GRNs). While ChIP-seq is commonly used to identify TF targets, it requires specific ChIP-grade antibodies and high cell numbers, often limiting its applicability. DNA adenine methyltransferase identification (DamID), developed and widely used in Drosophila, is a distinct technology to investigate protein-DNA interactions. Unlike ChIP-seq, it does not require antibodies, precipitation steps, or chemical protein-DNA crosslinking, but to date it has been seldom used in mammalian cells due to technical limitations. Here we describe an optimized DamID method coupled with next-generation sequencing (DamID-seq) in mouse cells and demonstrate the identification of the binding sites of two TFs, POU5F1 (also known as OCT4) and SOX2, in as few as 1000 embryonic stem cells (ESCs) and neural stem cells (NSCs), respectively. Furthermore, we have applied this technique in vivo for the first time in mammals. POU5F1 DamID-seq in the gastrulating mouse embryo at 7.5 d post coitum (dpc) successfully identified multiple POU5F1 binding sites proximal to genes involved in embryo development, neural tube formation, and mesoderm-cardiac tissue development, consistent with the pivotal role of this TF in post-implantation embryo. This technology paves the way to unprecedented investigation of TF-DNA interactions and GRNs in specific cell types of limited availability in mammals, including in vivo samples.


Asunto(s)
Genoma/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción/genética , Animales , Sitios de Unión/genética , Células Madre Embrionarias/metabolismo , Redes Reguladoras de Genes/genética , Ratones , Células-Madre Neurales/metabolismo , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/genética
4.
BMC Genomics ; 20(1): 100, 2019 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-30704403

RESUMEN

BACKGROUND: Along with the reorganization of epigenetic and transcriptional networks, somatic cell reprogramming brings about numerous changes at the level of RNA processing. These include the expression of specific transcript isoforms and 3' untranslated regions. A number of studies have uncovered RNA processing factors that modulate the efficiency of the reprogramming process. However, a comprehensive evaluation of the involvement of RNA processing factors in the reprogramming of somatic mammalian cells is lacking. RESULTS: Here, we used data from a large number of studies carried out in three mammalian species, mouse, chimpanzee and human, to uncover consistent changes in gene expression upon reprogramming of somatic cells. We found that a core set of nine splicing factors have consistent changes across the majority of data sets in all three species. Most striking among these are ESRP1 and ESRP2, which accelerate and enhance the efficiency of somatic cell reprogramming by promoting isoform expression changes associated with mesenchymal-to-epithelial transition. We further identify genes and processes in which splicing changes are observed in both human and mouse. CONCLUSIONS: Our results provide a general resource for gene expression and splicing changes that take place during somatic cell reprogramming. Furthermore, they support the concept that splicing factors with evolutionarily conserved, cell type-specific expression can modulate the efficiency of the process by reinforcing intermediate states resembling the cell types in which these factors are normally expressed.


Asunto(s)
Reprogramación Celular , Procesamiento Postranscripcional del ARN , Empalme del ARN , Animales , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Pan troglodytes , Isoformas de Proteínas , Proteínas de Unión al ARN/genética
5.
Nature ; 499(7456): 88-91, 2013 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-23728301

RESUMEN

The generation of induced pluripotent stem (iPS) cells presents a challenge to normal developmental processes. The low efficiency and heterogeneity of most methods have hindered understanding of the precise molecular mechanisms promoting, and roadblocks preventing, efficient reprogramming. Although several intermediate populations have been described, it has proved difficult to characterize the rare, asynchronous transition from these intermediate stages to iPS cells. The rapid expansion of minor reprogrammed cells in the heterogeneous population can also obscure investigation of relevant transition processes. Understanding the biological mechanisms essential for successful iPS cell generation requires both accurate capture of cells undergoing the reprogramming process and identification of the associated global gene expression changes. Here we demonstrate that in mouse embryonic fibroblasts, reprogramming follows an orderly sequence of stage transitions, marked by changes in the cell-surface markers CD44 and ICAM1, and a Nanog-enhanced green fluorescent protein (Nanog-eGFP) reporter. RNA-sequencing analysis of these populations demonstrates two waves of pluripotency gene upregulation, and unexpectedly, transient upregulation of several epidermis-related genes, demonstrating that reprogramming is not simply the reversal of the normal developmental processes. This novel high-resolution analysis enables the construction of a detailed reprogramming route map, and the improved understanding of the reprogramming process will lead to new reprogramming strategies.


Asunto(s)
Reprogramación Celular/fisiología , Receptores de Hialuranos/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Reprogramación Celular/genética , Epidermis/metabolismo , Fibroblastos , Citometría de Flujo , Perfilación de la Expresión Génica , Genes Reporteros , Receptores de Hialuranos/genética , Molécula 1 de Adhesión Intercelular/genética , Ratones , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Regulación hacia Arriba/genética
6.
Stem Cells ; 33(3): 699-712, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25377420

RESUMEN

Identifying novel players of the pluripotency gene regulatory network centered on Oct4, Sox2, and Nanog as well as delineating the interactions within the complex network is key to understanding self-renewal and early cell fate commitment of embryonic stem cells (ESC). While overexpression of the transcriptional regulator Cited2 sustains ESC pluripotency, its role in ESC functions remains unclear. Here, we show that Cited2 is important for proliferation, survival, and self-renewal of mouse ESC. We position Cited2 within the pluripotency gene regulatory network by defining Nanog, Tbx3, and Klf4 as its direct targets. We also demonstrate that the defects caused by Cited2 depletion are, at least in part, rescued by Nanog constitutive expression. Finally, we demonstrate that Cited2 is required for and enhances reprogramming of mouse embryonic fibroblasts to induced pluripotent stem cells.


Asunto(s)
Células Madre Embrionarias/fisiología , Proteínas de Homeodominio/biosíntesis , Células Madre Pluripotentes/fisiología , Proteínas Represoras/deficiencia , Transactivadores/deficiencia , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Redes Reguladoras de Genes , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factor 4 Similar a Kruppel , Ratones , Proteína Homeótica Nanog , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Transfección
7.
Nature ; 458(7239): 771-5, 2009 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-19252477

RESUMEN

Reprogramming of somatic cells to pluripotency, thereby creating induced pluripotent stem (iPS) cells, promises to transform regenerative medicine. Most instances of direct reprogramming have been achieved by forced expression of defined factors using multiple viral vectors. However, such iPS cells contain a large number of viral vector integrations, any one of which could cause unpredictable genetic dysfunction. Whereas c-Myc is dispensable for reprogramming, complete elimination of the other exogenous factors is also desired because ectopic expression of either Oct4 (also known as Pou5f1) or Klf4 can induce dysplasia. Two transient transfection-reprogramming methods have been published to address this issue. However, the efficiency of both approaches is extremely low, and neither has been applied successfully to human cells so far. Here we show that non-viral transfection of a single multiprotein expression vector, which comprises the coding sequences of c-Myc, Klf4, Oct4 and Sox2 linked with 2A peptides, can reprogram both mouse and human fibroblasts. Moreover, the transgene can be removed once reprogramming has been achieved. iPS cells produced with this non-viral vector show robust expression of pluripotency markers, indicating a reprogrammed state confirmed functionally by in vitro differentiation assays and formation of adult chimaeric mice. When the single-vector reprogramming system was combined with a piggyBac transposon, we succeeded in establishing reprogrammed human cell lines from embryonic fibroblasts with robust expression of pluripotency markers. This system minimizes genome modification in iPS cells and enables complete elimination of exogenous reprogramming factors, efficiently providing iPS cells that are applicable to regenerative medicine, drug screening and the establishment of disease models.


Asunto(s)
Reprogramación Celular/genética , Vectores Genéticos/genética , Células Madre Pluripotentes/citología , Transfección/métodos , Animales , Biomarcadores/análisis , Línea Celular , Células Cultivadas , Fibroblastos/citología , Perfilación de la Expresión Génica , Humanos , Factor 4 Similar a Kruppel , Ratones , Células Madre Pluripotentes/metabolismo , Transgenes/genética
8.
Nature ; 458(7239): 766-70, 2009 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-19252478

RESUMEN

Transgenic expression of just four defined transcription factors (c-Myc, Klf4, Oct4 and Sox2) is sufficient to reprogram somatic cells to a pluripotent state. The resulting induced pluripotent stem (iPS) cells resemble embryonic stem cells in their properties and potential to differentiate into a spectrum of adult cell types. Current reprogramming strategies involve retroviral, lentiviral, adenoviral and plasmid transfection to deliver reprogramming factor transgenes. Although the latter two methods are transient and minimize the potential for insertion mutagenesis, they are currently limited by diminished reprogramming efficiencies. piggyBac (PB) transposition is host-factor independent, and has recently been demonstrated to be functional in various human and mouse cell lines. The PB transposon/transposase system requires only the inverted terminal repeats flanking a transgene and transient expression of the transposase enzyme to catalyse insertion or excision events. Here we demonstrate successful and efficient reprogramming of murine and human embryonic fibroblasts using doxycycline-inducible transcription factors delivered by PB transposition. Stable iPS cells thus generated express characteristic pluripotency markers and succeed in a series of rigorous differentiation assays. By taking advantage of the natural propensity of the PB system for seamless excision, we show that the individual PB insertions can be removed from established iPS cell lines, providing an invaluable tool for discovery. In addition, we have demonstrated the traceless removal of reprogramming factors joined with viral 2A sequences delivered by a single transposon from murine iPS lines. We anticipate that the unique properties of this virus-independent simplification of iPS cell production will accelerate this field further towards full exploration of the reprogramming process and future cell-based therapies.


Asunto(s)
Diferenciación Celular , Reprogramación Celular/genética , Fibroblastos/citología , Fibroblastos/fisiología , Vectores Genéticos/genética , Células Madre Pluripotentes/fisiología , Animales , Línea Celular , Células Cultivadas , Elementos Transponibles de ADN , Fibroblastos/virología , Orden Génico , Técnicas de Transferencia de Gen , Humanos , Factor 4 Similar a Kruppel , Ratones , Ratones Desnudos , Alineación de Secuencia , Factores de Transcripción/genética , Transgenes/genética
9.
Nat Cell Biol ; 8(3): 285-92, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16462733

RESUMEN

Cells of early mammalian embryos have the potential to develop into any adult cell type, and are thus said to be pluripotent. Pluripotency is lost during embryogenesis as cells commit to specific developmental pathways. Although restriction of developmental potential is often associated with repression of inappropriate genetic programmes, the role of epigenetic silencing during early lineage commitment remains undefined. Here, we used mouse embryonic stem cells to study the function of epigenetic silencing in pluripotent cells. Embryonic stem cells lacking Mbd3 - a component of the nucleosome remodelling and histone deacetylation (NuRD) complex - were viable but failed to completely silence genes that are expressed before implantation of the embryo. Mbd3-deficient embryonic stem cells could be maintained in the absence of leukaemia inhibitory factor (LIF) and could initiate differentiation in embryoid bodies or chimeric embryos, but failed to commit to developmental lineages. Our findings define a role for epigenetic silencing in the cell-fate commitment of pluripotent cells.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Histona Desacetilasas/metabolismo , Células Madre Pluripotentes/citología , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/citología , Epigénesis Genética , Silenciador del Gen , Histona Desacetilasas/genética , Interleucina-6/fisiología , Factor Inhibidor de Leucemia , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2 , Ratones , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/genética
10.
Nat Commun ; 14(1): 488, 2023 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-36717582

RESUMEN

Induced pluripotent stem cell (iPSC) reprogramming is inefficient and understanding the molecular mechanisms underlying this inefficiency holds the key to successfully control cellular identity. Here, we report 24 reprogramming roadblock genes identified by CRISPR/Cas9-mediated genome-wide knockout (KO) screening. Of these, depletion of the predicted KRAB zinc finger protein (KRAB-ZFP) Zfp266 strongly and consistently enhances murine iPSC generation in several reprogramming settings, emerging as the most robust roadblock. We show that ZFP266 binds Short Interspersed Nuclear Elements (SINEs) adjacent to binding sites of pioneering factors, OCT4 (POU5F1), SOX2, and KLF4, and impedes chromatin opening. Replacing the KRAB co-suppressor with co-activator domains converts ZFP266 from an inhibitor to a potent facilitator of iPSC reprogramming. We propose that the SINE-KRAB-ZFP interaction is a critical regulator of chromatin accessibility at regulatory elements required for efficient cellular identity changes. In addition, this work serves as a resource to further illuminate molecular mechanisms hindering reprogramming.


Asunto(s)
Células Madre Pluripotentes Inducidas , Dedos de Zinc , Animales , Ratones , Reprogramación Celular/genética , Cromatina/genética , Cromatina/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel
11.
iScience ; 25(8): 104743, 2022 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-35942105

RESUMEN

Experimental and computational efforts are constantly made to elucidate mechanisms controlling cell fate decisions during development and reprogramming. One powerful computational method is to consider cell commitment and reprogramming as movements in an energy landscape. Here, we develop Computation of Energy Landscapes of Logical Gene Networks (CELLoGeNe), which maps Boolean implementation of gene regulatory networks (GRNs) into energy landscapes. CELLoGeNe removes inadvertent symmetries in the energy landscapes normally arising from standard Boolean operators. Furthermore, CELLoGeNe provides tools to visualize and stochastically analyze the shapes of multi-dimensional energy landscapes corresponding to epigenetic landscapes for development and reprogramming. We demonstrate CELLoGeNe on two GRNs governing different aspects of induced pluripotent stem cells, identifying experimentally validated attractors and revealing potential reprogramming roadblocks. CELLoGeNe is a general framework that can be applied to various biological systems offering a broad picture of intracellular dynamics otherwise inaccessible with existing methods.

12.
Cell Rep ; 24(6): 1496-1511.e8, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30089261

RESUMEN

During somatic reprogramming, Yamanaka's pioneer factors regulate a complex sequence of molecular events leading to the activation of a network of pluripotency factors, ultimately resulting in the acquisition and maintenance of a pluripotent state. Here, we show that, contrary to the pluripotency factors studied so far, overexpression of Mybl2 inhibits somatic reprogramming. Our results demonstrate that Mybl2 levels are crucial to the dynamics of the reprogramming process. Mybl2 overexpression changes chromatin conformation, affecting the accessibility of pioneer factors to the chromatin and promoting accessibility for early immediate response genes known to be reprogramming blockers. These changes in the chromatin landscape ultimately lead to a deregulation of key genes that are important for the mesenchymal-to-epithelial transition. This work defines Mybl2 level as a gatekeeper for the initiation of reprogramming, providing further insights into the tight regulation and required coordination of molecular events that are necessary for changes in cell fate identity during the reprogramming process.


Asunto(s)
Proteínas de Ciclo Celular/genética , Transactivadores/genética , Reprogramación Celular , Transición Epitelial-Mesenquimal , Humanos , Transfección
13.
Keio J Med ; 66(1): 14, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28356547

RESUMEN

The generation of induced pluripotent stem cells (iPSCs) achieved by overexpression of Oct4, Sox2, Klf4 and c-Myc, transformed our classical views of the cellular epigenetic landscape and delivered a new concept for cell and tissue engineering. In addition to iPSCs, several other cell types have also been generated by master transcription factor (TF)-mediated transdifferentiation. However, the critical molecular mechanisms amongst diverse cellular identity changes are not well understood. Through the investigation of reprogramming mechanisms, we recently revealed that over-expression of constitutive active Smad3 boosted not only iPSC generation, but also 3 other master TF-mediated conversions, from B cells to macrophages, myoblasts to adipocytes, and human fibroblasts to neurons. This demonstrated that there were common mechanisms underlying different master TF-mediated cell conversions. To illuminate such mechanisms further, we have recently performed CRISPR/Cas9-mediated genome-wide knockout screening during reprogramming with a lentiviral gRNA library containing 90,000 gRNAs. This screening provided us with ~15 novel reprogramming roadblock genes as well as ~20 candidate genes essential for the reprogramming process but not for ES cell self-renewal. This data set will be a valuable resource to further understand how overexpression of master TFs alters cellular identity, and to achieve more faithful, efficient cell conversions for regenerative medicine.(Presented at the 1934th Meeting, March 17, 2017).


Asunto(s)
Transdiferenciación Celular/genética , Reprogramación Celular , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Mioblastos/citología , Mioblastos/metabolismo , Neuronas/citología , Neuronas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo
14.
Cell Stem Cell ; 21(6): 791-805.e9, 2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29174331

RESUMEN

Reprogramming of cellular identity using exogenous expression of transcription factors (TFs) is a powerful and exciting tool for tissue engineering, disease modeling, and regenerative medicine. However, generation of desired cell types using this approach is often plagued by inefficiency, slow conversion, and an inability to produce mature functional cells. Here, we show that expression of constitutively active SMAD2/3 significantly improves the efficiency of induced pluripotent stem cell (iPSC) generation by the Yamanaka factors. Mechanistically, SMAD3 interacts with reprogramming factors and co-activators and co-occupies OCT4 target loci during reprogramming. Unexpectedly, active SMAD2/3 also markedly enhances three other TF-mediated direct reprogramming conversions, from B cells to macrophages, myoblasts to adipocytes, and human fibroblasts to neurons, highlighting broad and general roles for SMAD2/3 as cell-reprogramming potentiators. Our results suggest that co-expression of active SMAD2/3 could enhance multiple types of TF-based cell identity conversion and therefore be a powerful tool for cellular engineering.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factores de Transcripción/metabolismo , Línea Celular , Humanos , Factores de Transcripción/genética
15.
Methods Mol Biol ; 1357: 285-93, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25410291

RESUMEN

Generation of iPSCs is inefficient and the molecular mechanisms underlying reprogramming are not well understood. While several studies have demonstrated that reprogramming is not entirely a random process and contains predictable stepwise changes, varying degrees of cellular heterogeneity that arise in different reprogramming systems can obscure the process. Among several reprogramming systems available, delivery of polycistronic reprogramming factor expression cassettes with piggyBac transposon into mouse embryonic fibroblasts (MEFs) is one of the simplest and most robust reprogramming approaches that provide a low background of partially reprogrammed cells. Using two novel cell surface markers, ICAM1 and CD44, clear cell population changes undergoing reprogramming can be observed over a time course upon induction of the reprogramming factors. Consequently, this technique allows for easy identification of factors that enhance or delay reprogramming, and can be a useful strategy in elucidating key mechanisms for efficient generation of iPSCs.


Asunto(s)
Antígenos de Superficie/análisis , Técnicas de Reprogramación Celular/métodos , Reprogramación Celular/genética , Elementos Transponibles de ADN/genética , Receptores de Hialuranos/análisis , Células Madre Pluripotentes Inducidas/citología , Molécula 1 de Adhesión Intercelular/análisis , Animales , Separación Celular , Células Cultivadas , Fibroblastos/citología , Citometría de Flujo , Genes Reporteros , Genes myc , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas de Homeodominio/genética , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/fisiología , Proteínas Luminiscentes/genética , Ratones , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Proteínas Proto-Oncogénicas c-myc/fisiología , Proteínas Recombinantes/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/fisiología
17.
Stem Cell Reports ; 5(3): 350-64, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26278041

RESUMEN

Since the first generation of induced pluripotent stem cells (iPSCs), several reprogramming systems have been used to study its molecular mechanisms. However, the system of choice largely affects the reprogramming efficiency, influencing our view on the mechanisms. Here, we demonstrate that reprogramming triggered by less efficient polycistronic reprogramming cassettes not only highlights mesenchymal-to-epithelial transition (MET) as a roadblock but also faces more severe difficulties to attain a pluripotent state even post-MET. In contrast, more efficient cassettes can reprogram both wild-type and Nanog(-/-) fibroblasts with comparable efficiencies, routes, and kinetics, unlike the less efficient reprogramming systems. Moreover, we attribute a previously reported variation in the N terminus of KLF4 as a dominant factor underlying these critical differences. Our data establish that some reprogramming roadblocks are system dependent, highlighting the need to pursue mechanistic studies with close attention to the systems to better understand reprogramming.


Asunto(s)
Reprogramación Celular , Transición Epitelial-Mesenquimal , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Fibroblastos/citología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes Inducidas/citología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Noqueados , Proteína Homeótica Nanog
18.
Phys Rev E Stat Nonlin Soft Matter Phys ; 65(6 Pt 1): 061801, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12188750

RESUMEN

In order to clarify the structure formation processes in the induction period of polymer crystallization the annealing time dependence of depolarized light-scattering (DPLS) intensities has been investigated as a function of crystallization temperature for poly(ethylene terephthalate), poly(ethylene naphthalate), syndiotactic polystyrene, and isotactic polystyrene. It is found that the induction period may be separated into three stages: the first stage where the DPLS intensity hardly changes with time, the second stage where the intensity increases exponentially, and the third stage where it levels off. Considering that the DPLS provides information about the degree of parallel orientation of rigid polymer segments, the first stage whose time length depends on the annealing temperature may be assigned to a process where the polymer chains begin to partially assume a rigid conformation, generally a helical structure being almost the same as the structure in the corresponding crystal. This process is limited to a time when the average length of the rigid segments attains a critical value given by a Shimada, Doi, and Okano theory [J. Chem. Phys. 88, 7181 (1988)] above which spinodal decomposition (SD) is caused. The second and third stages correspond to the early and late stages of SD, respectively, which was confirmed by small-angle x-ray scattering measurements. The apparent activation energies obtained from the temperature dependence of the DPLS intensities for the three stages were 35-40, 25-50, and 180-400 kJ/mol, respectively, for all the polymers. The large apparent activation energies for the late stage of SD is discussed within a framework of Binder and Stauffer's theory [Phys. Rev. Lett. 33, 1006 (1974)].

19.
Curr Opin Genet Dev ; 28: 38-42, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25198100

RESUMEN

The generation of induced pluripotent stem cells (iPSCs) with Oct4, Sox2, Klf4, c-Myc has been described as 'direct' reprogramming in contrast to reprogramming via nuclear transfer. Interestingly, recent studies have suggested that the conversion process itself includes transient up-regulation and down-regulation of hundreds of genes, making unique intermediate populations. In a sense, the process of 4 factor reprogramming is indirect. Like in vitro differentiation, iPSC generation efficiency and kinetics largely depend on the external environment, as well as the amount and stoichiometry of exogenously expressed reprogramming factors. However, accumulating evidence indicates that when reprogramming succeeds, the process is not random but progresses in an ordered, step-wise manner. In this review, we summarize current knowledge detailing how somatic cells reach a pluripotent state.


Asunto(s)
Linaje de la Célula , Reprogramación Celular , Células Madre Pluripotentes/citología , Animales , Humanos , Factor 4 Similar a Kruppel
20.
Cell Stem Cell ; 15(1): 102-10, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24835571

RESUMEN

The Nucleosome Remodeling and Deacetylase (NuRD) complex is essential for embryonic development and pluripotent stem cell differentiation. In this study, we investigated whether NuRD is also involved in the reverse biological process of induction of pluripotency in neural stem cells. By knocking out MBD3, an essential scaffold subunit of the NuRD complex, at different time points in reprogramming, we found that efficient formation of reprogramming intermediates and induced pluripotent stem cells from neural stem cells requires NuRD activity. We also show that reprogramming of epiblast-derived stem cells to naive pluripotency requires NuRD complex function and that increased MBD3/NuRD levels can enhance reprogramming efficiency when coexpressed with the reprogramming factor NANOG. Our results therefore show that the MBD3/NuRD complex plays a key role in reprogramming in certain contexts and that a chromatin complex required for cell differentiation can also promote reversion back to a naive pluripotent cell state.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Células-Madre Neurales/fisiología , Células Madre Pluripotentes/fisiología , Animales , Desdiferenciación Celular/genética , Diferenciación Celular/genética , Línea Celular , Reprogramación Celular/genética , Proteínas de Unión al ADN/genética , Desarrollo Embrionario/genética , Técnicas de Inactivación de Genes , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Ratones , Ratones Endogámicos , Ratones Noqueados , Proteína Homeótica Nanog
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA