Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Nat Rev Mol Cell Biol ; 23(4): 231-249, 2022 04.
Article in English | MEDLINE | ID: mdl-35013589

ABSTRACT

X-chromosome inactivation (XCI) is the epigenetic mechanism that ensures X-linked dosage compensation between cells of females (XX karyotype) and males (XY). XCI is essential for female embryos to survive through development and requires the accurate spatiotemporal regulation of many different factors to achieve remarkable chromosome-wide gene silencing. As a result of XCI, the active and inactive X chromosomes are functionally and structurally different, with the inactive X chromosome undergoing a major conformational reorganization within the nucleus. In this Review, we discuss the multiple layers of genetic and epigenetic regulation that underlie initiation of XCI during development and then maintain it throughout life, in light of the most recent findings in this rapidly advancing field. We discuss exciting new insights into the regulation of X inactive-specific transcript (XIST), the trigger and master regulator of XCI, and into the mechanisms and dynamics that underlie the silencing of nearly all X-linked genes. Finally, given the increasing interest in understanding the impact of chromosome organization on gene regulation, we provide an overview of the factors that are thought to reshape the 3D structure of the inactive X chromosome and of the relevance of such structural changes for XCI establishment and maintenance.


Subject(s)
Epigenesis, Genetic , RNA, Long Noncoding , Epigenesis, Genetic/genetics , Female , Gene Silencing , Humans , Male , RNA, Long Noncoding/genetics , X Chromosome/genetics , X Chromosome Inactivation/genetics
2.
Nature ; 580(7801): 142-146, 2020 04.
Article in English | MEDLINE | ID: mdl-32238933

ABSTRACT

Paternal and maternal epigenomes undergo marked changes after fertilization1. Recent epigenomic studies have revealed the unusual chromatin landscapes that are present in oocytes, sperm and early preimplantation embryos, including atypical patterns of histone modifications2-4 and differences in chromosome organization and accessibility, both in gametes5-8 and after fertilization5,8-10. However, these studies have led to very different conclusions: the global absence of local topological-associated domains (TADs) in gametes and their appearance in the embryo8,9 versus the pre-existence of TADs and loops in the zygote5,11. The questions of whether parental structures can be inherited in the newly formed embryo and how these structures might relate to allele-specific gene regulation remain open. Here we map genomic interactions for each parental genome (including the X chromosome), using an optimized single-cell high-throughput chromosome conformation capture (HiC) protocol12,13, during preimplantation in the mouse. We integrate chromosome organization with allelic expression states and chromatin marks, and reveal that higher-order chromatin structure after fertilization coincides with an allele-specific enrichment of methylation of histone H3 at lysine 27. These early parental-specific domains correlate with gene repression and participate in parentally biased gene expression-including in recently described, transiently imprinted loci14. We also find TADs that arise in a non-parental-specific manner during a second wave of genome assembly. These de novo domains are associated with active chromatin. Finally, we obtain insights into the relationship between TADs and gene expression by investigating structural changes to the paternal X chromosome before and during X chromosome inactivation in preimplantation female embryos15. We find that TADs are lost as genes become silenced on the paternal X chromosome but linger in regions that escape X chromosome inactivation. These findings demonstrate the complex dynamics of three-dimensional genome organization and gene expression during early development.


Subject(s)
Blastocyst/cytology , Blastocyst/metabolism , Chromatin/metabolism , Embryonic Development/genetics , Fertilization/genetics , Germ Cells/cytology , Parents , Alleles , Animals , Chromatin/chemistry , Chromatin/genetics , Chromosome Positioning , Chromosomes, Mammalian/chemistry , Chromosomes, Mammalian/genetics , Chromosomes, Mammalian/metabolism , Female , Gene Expression Regulation, Developmental , Genome/genetics , Genomic Imprinting , Germ Cells/metabolism , Histones/chemistry , Histones/metabolism , Male , Methylation , Mice , Polycomb-Group Proteins/metabolism , Single-Cell Analysis , X Chromosome Inactivation/genetics
3.
Nature ; 578(7795): 455-460, 2020 02.
Article in English | MEDLINE | ID: mdl-32025035

ABSTRACT

Xist represents a paradigm for the function of long non-coding RNA in epigenetic regulation, although how it mediates X-chromosome inactivation (XCI) remains largely unexplained. Several proteins that bind to Xist RNA have recently been identified, including the transcriptional repressor SPEN1-3, the loss of which has been associated with deficient XCI at multiple loci2-6. Here we show in mice that SPEN is a key orchestrator of XCI in vivo and we elucidate its mechanism of action. We show that SPEN is essential for initiating gene silencing on the X chromosome in preimplantation mouse embryos and in embryonic stem cells. SPEN is dispensable for maintenance of XCI in neural progenitors, although it significantly decreases the expression of genes that escape XCI. We show that SPEN is immediately recruited to the X chromosome upon the upregulation of Xist, and is targeted to enhancers and promoters of active genes. SPEN rapidly disengages from chromatin upon gene silencing, suggesting that active transcription is required to tether SPEN to chromatin. We define the SPOC domain as a major effector of the gene-silencing function of SPEN, and show that tethering SPOC to Xist RNA is sufficient to mediate gene silencing. We identify the protein partners of SPOC, including NCoR/SMRT, the m6A RNA methylation machinery, the NuRD complex, RNA polymerase II and factors involved in the regulation of transcription initiation and elongation. We propose that SPEN acts as a molecular integrator for the initiation of XCI, bridging Xist RNA with the transcription machinery-as well as with nucleosome remodellers and histone deacetylases-at active enhancers and promoters.


Subject(s)
DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Silencing , RNA-Binding Proteins/metabolism , Transcription, Genetic , X Chromosome Inactivation/genetics , X Chromosome/genetics , Animals , Blastocyst/cytology , Blastocyst/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/chemistry , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic/genetics , Female , Histone Deacetylases/metabolism , Male , Methylation , Mice , Promoter Regions, Genetic/genetics , Protein Domains , RNA, Long Noncoding/genetics , RNA-Binding Proteins/chemistry
4.
Genome Res ; 29(10): 1659-1672, 2019 10.
Article in English | MEDLINE | ID: mdl-31515287

ABSTRACT

Induction and reversal of chromatin silencing is critical for successful development, tissue homeostasis, and the derivation of induced pluripotent stem cells (iPSCs). X-Chromosome inactivation (XCI) and reactivation (XCR) in female cells represent chromosome-wide transitions between active and inactive chromatin states. Although XCI has long been studied, providing important insights into gene regulation, the dynamics and mechanisms underlying the reversal of stable chromatin silencing of X-linked genes are much less understood. Here, we use allele-specific transcriptomics to study XCR during mouse iPSC reprogramming in order to elucidate the timing and mechanisms of chromosome-wide reversal of gene silencing. We show that XCR is hierarchical, with subsets of genes reactivating early, late, and very late during reprogramming. Early genes are activated before the onset of late pluripotency genes activation. Early genes are located genomically closer to genes that escape XCI, unlike genes reactivating late. Early genes also show increased pluripotency transcription factor (TF) binding. We also reveal that histone deacetylases (HDACs) restrict XCR in reprogramming intermediates and that the severe hypoacetylation state of the inactive X Chromosome (Xi) persists until late reprogramming stages. Altogether, these results reveal the timing of transcriptional activation of monoallelically repressed genes during iPSC reprogramming, and suggest that allelic activation involves the combined action of chromatin topology, pluripotency TFs, and chromatin regulators. These findings are important for our understanding of gene silencing, maintenance of cell identity, reprogramming, and disease.


Subject(s)
Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , RNA, Long Noncoding/genetics , X Chromosome Inactivation/genetics , Animals , Chromatin/genetics , Female , Gene Silencing , Genes, X-Linked/genetics , Histone Deacetylases/genetics , Mice , Transcriptional Activation/genetics , X Chromosome/genetics
5.
PLoS Genet ; 14(8): e1007581, 2018 08.
Article in English | MEDLINE | ID: mdl-30080860

ABSTRACT

Cis-regulation plays an essential role in the control of gene expression, and is particularly complex and poorly understood for developmental genes, which are subject to multiple levels of modulation. In this study, we performed a global analysis of the cis-acting elements involved in the control of the zebrafish developmental gene krox20. krox20 encodes a transcription factor required for hindbrain segmentation and patterning, a morphogenetic process highly conserved during vertebrate evolution. Chromatin accessibility analysis reveals a cis-regulatory landscape that includes 6 elements participating in the control of initiation and autoregulatory aspects of krox20 hindbrain expression. Combining transgenic reporter analyses and CRISPR/Cas9-mediated mutagenesis, we assign precise functions to each of these 6 elements and provide a comprehensive view of krox20 cis-regulation. Three important features emerged. First, cooperation between multiple cis-elements plays a major role in the regulation. Cooperation can surprisingly combine synergy and redundancy, and is not restricted to transcriptional enhancer activity (for example, 4 distinct elements cooperate through different modes to maintain autoregulation). Second, several elements are unexpectedly versatile, which allows them to be involved in different aspects of control of gene expression. Third, comparative analysis of the elements and their activities in several vertebrate species reveals that this versatility is underlain by major plasticity across evolution, despite the high conservation of the gene expression pattern. These characteristics are likely to be of broad significance for developmental genes.


Subject(s)
Early Growth Response Protein 2/genetics , Gene Expression Regulation, Developmental , Rhombencephalon/metabolism , Zebrafish Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , CRISPR-Cas Systems , Chromatin/metabolism , Early Growth Response Protein 2/physiology , Enhancer Elements, Genetic , Evolution, Molecular , Genetic Loci , Morphogenesis/genetics , Transcriptional Activation , Zebrafish/embryology
6.
Nature ; 506(7487): 235-9, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24336202

ABSTRACT

CCAAT/enhancer binding protein-α (C/EBPα) induces transdifferentiation of B cells into macrophages at high efficiencies and enhances reprogramming into induced pluripotent stem (iPS) cells when co-expressed with the transcription factors Oct4 (Pou5f1), Sox2, Klf4 and Myc (hereafter called OSKM). However, how C/EBPα accomplishes these effects is unclear. Here we find that in mouse primary B cells transient C/EBPα expression followed by OSKM activation induces a 100-fold increase in iPS cell reprogramming efficiency, involving 95% of the population. During this conversion, pluripotency and epithelial-mesenchymal transition genes become markedly upregulated, and 60% of the cells express Oct4 within 2 days. C/EBPα acts as a 'path-breaker' as it transiently makes the chromatin of pluripotency genes more accessible to DNase I. C/EBPα also induces the expression of the dioxygenase Tet2 and promotes its translocation to the nucleus where it binds to regulatory regions of pluripotency genes that become demethylated after OSKM induction. In line with these findings, overexpression of Tet2 enhances OSKM-induced B-cell reprogramming. Because the enzyme is also required for efficient C/EBPα-induced immune cell conversion, our data indicate that Tet2 provides a mechanistic link between iPS cell reprogramming and B-cell transdifferentiation. The rapid iPS reprogramming approach described here should help to fully elucidate the process and has potential clinical applications.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Transdifferentiation , Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cells, Cultured , Cellular Reprogramming/genetics , Chromatin/genetics , Chromatin/metabolism , Cytosine/metabolism , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deoxyribonuclease I/metabolism , Dioxygenases , Epithelial-Mesenchymal Transition/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Up-Regulation/genetics
7.
Nucleic Acids Res ; 46(W1): W209-W214, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29722874

ABSTRACT

RSAT (Regulatory Sequence Analysis Tools) is a suite of modular tools for the detection and the analysis of cis-regulatory elements in genome sequences. Its main applications are (i) motif discovery, including from genome-wide datasets like ChIP-seq/ATAC-seq, (ii) motif scanning, (iii) motif analysis (quality assessment, comparisons and clustering), (iv) analysis of regulatory variations, (v) comparative genomics. Six public servers jointly support 10 000 genomes from all kingdoms. Six novel or refactored programs have been added since the 2015 NAR Web Software Issue, including updated programs to analyse regulatory variants (retrieve-variation-seq, variation-scan, convert-variations), along with tools to extract sequences from a list of coordinates (retrieve-seq-bed), to select motifs from motif collections (retrieve-matrix), and to extract orthologs based on Ensembl Compara (get-orthologs-compara). Three use cases illustrate the integration of new and refactored tools to the suite. This Anniversary update gives a 20-year perspective on the software suite. RSAT is well-documented and available through Web sites, SOAP/WSDL (Simple Object Access Protocol/Web Services Description Language) web services, virtual machines and stand-alone programs at http://www.rsat.eu/.


Subject(s)
Regulatory Sequences, Nucleic Acid , Software , Genetic Variation , Genomics/history , High-Throughput Nucleotide Sequencing/history , History, 20th Century , History, 21st Century , Internet , Nucleotide Motifs , Software/history
8.
PLoS Genet ; 13(7): e1006903, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28749941

ABSTRACT

Developmental genes can harbour multiple transcriptional enhancers that act simultaneously or in succession to achieve robust and precise spatiotemporal expression. However, the mechanisms underlying cooperation between cis-acting elements are poorly documented, notably in vertebrates. The mouse gene Krox20 encodes a transcription factor required for the specification of two segments (rhombomeres) of the developing hindbrain. In rhombomere 3, Krox20 is subject to direct positive feedback governed by an autoregulatory enhancer, element A. In contrast, a second enhancer, element C, distant by 70 kb, is active from the initiation of transcription independent of the presence of the KROX20 protein. Here, using both enhancer knock-outs and investigations of chromatin organisation, we show that element C possesses a dual activity: besides its classical enhancer function, it is also permanently required in cis to potentiate the autoregulatory activity of element A, by increasing its chromatin accessibility. This work uncovers a novel, asymmetrical, long-range mode of cooperation between cis-acting elements that might be essential to avoid promiscuous activation of positive autoregulatory elements.


Subject(s)
Early Growth Response Protein 1/genetics , Enhancer Elements, Genetic , Regulatory Elements, Transcriptional/genetics , Rhombencephalon/growth & development , Animals , Body Patterning/genetics , Chromatin/genetics , Early Growth Response Protein 1/biosynthesis , Gene Expression Regulation, Developmental , Mice, Knockout , Mutation , Rhombencephalon/metabolism , Sequence Homology, Nucleic Acid
9.
Proc Natl Acad Sci U S A ; 114(23): 5792-5799, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28584084

ABSTRACT

Blood cells are derived from a common set of hematopoietic stem cells, which differentiate into more specific progenitors of the myeloid and lymphoid lineages, ultimately leading to differentiated cells. This developmental process is controlled by a complex regulatory network involving cytokines and their receptors, transcription factors, and chromatin remodelers. Using public data and data from our own molecular genetic experiments (quantitative PCR, Western blot, EMSA) or genome-wide assays (RNA-sequencing, ChIP-sequencing), we have assembled a comprehensive regulatory network encompassing the main transcription factors and signaling components involved in myeloid and lymphoid development. Focusing on B-cell and macrophage development, we defined a qualitative dynamical model recapitulating cytokine-induced differentiation of common progenitors, the effect of various reported gene knockdowns, and the reprogramming of pre-B cells into macrophages induced by the ectopic expression of specific transcription factors. The resulting network model can be used as a template for the integration of new hematopoietic differentiation and transdifferentiation data to foster our understanding of lymphoid/myeloid cell-fate decisions.


Subject(s)
Cell Differentiation/genetics , Cell Transdifferentiation/genetics , Lymphocytes/cytology , Models, Biological , Myeloid Cells/cytology , B-Lymphocytes/cytology , Gene Regulatory Networks , Macrophages/cytology
10.
J Biol Chem ; 292(46): 18924-18936, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28900037

ABSTRACT

The transcription factor C/EBPα is essential for myeloid differentiation and is frequently dysregulated in acute myeloid leukemia. Although studied extensively, the precise regulation of its gene by upstream factors has remained largely elusive. Here, we investigated its transcriptional activation during myeloid differentiation. We identified an evolutionarily conserved octameric sequence, CCCAGCAG, ∼100 bases upstream of the CEBPA transcription start site, and demonstrated through mutational analysis that this sequence is crucial for C/EBPα expression. This sequence is present in the genes encoding C/EBPα in humans, rodents, chickens, and frogs and is also present in the promoters of other C/EBP family members. We identified that ZNF143, the human homolog of the Xenopus transcriptional activator STAF, specifically binds to this 8-bp sequence to activate C/EBPα expression in myeloid cells through a mechanism that is distinct from that observed in liver cells and adipocytes. Altogether, our data suggest that ZNF143 plays an important role in the expression of C/EBPα in myeloid cells.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/genetics , Myeloid Cells/cytology , Promoter Regions, Genetic , Trans-Activators/metabolism , Transcriptional Activation , Base Sequence , Cell Line , Conserved Sequence , Gene Expression Regulation, Developmental , Hematopoiesis , Humans , Myeloid Cells/metabolism , Protein Binding
11.
Nat Struct Mol Biol ; 30(8): 1216-1223, 2023 08.
Article in English | MEDLINE | ID: mdl-37291424

ABSTRACT

Subnuclear compartmentalization has been proposed to play an important role in gene regulation by segregating active and inactive parts of the genome in distinct physical and biochemical environments. During X chromosome inactivation (XCI), the noncoding Xist RNA coats the X chromosome, triggers gene silencing and forms a dense body of heterochromatin from which the transcription machinery appears to be excluded. Phase separation has been proposed to be involved in XCI, and might explain the exclusion of the transcription machinery by preventing its diffusion into the Xist-coated territory. Here, using quantitative fluorescence microscopy and single-particle tracking, we show that RNA polymerase II (RNAPII) freely accesses the Xist territory during the initiation of XCI. Instead, the apparent depletion of RNAPII is due to the loss of its chromatin stably bound fraction. These findings indicate that initial exclusion of RNAPII from the inactive X reflects the absence of actively transcribing RNAPII, rather than a consequence of putative physical compartmentalization of the inactive X heterochromatin domain.


Subject(s)
RNA Polymerase II , RNA, Long Noncoding , RNA Polymerase II/metabolism , Heterochromatin , X Chromosome/genetics , X Chromosome/metabolism , X Chromosome Inactivation , Chromatin , RNA, Untranslated/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
12.
Methods Mol Biol ; 2214: 295-316, 2021.
Article in English | MEDLINE | ID: mdl-32944918

ABSTRACT

The adaptation of Hi-C protocols to enable the investigation of chromosome organization in single cells opens new avenues to study the dynamics of this process during embryogenesis. However, the analysis of single-cell Hi-C data is not yet standardized and raises novel bioinformatic challenges. Here we describe a complete workflow for the analysis of single-cell Hi-C data, with a main focus on allele-specific analysis based on data obtained from hybrid embryos.


Subject(s)
Blastocyst/cytology , Computational Biology/methods , Mice/embryology , Single-Cell Analysis/methods , Alleles , Animals , Blastocyst/metabolism , Cell Cycle , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Male , Mice/genetics , Software , Workflow
13.
Curr Top Dev Biol ; 139: 205-238, 2020.
Article in English | MEDLINE | ID: mdl-32450961

ABSTRACT

Boolean approaches and extensions thereof are becoming increasingly popular to model signaling and regulatory networks, including those controlling cell differentiation, pattern formation and embryonic development. Here, we describe a logical modeling framework relying on three steps: the delineation of a regulatory graph, the specification of multilevel components, and the encoding of Boolean rules specifying the behavior of model components depending on the levels or activities of their regulators. Referring to a non-deterministic, asynchronous updating scheme, we present several complementary methods and tools enabling the computation of stable activity patterns, the verification of the reachability of such patterns, as well as the generation of mean temporal evolution curves and the computation of the probabilities to reach distinct activity patterns. We apply this logical framework to the regulatory network controlling T lymphocyte specification. This process involves cross-regulations between specific T cell regulatory factors and factors driving alternative differentiation pathways, which remain accessible during the early steps of thymocyte development. Many transcription factors needed for T cell specification are required in other hematopoietic differentiation pathways and are combined in a fine-tuned, time-dependent fashion to achieve T cell commitment. Using the software GINsim, we integrated current knowledge into a dynamical model, which recapitulates the main developmental steps from early progenitors entering the thymus up to T cell commitment, as well as the impact of various documented environmental and genetic perturbations. Our model analysis further enabled the identification of several knowledge gaps. The model, software and whole analysis workflow are provided in computer-readable and executable form to ensure reproducibility and ease extensions.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Models, Genetic , T-Lymphocytes/metabolism , Thymus Gland/metabolism , Animals , Computer Simulation , T-Lymphocytes/cytology , Thymocytes/cytology , Thymocytes/metabolism , Thymus Gland/cytology , Thymus Gland/embryology , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Nat Genet ; 50(2): 238-249, 2018 02.
Article in English | MEDLINE | ID: mdl-29335546

ABSTRACT

Chromosomal architecture is known to influence gene expression, yet its role in controlling cell fate remains poorly understood. Reprogramming of somatic cells into pluripotent stem cells (PSCs) by the transcription factors (TFs) OCT4, SOX2, KLF4 and MYC offers an opportunity to address this question but is severely limited by the low proportion of responding cells. We have recently developed a highly efficient reprogramming protocol that synchronously converts somatic into pluripotent stem cells. Here, we used this system to integrate time-resolved changes in genome topology with gene expression, TF binding and chromatin-state dynamics. The results showed that TFs drive topological genome reorganization at multiple architectural levels, often before changes in gene expression. Removal of locus-specific topological barriers can explain why pluripotency genes are activated sequentially, instead of simultaneously, during reprogramming. Together, our results implicate genome topology as an instructive force for implementing transcriptional programs and cell fate in mammals.


Subject(s)
Cellular Reprogramming/genetics , Chromatin Assembly and Disassembly/genetics , Chromosome Structures/genetics , Genome , Transcription Factors/physiology , Animals , Binding Sites/genetics , Cells, Cultured , Chromosome Structures/metabolism , Dosage Compensation, Genetic/genetics , Female , Gene Expression Regulation , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/physiology , Mice , Mice, Transgenic , Protein Binding , X Chromosome Inactivation/genetics
15.
Cell Stem Cell ; 23(5): 727-741.e9, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30220521

ABSTRACT

Here, we report DNA methylation and hydroxymethylation dynamics at nucleotide resolution using C/EBPα-enhanced reprogramming of B cells into induced pluripotent cells (iPSCs). We observed successive waves of hydroxymethylation at enhancers, concomitant with a decrease in DNA methylation, suggesting active demethylation. Consistent with this finding, ablation of the DNA demethylase Tet2 almost completely abolishes reprogramming. C/EBPα, Klf4, and Tfcp2l1 each interact with Tet2 and recruit the enzyme to specific DNA sites. During reprogramming, some of these sites maintain high levels of 5hmC, and enhancers and promoters of key pluripotency factors become demethylated as early as 1 day after Yamanaka factor induction. Surprisingly, methylation changes precede chromatin opening in distinct chromatin regions, including Klf4 bound sites, revealing a pioneer factor activity associated with alternation in DNA methylation. Rapid changes in hydroxymethylation similar to those in B cells were also observed during compound-accelerated reprogramming of fibroblasts into iPSCs, highlighting the generality of our observations.


Subject(s)
Cellular Reprogramming/genetics , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/genetics , Induced Pluripotent Stem Cells/cytology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Dioxygenases , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Male , Mice , Mice, Knockout
16.
Mol Cell Biol ; 37(4)2017 02 15.
Article in English | MEDLINE | ID: mdl-27920256

ABSTRACT

Short-term and long-term transcriptional memory is the phenomenon whereby the kinetics or magnitude of gene induction is enhanced following a prior induction period. Short-term memory persists within one cell generation or in postmitotic cells, while long-term memory can survive multiple rounds of cell division. We have developed a tissue culture model to study the epigenetic basis for long-term transcriptional memory (LTTM) and subsequently used this model to better understand the epigenetic mechanisms that enable heritable memory of temporary stimuli. We find that a pulse of transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) induces LTTM on a subset of target genes that survives nine cell divisions. The chromatin landscape at genes that acquire LTTM is more repressed than at those genes that do not exhibit memory, akin to a latent state. We show through chromatin immunoprecipitation (ChIP) and chemical inhibitor studies that RNA polymerase II (Pol II) elongation is important for establishing memory in this model but that Pol II itself is not retained as part of the memory mechanism. More generally, our work reveals that a transcription factor involved in lineage specification can induce LTTM and that failure to rerepress chromatin is one epigenetic mechanism underlying transcriptional memory.


Subject(s)
Chromatin/metabolism , Transcription Factors/metabolism , Transcription, Genetic , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/drug effects , Histones/metabolism , Lipopolysaccharides/pharmacology , Lysine/metabolism , Methylation/drug effects , Protein Binding/drug effects , RNA Polymerase II/metabolism , Transcription, Genetic/drug effects
17.
Cell Stem Cell ; 21(6): 791-805.e9, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29174331

ABSTRACT

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.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcription Factors/metabolism , Cell Line , Humans , Transcription Factors/genetics
18.
Nat Cell Biol ; 18(4): 371-81, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26974661

ABSTRACT

Reprogramming somatic cells into induced pluripotent stem cells (iPSCs) is typically inefficient and has been explained by elite-cell and stochastic models. We recently reported that B cells exposed to a pulse of C/EBPα (Bα' cells) behave as elite cells, in that they can be rapidly and efficiently reprogrammed into iPSCs by the Yamanaka factors OSKM. Here we show that C/EBPα post-transcriptionally increases the abundance of several hundred proteins, including Lsd1, Hdac1, Brd4, Med1 and Cdk9, components of chromatin-modifying complexes present at super-enhancers. Lsd1 was found to be required for B cell gene silencing and Brd4 for the activation of the pluripotency program. C/EBPα also promotes chromatin accessibility in pluripotent cells and upregulates Klf4 by binding to two haematopoietic enhancers. Bα' cells share many properties with granulocyte/macrophage progenitors, naturally occurring elite cells that are obligate targets for leukaemic transformation, whose formation strictly requires C/EBPα.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/genetics , Cellular Reprogramming/genetics , Histone Demethylases/genetics , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Transcription Factors/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , B-Lymphocytes/metabolism , Blotting, Western , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Line , Cells, Cultured , Female , Gene Expression Profiling/methods , Gene Ontology , HEK293 Cells , Histone Demethylases/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/metabolism , Nuclear Proteins/metabolism , Proteomics/methods , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , Up-Regulation
19.
Stem Cell Reports ; 5(2): 232-47, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26235892

ABSTRACT

Transcription-factor-induced somatic cell conversions are highly relevant for both basic and clinical research yet their mechanism is not fully understood and it is unclear whether they reflect normal differentiation processes. Here we show that during pre-B-cell-to-macrophage transdifferentiation, C/EBPα binds to two types of myeloid enhancers in B cells: pre-existing enhancers that are bound by PU.1, providing a platform for incoming C/EBPα; and de novo enhancers that are targeted by C/EBPα, acting as a pioneer factor for subsequent binding by PU.1. The order of factor binding dictates the upregulation kinetics of nearby genes. Pre-existing enhancers are broadly active throughout the hematopoietic lineage tree, including B cells. In contrast, de novo enhancers are silent in most cell types except in myeloid cells where they become activated by C/EBP factors. Our data suggest that C/EBPα recapitulates physiological developmental processes by short-circuiting two macrophage enhancer pathways in pre-B cells.


Subject(s)
B-Lymphocytes/metabolism , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Transdifferentiation , Myeloid Cells/metabolism , Myelopoiesis , Proto-Oncogene Proteins c-ets/metabolism , B-Lymphocytes/cytology , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Line , Humans , Myeloid Cells/cytology , Proto-Oncogene Proteins c-ets/genetics
20.
Sci Data ; 1: 140008, 2014.
Article in English | MEDLINE | ID: mdl-25977766

ABSTRACT

The reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) is lengthy and inefficient. The development of a reprogramming system that allows rapid and synchronous reprogramming to pluripotency is imperative for understanding the mechanism of iPSC formation and for future therapeutic applications. We have recently reported that a short expression in mouse primary B cells of the transcription factor C/EBPα before the induction of pluripotency factors increases the iPSC reprogramming efficiency >100-fold, involving 95% of the cells within a week. Here we present a dataset containing the time course of gene expression during this process as determined by microarray and RNA-seq techniques.


Subject(s)
Cellular Reprogramming/genetics , Gene Expression Profiling , Induced Pluripotent Stem Cells , Animals , B-Lymphocytes/cytology , B-Lymphocytes/physiology , CCAAT-Enhancer-Binding Proteins/genetics , Cell Differentiation/genetics , Gene Expression , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Mice , Microarray Analysis
SELECTION OF CITATIONS
SEARCH DETAIL