Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Curr Biol ; 34(3): 505-518.e6, 2024 02 05.
Article in English | MEDLINE | ID: mdl-38215744

ABSTRACT

Germ cells are essential to sexual reproduction. Across the animal kingdom, extracellular signaling isoprenoids, such as retinoic acids (RAs) in vertebrates and juvenile hormones (JHs) in invertebrates, facilitate multiple processes in reproduction. Here we investigated the role of these potent signaling molecules in embryonic germ cell development, using JHs in Drosophila melanogaster as a model system. In contrast to their established endocrine roles during larval and adult germline development, we found that JH signaling acts locally during embryonic development. Using an in vivo biosensor, we observed active JH signaling first within and near primordial germ cells (PGCs) as they migrate to the developing gonad. Through in vivo and in vitro assays, we determined that JHs are both necessary and sufficient for PGC migration. Analysis into the mechanisms of this newly uncovered paracrine JH function revealed that PGC migration was compromised when JHs were decreased or increased, suggesting that specific titers or spatiotemporal JH dynamics are required for robust PGC colonization of the gonad. Compromised PGC migration can impair fertility and cause germ cell tumors in many species, including humans. In mammals, retinoids have many roles in development and reproduction. We found that like JHs in Drosophila, RA was sufficient to impact mouse PGC migration in vitro. Together, our study reveals a previously unanticipated role of isoprenoids as local effectors of pre-gonadal PGC development and suggests a broadly shared mechanism in PGC migration.


Subject(s)
Drosophila melanogaster , Juvenile Hormones , Humans , Mice , Animals , Germ Cells , Drosophila , Gonads , Terpenes , Cell Movement , Mammals
2.
Nat Struct Mol Biol ; 31(1): 125-140, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38053013

ABSTRACT

Mammalian embryogenesis commences with two pivotal and binary cell fate decisions that give rise to three essential lineages: the trophectoderm, the epiblast and the primitive endoderm. Although key signaling pathways and transcription factors that control these early embryonic decisions have been identified, the non-coding regulatory elements through which transcriptional regulators enact these fates remain understudied. Here, we characterize, at a genome-wide scale, enhancer activity and 3D connectivity in embryo-derived stem cell lines that represent each of the early developmental fates. We observe extensive enhancer remodeling and fine-scale 3D chromatin rewiring among the three lineages, which strongly associate with transcriptional changes, although distinct groups of genes are irresponsive to topological changes. In each lineage, a high degree of connectivity, or 'hubness', positively correlates with levels of gene expression and enriches for cell-type specific and essential genes. Genes within 3D hubs also show a significantly stronger probability of coregulation across lineages compared to genes in linear proximity or within the same contact domains. By incorporating 3D chromatin features, we build a predictive model for transcriptional regulation (3D-HiChAT) that outperforms models using only 1D promoter or proximal variables to predict levels and cell-type specificity of gene expression. Using 3D-HiChAT, we identify, in silico, candidate functional enhancers and hubs in each cell lineage, and with CRISPRi experiments, we validate several enhancers that control gene expression in their respective lineages. Our study identifies 3D regulatory hubs associated with the earliest mammalian lineages and describes their relationship to gene expression and cell identity, providing a framework to comprehensively understand lineage-specific transcriptional behaviors.


Subject(s)
Gene Expression Regulation, Developmental , Regulatory Sequences, Nucleic Acid , Animals , Promoter Regions, Genetic/genetics , Chromatin/genetics , Cell Lineage/genetics , Gene Expression , Enhancer Elements, Genetic/genetics , Mammals/genetics
3.
bioRxiv ; 2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37577543

ABSTRACT

Mammalian embryogenesis commences with two pivotal and binary cell fate decisions that give rise to three essential lineages, the trophectoderm (TE), the epiblast (EPI) and the primitive endoderm (PrE). Although key signaling pathways and transcription factors that control these early embryonic decisions have been identified, the non-coding regulatory elements via which transcriptional regulators enact these fates remain understudied. To address this gap, we have characterized, at a genome-wide scale, enhancer activity and 3D connectivity in embryo-derived stem cell lines that represent each of the early developmental fates. We observed extensive enhancer remodeling and fine-scale 3D chromatin rewiring among the three lineages, which strongly associate with transcriptional changes, although there are distinct groups of genes that are irresponsive to topological changes. In each lineage, a high degree of connectivity or "hubness" positively correlates with levels of gene expression and enriches for cell-type specific and essential genes. Genes within 3D hubs also show a significantly stronger probability of coregulation across lineages, compared to genes in linear proximity or within the same contact domains. By incorporating 3D chromatin features, we build a novel predictive model for transcriptional regulation (3D-HiChAT), which outperformed models that use only 1D promoter or proximal variables in predicting levels and cell-type specificity of gene expression. Using 3D-HiChAT, we performed genome-wide in silico perturbations to nominate candidate functional enhancers and hubs in each cell lineage, and with CRISPRi experiments we validated several novel enhancers that control expression of one or more genes in their respective lineages. Our study comprehensively identifies 3D regulatory hubs associated with the earliest mammalian lineages and describes their relationship to gene expression and cell identity, providing a framework to understand lineage-specific transcriptional behaviors.

4.
Cell Rep ; 38(11): 110524, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35294876

ABSTRACT

In pluripotent cells, a delicate activation-repression balance maintains pro-differentiation genes ready for rapid activation. The identity of transcription factors (TFs) that specifically repress pro-differentiation genes remains obscure. By targeting ∼1,700 TFs with CRISPR loss-of-function screen, we found that ZBTB11 and ZFP131 are required for embryonic stem cell (ESC) pluripotency. ESCs without ZBTB11 or ZFP131 lose colony morphology, reduce proliferation rate, and upregulate transcription of genes associated with three germ layers. ZBTB11 and ZFP131 bind proximally to pro-differentiation genes. ZBTB11 or ZFP131 loss leads to an increase in H3K4me3, negative elongation factor (NELF) complex release, and concomitant transcription at associated genes. Together, our results suggest that ZBTB11 and ZFP131 maintain pluripotency by preventing premature expression of pro-differentiation genes and present a generalizable framework to maintain cellular potency.


Subject(s)
Embryonic Stem Cells , Pluripotent Stem Cells , Animals , Humans , Mice , Cell Differentiation/genetics , CRISPR-Cas Systems , Embryonic Stem Cells/metabolism , Germ Layers/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Dev Cell ; 56(22): 3052-3065.e5, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34710357

ABSTRACT

Loss of imprinting (LOI) results in severe developmental defects, but the mechanisms preventing LOI remain incompletely understood. Here, we dissect the functional components of the imprinting control region of the essential Dlk1-Dio3 locus (called IG-DMR) in pluripotent stem cells. We demonstrate that the IG-DMR consists of two antagonistic elements: a paternally methylated CpG island that prevents recruitment of TET dioxygenases and a maternally unmethylated non-canonical enhancer that ensures expression of the Gtl2 lncRNA by counteracting de novo DNA methyltransferases. Genetic or epigenetic editing of these elements leads to distinct LOI phenotypes with characteristic alternations of allele-specific gene expression, DNA methylation, and 3D chromatin topology. Although repression of the Gtl2 promoter results in dysregulated imprinting, the stability of LOI phenotypes depends on the IG-DMR, suggesting a functional hierarchy. These findings establish the IG-DMR as a bipartite control element that maintains imprinting by allele-specific restriction of the DNA (de)methylation machinery.


Subject(s)
Alleles , Calcium-Binding Proteins/genetics , DNA Methylation/genetics , Intercellular Signaling Peptides and Proteins/genetics , Animals , Chromosomes/genetics , Genomic Imprinting/genetics , Iodide Peroxidase/genetics , Mice , Promoter Regions, Genetic/genetics , RNA, Long Noncoding/genetics
6.
Stem Cell Reports ; 15(6): 1233-1245, 2020 12 08.
Article in English | MEDLINE | ID: mdl-32976761

ABSTRACT

Methylation of histone 3 at lysine 9 (H3K9) constitutes a roadblock for cellular reprogramming. Interference with methyltransferases or activation of demethylases by the cofactor ascorbic acid (AA) facilitates the derivation of induced pluripotent stem cells (iPSCs), but possible interactions between specific methyltransferases and AA treatment remain insufficiently explored. We show that chemical inhibition of the methyltransferases EHMT1 and EHMT2 counteracts iPSC formation in an enhanced reprogramming system in the presence of AA, an effect that is dependent on EHMT1. EHMT inhibition during enhanced reprogramming is associated with rapid loss of H3K9 dimethylation, inefficient downregulation of somatic genes, and failed mesenchymal-to-epithelial transition. Furthermore, transient EHMT inhibition during reprogramming yields iPSCs that fail to efficiently give rise to viable mice upon blastocyst injection. Our observations establish novel functions of H3K9 methyltransferases and suggest that a functional balance between AA-stimulated enzymes and EHMTs supports efficient and less error-prone iPSC reprogramming to pluripotency.


Subject(s)
Cellular Reprogramming , Histone-Lysine N-Methyltransferase/metabolism , Induced Pluripotent Stem Cells/enzymology , Animals , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Induced Pluripotent Stem Cells/cytology , Methylation , Mice
7.
Cell Rep ; 30(11): 3597-3604.e3, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32187532

ABSTRACT

Cultured pluripotent cells accumulate detrimental chromatin alterations, including DNA methylation changes at imprinted genes known as loss of imprinting (LOI). Although the occurrence of LOI is considered a stochastic phenomenon, here we document a genetic determinant that segregates mouse pluripotent cells into stable and unstable cell lines. Unstable lines exhibit hypermethylation at Dlk1-Dio3 and other imprinted loci, in addition to impaired developmental potential. Stimulation of demethylases by ascorbic acid prevents LOI and loss of developmental potential. Susceptibility to LOI greatly differs between commonly used mouse strains, which we use to map a causal region on chromosome 13 with quantitative trait locus (QTL) analysis. Our observations identify a strong genetic determinant of locus-specific chromatin abnormalities in pluripotent cells and provide a non-invasive way to suppress them. This highlights the importance of considering genetics in conjunction with culture conditions for assuring the quality of pluripotent cells for biomedical applications.


Subject(s)
Chromosomes, Mammalian/genetics , Genetic Loci , Genomic Imprinting , Pluripotent Stem Cells/metabolism , Animals , Ascorbic Acid/pharmacology , Calcium-Binding Proteins/genetics , Cell Line , DNA Methylation/genetics , Embryonic Development/drug effects , Epigenesis, Genetic , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/metabolism , Quantitative Trait Loci/genetics
8.
Genome Biol ; 20(1): 248, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31752933

ABSTRACT

Activation of regulatory elements is thought to be inversely correlated with DNA methylation levels. However, it is difficult to determine whether DNA methylation is compatible with chromatin accessibility or transcription factor (TF) binding if assays are performed separately. We developed a fast, low-input, low sequencing depth method, EpiMethylTag, that combines ATAC-seq or ChIP-seq (M-ATAC or M-ChIP) with bisulfite conversion, to simultaneously examine accessibility/TF binding and methylation on the same DNA. Here we demonstrate that EpiMethylTag can be used to study the functional interplay between chromatin accessibility and TF binding (CTCF and KLF4) at methylated sites.


Subject(s)
Chromatin Immunoprecipitation Sequencing , DNA Methylation , Genomics/methods , Animals , Chromatin/metabolism , Humans , Kruppel-Like Factor 4 , Transcription Factors/metabolism
9.
Nat Cell Biol ; 21(10): 1179-1190, 2019 10.
Article in English | MEDLINE | ID: mdl-31548608

ABSTRACT

Cell fate transitions are accompanied by global transcriptional, epigenetic and topological changes driven by transcription factors, as is exemplified by reprogramming somatic cells to pluripotent stem cells through the expression of OCT4, KLF4, SOX2 and cMYC. How transcription factors orchestrate the complex molecular changes around their target gene loci remains incompletely understood. Here, using KLF4 as a paradigm, we provide a transcription-factor-centric view of chromatin reorganization and its association with three-dimensional enhancer rewiring and transcriptional changes during the reprogramming of mouse embryonic fibroblasts to pluripotent stem cells. Inducible depletion of KLF factors in PSCs caused a genome-wide decrease in enhancer connectivity, whereas disruption of individual KLF4 binding sites within pluripotent-stem-cell-specific enhancers was sufficient to impair enhancer-promoter contacts and reduce the expression of associated genes. Our study provides an integrative view of the complex activities of a lineage-specifying transcription factor and offers novel insights into the nature of the molecular events that follow transcription factor binding.


Subject(s)
Cellular Reprogramming/genetics , Chromatin Assembly and Disassembly/genetics , Enhancer Elements, Genetic , Kruppel-Like Transcription Factors/metabolism , Mouse Embryonic Stem Cells/metabolism , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Kruppel-Like Factor 4 , Male , Mice , Pluripotent Stem Cells/metabolism
10.
Cell Stem Cell ; 25(3): 297-298, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31491390

ABSTRACT

An example of the peer review process for "Distinct molecular trajectories converge to induce naive pluripotency" (Stuart et al., 2019) is presented here.

11.
Curr Opin Genet Dev ; 52: 77-85, 2018 10.
Article in English | MEDLINE | ID: mdl-29925040

ABSTRACT

The discovery of induced pluripotent stem cells (iPSCs) has solidified the concept of transcription factors as major players in controlling cell identity and provided a tractable tool to study how somatic cell identity can be dismantled and pluripotency established. A number of landmark studies have established hallmarks and roadmaps of iPSC formation by describing relative kinetics of transcriptional, protein and epigenetic changes, including alterations in DNA methylation and histone modifications. Recently, technological advancements such as single-cell analyses, high-resolution genome-wide chromatin assays and more efficient reprogramming systems have been used to challenge and refine our understanding of the reprogramming process. Here, we will outline novel insights into the molecular mechanisms underlying iPSC formation, focusing on how the core reprogramming factors OCT4, KLF4, SOX2 and MYC (OKSM) drive changes in gene expression, chromatin state and 3D genome topology. In addition, we will discuss unexpected consequences of reprogramming factor expression in in vitro and in vivo systems that may point towards new applications of iPSC technology.


Subject(s)
Cellular Reprogramming/genetics , Chromatin/genetics , DNA Methylation/genetics , Induced Pluripotent Stem Cells/cytology , Epigenesis, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4
12.
Cell Rep ; 22(4): 876-884, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29420174

ABSTRACT

The ability of induced pluripotent stem cells (iPSCs) to differentiate into all adult cell types makes them attractive for research and regenerative medicine; however, it remains unknown when and how this capacity is established. We characterized the acquisition of developmental pluripotency in a suitable reprogramming system to show that iPSCs prior to passaging become capable of generating all tissues upon injection into preimplantation embryos. The developmental potential of nascent iPSCs is comparable to or even surpasses that of established pluripotent cells. Further functional assays and genome-wide molecular analyses suggest that cells acquiring developmental pluripotency exhibit a unique combination of properties that distinguish them from canonical naive and primed pluripotency states. These include reduced clonal self-renewal potential and the elevated expression of differentiation-associated transcriptional regulators. Our observations close a gap in the understanding of induced pluripotency and provide an improved roadmap of cellular reprogramming with ramifications for the use of iPSCs.


Subject(s)
Gene Expression Regulation/genetics , Induced Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation , Humans , Mice
13.
Nat Cell Biol ; 19(4): 341-351, 2017 04.
Article in English | MEDLINE | ID: mdl-28319092

ABSTRACT

Self-renewing naive mouse embryonic stem cells (mESCs) contain few mitochondria, which increase in number and volume at the onset of differentiation. KBP (encoded by Kif1bp) is an interactor of the mitochondrial-associated kinesin Kif1Bα. We found that TDH, responsible for mitochondrial production of acetyl-CoA in mESCs, and the acetyltransferase GCN5L1 cooperate to acetylate Lys501 in KBP, allowing its recognition by and degradation via Fbxo15, an F-box protein transcriptionally controlled by the pluripotency core factors and repressed following differentiation. Defects in KBP degradation in mESCs result in an unscheduled increase in mitochondrial biogenesis, enhanced respiration and ROS production, and inhibition of cell proliferation. Silencing of Kif1Bα reverts the aberrant increase in mitochondria induced by KBP stabilization. Notably, following differentiation, Kif1bp-/- mESCs display impaired expansion of the mitochondrial mass and form smaller embryoid bodies. Thus, KBP proteolysis limits the accumulation of mitochondria in mESCs to preserve their optimal fitness, whereas KBP accumulation promotes mitochondrial biogenesis in differentiating cells.


Subject(s)
Alcohol Oxidoreductases/metabolism , Carrier Proteins/metabolism , F-Box Proteins/metabolism , Mitochondria/metabolism , Mouse Embryonic Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Organelle Biogenesis , Acetylation , Animals , Cell Differentiation , Cell Proliferation , Cell Respiration , Cell Self Renewal , HEK293 Cells , Humans , Kinesins/metabolism , Lentivirus/metabolism , Mice , Mitochondrial Proteins , Mutant Proteins/metabolism , Protein Binding , Protein Stability , Proteolysis , RNA, Small Interfering/metabolism , Substrate Specificity
14.
Cell Rep ; 17(12): 3395-3406, 2016 12 20.
Article in English | MEDLINE | ID: mdl-28009305

ABSTRACT

Reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) is typically an inefficient and asynchronous process. A variety of technological efforts have been made to accelerate and/or synchronize this process. To define a unified framework to study and compare the dynamics of reprogramming under different conditions, we developed an in silico analysis platform based on mathematical modeling. Our approach takes into account the variability in experimental results stemming from probabilistic growth and death of cells and potentially heterogeneous reprogramming rates. We suggest that reprogramming driven by the Yamanaka factors alone is a more heterogeneous process, possibly due to cell-specific reprogramming rates, which could be homogenized by the addition of additional factors. We validated our approach using publicly available reprogramming datasets, including data on early reprogramming dynamics as well as cell count data, and thus we demonstrated the general utility and predictive power of our methodology for investigating reprogramming and other cell fate change systems.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells , Models, Theoretical , Computer Simulation , Humans , Models, Statistical
15.
Development ; 143(22): 4161-4166, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27729406

ABSTRACT

Genomic imprinting results in the monoallelic expression of genes that encode important regulators of growth and proliferation. Dysregulation of imprinted genes, such as those within the Dlk1-Dio3 locus, is associated with developmental syndromes and specific diseases. Our ability to interrogate causes of imprinting instability has been hindered by the absence of suitable model systems. Here, we describe a Dlk1 knock-in reporter mouse that enables single-cell visualization of allele-specific expression and prospective isolation of cells, simultaneously. We show that this 'imprinting reporter mouse' can be used to detect tissue-specific Dlk1 expression patterns in developing embryos. We also apply this system to pluripotent cell culture and demonstrate that it faithfully indicates DNA methylation changes induced upon cellular reprogramming. Finally, the reporter system reveals the role of elevated oxygen levels in eroding imprinted Dlk1 expression during prolonged culture and in vitro differentiation. The possibility to study allele-specific expression in different contexts makes our reporter system a useful tool to dissect the regulation of genomic imprinting in normal development and disease.


Subject(s)
Embryonic Development/genetics , Genes, Reporter , Genomic Imprinting , Genomic Instability/genetics , Intercellular Signaling Peptides and Proteins/genetics , Pluripotent Stem Cells/metabolism , Animals , Calcium-Binding Proteins , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Embryo, Mammalian , Female , Genetic Loci , Genomic Imprinting/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Genetic , Oxygen/pharmacology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects
16.
Nat Cell Biol ; 18(11): 1127-1138, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27749823

ABSTRACT

Pluripotent embryonic stem cells (ESCs) self-renew or differentiate into all tissues of the developing embryo and cell-specification factors are necessary to balance gene expression. Here we delineate the function of the PHD-finger protein 5a (Phf5a) in ESC self-renewal and ascribe its role in regulating pluripotency, cellular reprogramming and myoblast specification. We demonstrate that Phf5a is essential for maintaining pluripotency, since depleted ESCs exhibit hallmarks of differentiation. Mechanistically, we attribute Phf5a function to the stabilization of the Paf1 transcriptional complex and control of RNA polymerase II elongation on pluripotency loci. Apart from an ESC-specific factor, we demonstrate that Phf5a controls differentiation of adult myoblasts. Our findings suggest a potent mode of regulation by Phf5a in stem cells, which directs their transcriptional programme, ultimately regulating maintenance of pluripotency and cellular reprogramming.


Subject(s)
Carrier Proteins/genetics , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Mouse Embryonic Stem Cells/cytology , Myoblasts/cytology , Pluripotent Stem Cells/cytology , Transcription, Genetic , Aging , Animals , Cell Line , Cell Proliferation/genetics , DNA-Binding Proteins , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Inbred C57BL , RNA-Binding Proteins , Trans-Activators
17.
Nature ; 534(7607): 387-90, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27281218

ABSTRACT

Developmental specification of germ cells lies at the heart of inheritance, as germ cells contain all of the genetic and epigenetic information transmitted between generations. The critical developmental event distinguishing germline from somatic lineages is the differentiation of primordial germ cells (PGCs), precursors of sex-specific gametes that produce an entire organism upon fertilization. Germ cells toggle between uni- and pluripotent states as they exhibit their own 'latent' form of pluripotency. For example, PGCs express a number of transcription factors in common with embryonic stem (ES) cells, including OCT4 (encoded by Pou5f1), SOX2, NANOG and PRDM14 (refs 2, 3, 4). A biochemical mechanism by which these transcription factors converge on chromatin to produce the dramatic rearrangements underlying ES-cell- and PGC-specific transcriptional programs remains poorly understood. Here we identify a novel co-repressor protein, CBFA2T2, that regulates pluripotency and germline specification in mice. Cbfa2t2(-/-) mice display severe defects in PGC maturation and epigenetic reprogramming. CBFA2T2 forms a biochemical complex with PRDM14, a germline-specific transcription factor. Mechanistically, CBFA2T2 oligomerizes to form a scaffold upon which PRDM14 and OCT4 are stabilized on chromatin. Thus, in contrast to the traditional 'passenger' role of a co-repressor, CBFA2T2 functions synergistically with transcription factors at the crossroads of the fundamental developmental plasticity between uni- and pluripotency.


Subject(s)
Germ Cells/metabolism , Pluripotent Stem Cells/metabolism , Repressor Proteins/metabolism , Animals , Cell Line , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/genetics , Female , Gene Expression Regulation, Developmental/genetics , Germ Cells/cytology , Germ Cells/pathology , Humans , Male , Mice , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Protein Binding , RNA-Binding Proteins , Repressor Proteins/chemistry , Repressor Proteins/deficiency , Repressor Proteins/genetics , Transcription Factors/metabolism
18.
Nature ; 528(7581): 218-24, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26659182

ABSTRACT

Cellular differentiation involves profound remodelling of chromatic landscapes, yet the mechanisms by which somatic cell identity is subsequently maintained remain incompletely understood. To further elucidate regulatory pathways that safeguard the somatic state, we performed two comprehensive RNA interference (RNAi) screens targeting chromatin factors during transcription-factor-mediated reprogramming of mouse fibroblasts to induced pluripotent stem cells (iPS cells). Subunits of the chromatin assembly factor-1 (CAF-1) complex, including Chaf1a and Chaf1b, emerged as the most prominent hits from both screens, followed by modulators of lysine sumoylation and heterochromatin maintenance. Optimal modulation of both CAF-1 and transcription factor levels increased reprogramming efficiency by several orders of magnitude and facilitated iPS cell formation in as little as 4 days. Mechanistically, CAF-1 suppression led to a more accessible chromatin structure at enhancer elements early during reprogramming. These changes were accompanied by a decrease in somatic heterochromatin domains, increased binding of Sox2 to pluripotency-specific targets and activation of associated genes. Notably, suppression of CAF-1 also enhanced the direct conversion of B cells into macrophages and fibroblasts into neurons. Together, our findings reveal the histone chaperone CAF-1 to be a novel regulator of somatic cell identity during transcription-factor-induced cell-fate transitions and provide a potential strategy to modulate cellular plasticity in a regenerative setting.


Subject(s)
Cellular Reprogramming/genetics , Chromatin Assembly Factor-1/metabolism , Animals , Cells, Cultured , Chromatin/metabolism , Chromatin Assembly Factor-1/antagonists & inhibitors , Chromatin Assembly Factor-1/genetics , Gene Expression Regulation/genetics , Heterochromatin/metabolism , Mice , Nucleosomes/metabolism , RNA Interference , Transduction, Genetic
19.
Cell Stem Cell ; 16(1): 9-10, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25575077

ABSTRACT

A series of five related publications describe an alternative pluripotent state that is dependent on continuous high levels of exogenous reprogramming factor expression. A comprehensive effort to molecularly compare the acquisition of this state to induced pluripotency aims at providing new insights into the mechanisms underlying cellular reprogramming.

20.
Stem Cell Reports ; 3(4): 574-84, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25358786

ABSTRACT

The differentiated state of somatic cells provides barriers for the derivation of induced pluripotent stem cells (iPSCs). To address why some cell types reprogram more readily than others, we studied the effect of combined modulation of cellular signaling pathways. Surprisingly, inhibition of transforming growth factor ß (TGF-ß) together with activation of Wnt signaling in the presence of ascorbic acid allows >80% of murine fibroblasts to acquire pluripotency after 1 week of reprogramming factor expression. In contrast, hepatic and blood progenitors predominantly required only TGF-ß inhibition or canonical Wnt activation, respectively, to reprogram at efficiencies approaching 100%. Strikingly, blood progenitors reactivated endogenous pluripotency loci in a highly synchronous manner, and we demonstrate that expression of specific chromatin-modifying enzymes and reduced TGF-ß/mitogen-activated protein (MAP) kinase activity are intrinsic properties associated with the unique reprogramming response of these cells. Our observations define cell-type-specific requirements for the rapid and synchronous reprogramming of somatic cells.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , MAP Kinase Signaling System , Wnt Signaling Pathway , Animals , Ascorbic Acid/pharmacology , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Mice , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...