Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 544
Filter
Add more filters

Publication year range
1.
Cell ; 186(5): 957-974.e28, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36812912

ABSTRACT

Bats are distinctive among mammals due to their ability to fly, use laryngeal echolocation, and tolerate viruses. However, there are currently no reliable cellular models for studying bat biology or their response to viral infections. Here, we created induced pluripotent stem cells (iPSCs) from two species of bats: the wild greater horseshoe bat (Rhinolophus ferrumequinum) and the greater mouse-eared bat (Myotis myotis). The iPSCs from both bat species showed similar characteristics and had a gene expression profile resembling that of cells attacked by viruses. They also had a high number of endogenous viral sequences, particularly retroviruses. These results suggest that bats have evolved mechanisms to tolerate a large load of viral sequences and may have a more intertwined relationship with viruses than previously thought. Further study of bat iPSCs and their differentiated progeny will provide insights into bat biology, virus host relationships, and the molecular basis of bats' special traits.


Subject(s)
Chiroptera , Pluripotent Stem Cells , Virus Diseases , Viruses , Animals , Viruses/genetics , Transcriptome , Phylogeny
2.
Cell ; 184(1): 207-225.e24, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33333019

ABSTRACT

Regulation of biological processes typically incorporates mechanisms that initiate and terminate the process and, where understood, these mechanisms often involve feedback control. Regulation of transcription is a fundamental cellular process where the mechanisms involved in initiation have been studied extensively, but those involved in arresting the process are poorly understood. Modeling of the potential roles of RNA in transcriptional control suggested a non-equilibrium feedback control mechanism where low levels of RNA promote condensates formed by electrostatic interactions whereas relatively high levels promote dissolution of these condensates. Evidence from in vitro and in vivo experiments support a model where RNAs produced during early steps in transcription initiation stimulate condensate formation, whereas the burst of RNAs produced during elongation stimulate condensate dissolution. We propose that transcriptional regulation incorporates a feedback mechanism whereby transcribed RNAs initially stimulate but then ultimately arrest the process.


Subject(s)
Feedback, Physiological , RNA/genetics , Transcription, Genetic , Animals , Mediator Complex/metabolism , Mice , Models, Biological , Mouse Embryonic Stem Cells/metabolism , RNA/biosynthesis , Static Electricity
3.
Cell ; 172(5): 979-992.e6, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29456084

ABSTRACT

Fragile X syndrome (FXS), the most common genetic form of intellectual disability in males, is caused by silencing of the FMR1 gene associated with hypermethylation of the CGG expansion mutation in the 5' UTR of FMR1 in FXS patients. Here, we applied recently developed DNA methylation editing tools to reverse this hypermethylation event. Targeted demethylation of the CGG expansion by dCas9-Tet1/single guide RNA (sgRNA) switched the heterochromatin status of the upstream FMR1 promoter to an active chromatin state, restoring a persistent expression of FMR1 in FXS iPSCs. Neurons derived from methylation-edited FXS iPSCs rescued the electrophysiological abnormalities and restored a wild-type phenotype upon the mutant neurons. FMR1 expression in edited neurons was maintained in vivo after engrafting into the mouse brain. Finally, demethylation of the CGG repeats in post-mitotic FXS neurons also reactivated FMR1. Our data establish that demethylation of the CGG expansion is sufficient for FMR1 reactivation, suggesting potential therapeutic strategies for FXS.


Subject(s)
DNA Methylation/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Gene Editing , Neurons/pathology , Animals , CRISPR-Associated Protein 9/metabolism , Epigenesis, Genetic , HEK293 Cells , Heterochromatin/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Kinetics , Male , Mice , Neurons/metabolism , Phenotype , Promoter Regions, Genetic , RNA, Guide, Kinetoplastida/metabolism , Trinucleotide Repeat Expansion/genetics
4.
Cell ; 175(7): 1842-1855.e16, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30449618

ABSTRACT

Gene expression is controlled by transcription factors (TFs) that consist of DNA-binding domains (DBDs) and activation domains (ADs). The DBDs have been well characterized, but little is known about the mechanisms by which ADs effect gene activation. Here, we report that diverse ADs form phase-separated condensates with the Mediator coactivator. For the OCT4 and GCN4 TFs, we show that the ability to form phase-separated droplets with Mediator in vitro and the ability to activate genes in vivo are dependent on the same amino acid residues. For the estrogen receptor (ER), a ligand-dependent activator, we show that estrogen enhances phase separation with Mediator, again linking phase separation with gene activation. These results suggest that diverse TFs can interact with Mediator through the phase-separating capacity of their ADs and that formation of condensates with Mediator is involved in gene activation.


Subject(s)
Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Receptors, Estrogen/metabolism , Transcriptional Activation/physiology , Animals , HEK293 Cells , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Octamer Transcription Factor-3/genetics , Protein Domains , Receptors, Estrogen/genetics
5.
Cell ; 168(4): 629-643, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28187285

ABSTRACT

Cancer arises from genetic alterations that invariably lead to dysregulated transcriptional programs. These dysregulated programs can cause cancer cells to become highly dependent on certain regulators of gene expression. Here, we discuss how transcriptional control is disrupted by genetic alterations in cancer cells, why transcriptional dependencies can develop as a consequence of dysregulated programs, and how these dependencies provide opportunities for novel therapeutic interventions in cancer.


Subject(s)
Neoplasms/drug therapy , Neoplasms/genetics , Animals , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Homeostasis , Humans , Neoplasms/pathology , Transcription Factors/metabolism , Transcription, Genetic
6.
Cell ; 168(6): 1000-1014.e15, 2017 Mar 09.
Article in English | MEDLINE | ID: mdl-28283057

ABSTRACT

Super-enhancers are an emerging subclass of regulatory regions controlling cell identity and disease genes. However, their biological function and impact on miRNA networks are unclear. Here, we report that super-enhancers drive the biogenesis of master miRNAs crucial for cell identity by enhancing both transcription and Drosha/DGCR8-mediated primary miRNA (pri-miRNA) processing. Super-enhancers, together with broad H3K4me3 domains, shape a tissue-specific and evolutionarily conserved atlas of miRNA expression and function. CRISPR/Cas9 genomics revealed that super-enhancer constituents act cooperatively and facilitate Drosha/DGCR8 recruitment and pri-miRNA processing to boost cell-specific miRNA production. The BET-bromodomain inhibitor JQ1 preferentially inhibits super-enhancer-directed cotranscriptional pri-miRNA processing. Furthermore, super-enhancers are characterized by pervasive interaction with DGCR8/Drosha and DGCR8/Drosha-regulated mRNA stability control, suggesting unique RNA regulation at super-enhancers. Finally, super-enhancers mark multiple miRNAs associated with cancer hallmarks. This study presents principles underlying miRNA biology in health and disease and an unrecognized higher-order property of super-enhancers in RNA processing beyond transcription.


Subject(s)
Enhancer Elements, Genetic , MicroRNAs/metabolism , Animals , Azepines/pharmacology , Gene Expression Regulation , Histone Code , Humans , Mice , Neoplasms/genetics , Organ Specificity , RNA Processing, Post-Transcriptional/drug effects , Transcription Factors/metabolism , Transcription, Genetic , Triazoles/pharmacology
7.
Cell ; 169(1): 13-23, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28340338

ABSTRACT

Phase-separated multi-molecular assemblies provide a general regulatory mechanism to compartmentalize biochemical reactions within cells. We propose that a phase separation model explains established and recently described features of transcriptional control. These features include the formation of super-enhancers, the sensitivity of super-enhancers to perturbation, the transcriptional bursting patterns of enhancers, and the ability of an enhancer to produce simultaneous activation at multiple genes. This model provides a conceptual framework to further explore principles of gene control in mammals.


Subject(s)
Gene Expression Regulation , Models, Biological , Transcription, Genetic , Animals , Enhancer Elements, Genetic , Eukaryotic Cells/metabolism , Humans , Transcription Factors/metabolism , Transcriptional Activation
8.
Cell ; 171(7): 1573-1588.e28, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29224777

ABSTRACT

There is considerable evidence that chromosome structure plays important roles in gene control, but we have limited understanding of the proteins that contribute to structural interactions between gene promoters and their enhancer elements. Large DNA loops that encompass genes and their regulatory elements depend on CTCF-CTCF interactions, but most enhancer-promoter interactions do not employ this structural protein. Here, we show that the ubiquitously expressed transcription factor Yin Yang 1 (YY1) contributes to enhancer-promoter structural interactions in a manner analogous to DNA interactions mediated by CTCF. YY1 binds to active enhancers and promoter-proximal elements and forms dimers that facilitate the interaction of these DNA elements. Deletion of YY1 binding sites or depletion of YY1 protein disrupts enhancer-promoter looping and gene expression. We propose that YY1-mediated enhancer-promoter interactions are a general feature of mammalian gene control.


Subject(s)
Enhancer Elements, Genetic , Promoter Regions, Genetic , YY1 Transcription Factor/metabolism , Animals , CCCTC-Binding Factor/metabolism , Embryonic Stem Cells/metabolism , Humans , Mice
9.
Cell ; 167(5): 1188-1200, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863240

ABSTRACT

Understanding how transcriptional enhancers control over 20,000 protein-coding genes to maintain cell-type-specific gene expression programs in all human cells is a fundamental challenge in regulatory biology. Recent studies suggest that gene regulatory elements and their target genes generally occur within insulated neighborhoods, which are chromosomal loop structures formed by the interaction of two DNA sites bound by the CTCF protein and occupied by the cohesin complex. Here, we review evidence that insulated neighborhoods provide for specific enhancer-gene interactions, are essential for both normal gene activation and repression, form a chromosome scaffold that is largely preserved throughout development, and are perturbed by genetic and epigenetic factors in disease. Insulated neighborhoods are a powerful paradigm for gene control that provides new insights into development and disease.


Subject(s)
Chromosomes/metabolism , Gene Expression Regulation , Animals , CCCTC-Binding Factor , Enhancer Elements, Genetic , Humans , Insulator Elements , Mammals/metabolism , Repressor Proteins/metabolism
10.
Cell ; 167(1): 233-247.e17, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27662091

ABSTRACT

Mammalian DNA methylation is a critical epigenetic mechanism orchestrating gene expression networks in many biological processes. However, investigation of the functions of specific methylation events remains challenging. Here, we demonstrate that fusion of Tet1 or Dnmt3a with a catalytically inactive Cas9 (dCas9) enables targeted DNA methylation editing. Targeting of the dCas9-Tet1 or -Dnmt3a fusion protein to methylated or unmethylated promoter sequences caused activation or silencing, respectively, of an endogenous reporter. Targeted demethylation of the BDNF promoter IV or the MyoD distal enhancer by dCas9-Tet1 induced BDNF expression in post-mitotic neurons or activated MyoD facilitating reprogramming of fibroblasts into myoblasts, respectively. Targeted de novo methylation of a CTCF loop anchor site by dCas9-Dnmt3a blocked CTCF binding and interfered with DNA looping, causing altered gene expression in the neighboring loop. Finally, we show that these tools can edit DNA methylation in mice, demonstrating their wide utility for functional studies of epigenetic regulation.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Editing/methods , Proto-Oncogene Proteins/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Brain-Derived Neurotrophic Factor/genetics , CCCTC-Binding Factor , CRISPR-Associated Protein 9 , Cell Line , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA-Binding Proteins/genetics , Endonucleases/genetics , Endonucleases/metabolism , Enhancer Elements, Genetic , Genome , Mice , MyoD Protein/metabolism , Neurons/metabolism , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism
11.
Mol Cell ; 83(14): 2449-2463.e13, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37402367

ABSTRACT

Transcription factors (TFs) orchestrate the gene expression programs that define each cell's identity. The canonical TF accomplishes this with two domains, one that binds specific DNA sequences and the other that binds protein coactivators or corepressors. We find that at least half of TFs also bind RNA, doing so through a previously unrecognized domain with sequence and functional features analogous to the arginine-rich motif of the HIV transcriptional activator Tat. RNA binding contributes to TF function by promoting the dynamic association between DNA, RNA, and TF on chromatin. TF-RNA interactions are a conserved feature important for vertebrate development and disrupted in disease. We propose that the ability to bind DNA, RNA, and protein is a general property of many TFs and is fundamental to their gene regulatory function.


Subject(s)
RNA , Transcription Factors , Transcription Factors/metabolism , RNA/metabolism , Binding Sites , Protein Binding , DNA/genetics
12.
Cell ; 163(1): 174-86, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26406377

ABSTRACT

Triple-negative breast cancer (TNBC) is a highly aggressive form of breast cancer that exhibits extremely high levels of genetic complexity and yet a relatively uniform transcriptional program. We postulate that TNBC might be highly dependent on uninterrupted transcription of a key set of genes within this gene expression program and might therefore be exceptionally sensitive to inhibitors of transcription. Utilizing kinase inhibitors and CRISPR/Cas9-mediated gene editing, we show here that triple-negative but not hormone receptor-positive breast cancer cells are exceptionally dependent on CDK7, a transcriptional cyclin-dependent kinase. TNBC cells are unique in their dependence on this transcriptional CDK and suffer apoptotic cell death upon CDK7 inhibition. An "Achilles cluster" of TNBC-specific genes is especially sensitive to CDK7 inhibition and frequently associated with super-enhancers. We conclude that CDK7 mediates transcriptional addiction to a vital cluster of genes in TNBC and CDK7 inhibition may be a useful therapy for this challenging cancer.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Neoplastic , Transcription, Genetic , Triple Negative Breast Neoplasms/genetics , Animals , Cell Line, Tumor , Cyclin-Dependent Kinases/antagonists & inhibitors , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Cyclin-Dependent Kinase-Activating Kinase
13.
Cell ; 159(2): 374-387, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25303531

ABSTRACT

The pluripotent state of embryonic stem cells (ESCs) is produced by active transcription of genes that control cell identity and repression of genes encoding lineage-specifying developmental regulators. Here, we use ESC cohesin ChIA-PET data to identify the local chromosomal structures at both active and repressed genes across the genome. The results produce a map of enhancer-promoter interactions and reveal that super-enhancer-driven genes generally occur within chromosome structures that are formed by the looping of two interacting CTCF sites co-occupied by cohesin. These looped structures form insulated neighborhoods whose integrity is important for proper expression of local genes. We also find that repressed genes encoding lineage-specifying developmental regulators occur within insulated neighborhoods. These results provide insights into the relationship between transcriptional control of cell identity genes and control of local chromosome structure.


Subject(s)
Chromosomes, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Animals , CCCTC-Binding Factor , Cell Cycle Proteins/metabolism , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/metabolism , Embryonic Stem Cells/cytology , Genome , High-Throughput Nucleotide Sequencing , Mice , Organ Specificity , Pluripotent Stem Cells/metabolism , Repressor Proteins/metabolism , Sequence Analysis, DNA , Cohesins
14.
Cell ; 159(5): 1126-1139, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25416950

ABSTRACT

The MYC oncoproteins are thought to stimulate tumor cell growth and proliferation through amplification of gene transcription, a mechanism that has thwarted most efforts to inhibit MYC function as potential cancer therapy. Using a covalent inhibitor of cyclin-dependent kinase 7 (CDK7) to disrupt the transcription of amplified MYCN in neuroblastoma cells, we demonstrate downregulation of the oncoprotein with consequent massive suppression of MYCN-driven global transcriptional amplification. This response translated to significant tumor regression in a mouse model of high-risk neuroblastoma, without the introduction of systemic toxicity. The striking treatment selectivity of MYCN-overexpressing cells correlated with preferential downregulation of super-enhancer-associated genes, including MYCN and other known oncogenic drivers in neuroblastoma. These results indicate that CDK7 inhibition, by selectively targeting the mechanisms that promote global transcriptional amplification in tumor cells, may be useful therapy for cancers that are driven by MYC family oncoproteins.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Disease Models, Animal , Neuroblastoma/drug therapy , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Phenylenediamines/therapeutic use , Protein Kinase Inhibitors/pharmacology , Pyrimidines/therapeutic use , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinases/metabolism , Humans , N-Myc Proto-Oncogene Protein , Transcription, Genetic/drug effects , Cyclin-Dependent Kinase-Activating Kinase
15.
Cell ; 152(6): 1237-51, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23498934

ABSTRACT

The gene expression programs that establish and maintain specific cell states in humans are controlled by thousands of transcription factors, cofactors, and chromatin regulators. Misregulation of these gene expression programs can cause a broad range of diseases. Here, we review recent advances in our understanding of transcriptional regulation and discuss how these have provided new insights into transcriptional misregulation in disease.


Subject(s)
Disease/genetics , Gene Expression Regulation , Transcription, Genetic , Animals , Gene Regulatory Networks , Humans , Neoplasms/genetics , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
16.
Cell ; 153(2): 307-19, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23582322

ABSTRACT

Master transcription factors Oct4, Sox2, and Nanog bind enhancer elements and recruit Mediator to activate much of the gene expression program of pluripotent embryonic stem cells (ESCs). We report here that the ESC master transcription factors form unusual enhancer domains at most genes that control the pluripotent state. These domains, which we call super-enhancers, consist of clusters of enhancers that are densely occupied by the master regulators and Mediator. Super-enhancers differ from typical enhancers in size, transcription factor density and content, ability to activate transcription, and sensitivity to perturbation. Reduced levels of Oct4 or Mediator cause preferential loss of expression of super-enhancer-associated genes relative to other genes, suggesting how changes in gene expression programs might be accomplished during development. In other more differentiated cells, super-enhancers containing cell-type-specific master transcription factors are also found at genes that define cell identity. Super-enhancers thus play key roles in the control of mammalian cell identity.


Subject(s)
Cell Lineage , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Mediator Complex/metabolism , Transcription Factors/metabolism , Animals , B-Lymphocytes/metabolism , Cell Line , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Mice , Transcription, Genetic
17.
Cell ; 153(2): 320-34, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23582323

ABSTRACT

Chromatin regulators have become attractive targets for cancer therapy, but it is unclear why inhibition of these ubiquitous regulators should have gene-specific effects in tumor cells. Here, we investigate how inhibition of the widely expressed transcriptional coactivator BRD4 leads to selective inhibition of the MYC oncogene in multiple myeloma (MM). BRD4 and Mediator were found to co-occupy thousands of enhancers associated with active genes. They also co-occupied a small set of exceptionally large super-enhancers associated with genes that feature prominently in MM biology, including the MYC oncogene. Treatment of MM tumor cells with the BET-bromodomain inhibitor JQ1 led to preferential loss of BRD4 at super-enhancers and consequent transcription elongation defects that preferentially impacted genes with super-enhancers, including MYC. Super-enhancers were found at key oncogenic drivers in many other tumor cells. These observations have implications for the discovery of cancer therapeutics directed at components of super-enhancers in diverse tumor types.


Subject(s)
Antineoplastic Agents/pharmacology , Azepines/pharmacology , Enhancer Elements, Genetic , Mediator Complex/metabolism , Neoplasms/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Triazoles/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Chromatin , Gene Expression Regulation, Neoplastic/drug effects , Genome-Wide Association Study , Humans , Mediator Complex/antagonists & inhibitors , Multiple Myeloma/genetics , Nuclear Proteins/antagonists & inhibitors , Transcription Elongation, Genetic , Transcription Factors/antagonists & inhibitors
18.
Cell ; 155(4): 934-47, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24119843

ABSTRACT

Super-enhancers are large clusters of transcriptional enhancers that drive expression of genes that define cell identity. Improved understanding of the roles that super-enhancers play in biology would be afforded by knowing the constellation of factors that constitute these domains and by identifying super-enhancers across the spectrum of human cell types. We describe here the population of transcription factors, cofactors, chromatin regulators, and transcription apparatus occupying super-enhancers in embryonic stem cells and evidence that super-enhancers are highly transcribed. We produce a catalog of super-enhancers in a broad range of human cell types and find that super-enhancers associate with genes that control and define the biology of these cells. Interestingly, disease-associated variation is especially enriched in the super-enhancers of disease-relevant cell types. Furthermore, we find that cancer cells generate super-enhancers at oncogenes and other genes important in tumor pathogenesis. Thus, super-enhancers play key roles in human cell identity in health and in disease.


Subject(s)
Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Neoplasms/genetics , Animals , Chromatin/metabolism , Humans , Neoplasms/pathology , Polymorphism, Single Nucleotide , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Transcription, Genetic
19.
Cell ; 154(1): 61-74, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23827675

ABSTRACT

The recent discovery that normal and neoplastic epithelial cells re-enter the stem cell state raised the intriguing possibility that the aggressiveness of carcinomas derives not from their existing content of cancer stem cells (CSCs) but from their proclivity to generate new CSCs from non-CSC populations. Here, we demonstrate that non-CSCs of human basal breast cancers are plastic cell populations that readily switch from a non-CSC to CSC state. The observed cell plasticity is dependent on ZEB1, a key regulator of the epithelial-mesenchymal transition. We find that plastic non-CSCs maintain the ZEB1 promoter in a bivalent chromatin configuration, enabling them to respond readily to microenvironmental signals, such as TGFß. In response, the ZEB1 promoter converts from a bivalent to active chromatin configuration, ZEB1 transcription increases, and non-CSCs subsequently enter the CSC state. Our findings support a dynamic model in which interconversions between low and high tumorigenic states occur frequently, thereby increasing tumorigenic and malignant potential.


Subject(s)
Breast Neoplasms/pathology , Chromatin/metabolism , Homeodomain Proteins/metabolism , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , Transcription Factors/metabolism , Animals , Breast Neoplasms/genetics , Epithelial Cells/pathology , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Humans , Hyaluronan Receptors/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism , Zinc Finger E-box-Binding Homeobox 1
20.
Mol Cell ; 78(3): 459-476.e13, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32240602

ABSTRACT

The cyclin-dependent kinase 1 (Cdk1) drives cell division. To uncover additional functions of Cdk1, we generated knockin mice expressing an analog-sensitive version of Cdk1 in place of wild-type Cdk1. In our study, we focused on embryonic stem cells (ESCs), because this cell type displays particularly high Cdk1 activity. We found that in ESCs, a large fraction of Cdk1 substrates is localized on chromatin. Cdk1 phosphorylates many proteins involved in epigenetic regulation, including writers and erasers of all major histone marks. Consistent with these findings, inhibition of Cdk1 altered histone-modification status of ESCs. High levels of Cdk1 in ESCs phosphorylate and partially inactivate Dot1l, the H3K79 methyltransferase responsible for placing activating marks on gene bodies. Decrease of Cdk1 activity during ESC differentiation de-represses Dot1l, thereby allowing coordinated expression of differentiation genes. These analyses indicate that Cdk1 functions to maintain the epigenetic identity of ESCs.


Subject(s)
CDC2 Protein Kinase/metabolism , Embryonic Stem Cells/physiology , Epigenesis, Genetic , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , CDC2 Protein Kinase/genetics , Cell Differentiation , Cells, Cultured , Chromatin Immunoprecipitation/methods , Female , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , MCF-7 Cells , Male , Mice , Mice, Knockout , Phosphorylation , Saccharomyces cerevisiae Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL