Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters











Publication year range
1.
Nat Cell Biol ; 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251719

ABSTRACT

The ten-eleven translocation (TET) family of dioxygenases maintain stable local DNA demethylation during cell division and lineage specification. As the major catalytic product of TET enzymes, 5-hydroxymethylcytosine is selectively enriched at specific genomic regions, such as enhancers, in a tissue-dependent manner. However, the mechanisms underlying this selectivity remain unresolved. Here we unveil a low-complexity insert domain within TET2 that facilitates its biomolecular condensation with epigenetic modulators, such as UTX and MLL4. This co-condensation fosters a permissive chromatin environment for precise DNA demethylation. Disrupting low-complexity insert-mediated condensation alters the genomic binding of TET2 to cause promiscuous DNA demethylation and genome reorganization. These changes influence the expression of key genes implicated in leukaemogenesis to curtail leukaemia cell proliferation. Collectively, this study establishes the pivotal role of TET2 condensation in orchestrating precise DNA demethylation and gene transcription to support tumour cell growth.

2.
Nat Cell Biol ; 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169219

ABSTRACT

Post-transcriptional mechanisms are fundamental safeguards of progenitor cell identity and are often dysregulated in cancer. Here, we identified regulators of P-bodies as crucial vulnerabilities in acute myeloid leukaemia (AML) through genome-wide CRISPR screens in normal and malignant haematopoietic progenitors. We found that leukaemia cells harbour aberrantly elevated numbers of P-bodies and show that P-body assembly is crucial for initiation and maintenance of AML. Notably, P-body loss had little effect upon homoeostatic haematopoiesis but impacted regenerative haematopoiesis. Molecular characterization of P-bodies purified from human AML cells unveiled their critical role in sequestering messenger RNAs encoding potent tumour suppressors from the translational machinery. P-body dissolution promoted translation of these mRNAs, which in turn rewired gene expression and chromatin architecture in leukaemia cells. Collectively, our findings highlight the contrasting and unique roles of RNA sequestration in P-bodies during tissue homoeostasis and oncogenesis. These insights open potential avenues for understanding myeloid leukaemia and future therapeutic interventions.

3.
Nat Cell Biol ; 26(4): 593-603, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38553595

ABSTRACT

Loss of protein function is a driving force of ageing. We have identified peptidyl-prolyl isomerase A (PPIA or cyclophilin A) as a dominant chaperone in haematopoietic stem and progenitor cells. Depletion of PPIA accelerates stem cell ageing. We found that proteins with intrinsically disordered regions (IDRs) are frequent PPIA substrates. IDRs facilitate interactions with other proteins or nucleic acids and can trigger liquid-liquid phase separation. Over 20% of PPIA substrates are involved in the formation of supramolecular membrane-less organelles. PPIA affects regulators of stress granules (PABPC1), P-bodies (DDX6) and nucleoli (NPM1) to promote phase separation and increase cellular stress resistance. Haematopoietic stem cell ageing is associated with a post-transcriptional decrease in PPIA expression and reduced translation of IDR-rich proteins. Here we link the chaperone PPIA to the synthesis of intrinsically disordered proteins, which indicates that impaired protein interaction networks and macromolecular condensation may be potential determinants of haematopoietic stem cell ageing.


Subject(s)
Intrinsically Disordered Proteins , Intrinsically Disordered Proteins/chemistry , Cyclophilin A/genetics , Cyclophilin A/metabolism , RNA-Binding Proteins , Hematopoietic Stem Cells/metabolism
4.
Nat Cell Biol ; 25(8): 1121-1134, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37460697

ABSTRACT

The epigenetic mechanisms that maintain differentiated cell states remain incompletely understood. Here we employed histone mutants to uncover a crucial role for H3K36 methylation in the maintenance of cell identities across diverse developmental contexts. Focusing on the experimental induction of pluripotency, we show that H3K36M-mediated depletion of H3K36 methylation endows fibroblasts with a plastic state poised to acquire pluripotency in nearly all cells. At a cellular level, H3K36M facilitates epithelial plasticity by rendering fibroblasts insensitive to TGFß signals. At a molecular level, H3K36M enables the decommissioning of mesenchymal enhancers and the parallel activation of epithelial/stem cell enhancers. This enhancer rewiring is Tet dependent and redirects Sox2 from promiscuous somatic to pluripotency targets. Our findings reveal a previously unappreciated dual role for H3K36 methylation in the maintenance of cell identity by integrating a crucial developmental pathway into sustained expression of cell-type-specific programmes, and by opposing the expression of alternative lineage programmes through enhancer methylation.


Subject(s)
Epigenesis, Genetic , Histones , Methylation , Histones/genetics , Histones/metabolism , Cell Differentiation/genetics , Fibroblasts/metabolism , Cell Lineage/genetics
5.
Cell Stem Cell ; 30(4): 335-337, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37028396

ABSTRACT

Small molecule-induced cell fate transitions are characterized by low efficiency and slow kinetics. An optimized chemical reprogramming approach now facilitates the robust and rapid conversion of somatic cells to pluripotent stem cells, unlocking exciting avenues to study and manipulate human cell identity.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Humans , Cellular Reprogramming , Cell Differentiation
6.
Cell Reprogram ; 25(1): 9-10, 2023 02.
Article in English | MEDLINE | ID: mdl-36594927

ABSTRACT

Short-term expression of Yamanaka factors early in life promotes epigenetic reprogramming and an increased healthy lifespan in a mouse model of accelerated aging.


Subject(s)
Cellular Reprogramming , Longevity , Animals , Mice , Epigenesis, Genetic , Rejuvenation , Aging/genetics
7.
Science ; 378(6623): 983-989, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36454826

ABSTRACT

Neurons harbor high levels of single-strand DNA breaks (SSBs) that are targeted to neuronal enhancers, but the source of this endogenous damage remains unclear. Using two systems of postmitotic lineage specification-induced pluripotent stem cell-derived neurons and transdifferentiated macrophages-we show that thymidine DNA glycosylase (TDG)-driven excision of methylcytosines oxidized with ten-eleven translocation enzymes (TET) is a source of SSBs. Although macrophage differentiation favors short-patch base excision repair to fill in single-nucleotide gaps, neurons also frequently use the long-patch subpathway. Disrupting this gap-filling process using anti-neoplastic cytosine analogs triggers a DNA damage response and neuronal cell death, which is dependent on TDG. Thus, TET-mediated active DNA demethylation promotes endogenous DNA damage, a process that normally safeguards cell identity but can also provoke neurotoxicity after anticancer treatments.


Subject(s)
DNA Breaks, Single-Stranded , DNA Demethylation , DNA Repair , Enhancer Elements, Genetic , Induced Pluripotent Stem Cells , Neurons , Thymine DNA Glycosylase , Cell Differentiation , Neurons/enzymology , 5-Methylcytosine/metabolism , Humans , Cell Transdifferentiation
8.
Nat Cell Biol ; 24(9): 1326-1327, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36075977
9.
Genes Dev ; 35(21-22): 1527-1547, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34711655

ABSTRACT

Understanding the genetic control of human embryonic stem cell function is foundational for developmental biology and regenerative medicine. Here we describe an integrated genome-scale loss- and gain-of-function screening approach to identify genetic networks governing embryonic stem cell proliferation and differentiation into the three germ layers. We identified a deep link between pluripotency maintenance and survival by showing that genetic alterations that cause pluripotency dissolution simultaneously increase apoptosis resistance. We discovered that the chromatin-modifying complex SAGA and in particular its subunit TADA2B are central regulators of pluripotency, survival, growth, and lineage specification. Joint analysis of all screens revealed that genetic alterations that broadly inhibit differentiation across multiple germ layers drive proliferation and survival under pluripotency-maintaining conditions and coincide with known cancer drivers. Our results show the power of integrated multilayer genetic screening for the robust mapping of complex genetic networks.


Subject(s)
Human Embryonic Stem Cells , Cell Differentiation/genetics , Embryonic Stem Cells , Gain of Function Mutation , Germ Layers , Humans
11.
Genes Dev ; 35(17-18): 1209-1228, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34413137

ABSTRACT

The generation of myotubes from fibroblasts upon forced MyoD expression is a classic example of transcription factor-induced reprogramming. We recently discovered that additional modulation of signaling pathways with small molecules facilitates reprogramming to more primitive induced myogenic progenitor cells (iMPCs). Here, we dissected the transcriptional and epigenetic dynamics of mouse fibroblasts undergoing reprogramming to either myotubes or iMPCs using a MyoD-inducible transgenic model. Induction of MyoD in fibroblasts combined with small molecules generated Pax7+ iMPCs with high similarity to primary muscle stem cells. Analysis of intermediate stages of iMPC induction revealed that extinction of the fibroblast program preceded induction of the stem cell program. Moreover, key stem cell genes gained chromatin accessibility prior to their transcriptional activation, and these regions exhibited a marked loss of DNA methylation dependent on the Tet enzymes. In contrast, myotube generation was associated with few methylation changes, incomplete and unstable reprogramming, and an insensitivity to Tet depletion. Finally, we showed that MyoD's ability to bind to unique bHLH targets was crucial for generating iMPCs but dispensable for generating myotubes. Collectively, our analyses elucidate the role of MyoD in myogenic reprogramming and derive general principles by which transcription factors and signaling pathways cooperate to rewire cell identity.


Subject(s)
Muscle Development , MyoD Protein , Animals , Cell Differentiation/genetics , Mice , Muscle Development/genetics , Muscle Fibers, Skeletal , Muscle, Skeletal , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/metabolism , Stem Cells/metabolism
12.
Genome Biol ; 22(1): 171, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34082786

ABSTRACT

BACKGROUND: Somatic cell reprogramming is the process that allows differentiated cells to revert to a pluripotent state. In contrast to the extensively studied rewiring of epigenetic and transcriptional programs required for reprogramming, the dynamics of post-transcriptional changes and their associated regulatory mechanisms remain poorly understood. Here we study the dynamics of alternative splicing changes occurring during efficient reprogramming of mouse B cells into induced pluripotent stem (iPS) cells and compare them to those occurring during reprogramming of mouse embryonic fibroblasts. RESULTS: We observe a significant overlap between alternative splicing changes detected in the two reprogramming systems, which are generally uncoupled from changes in transcriptional levels. Correlation between gene expression of potential regulators and specific clusters of alternative splicing changes enables the identification and subsequent validation of CPSF3 and hnRNP UL1 as facilitators, and TIA1 as repressor of mouse embryonic fibroblasts reprogramming. We further find that these RNA-binding proteins control partially overlapping programs of splicing regulation, involving genes relevant for developmental and morphogenetic processes. CONCLUSIONS: Our results reveal common programs of splicing regulation during reprogramming of different cell types and identify three novel regulators of this process and their targets.


Subject(s)
Alternative Splicing/genetics , Cellular Reprogramming/genetics , Cleavage And Polyadenylation Specificity Factor/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , T-Cell Intracellular Antigen-1/metabolism , Animals , B-Lymphocytes/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Mice
13.
Development ; 146(23)2019 12 02.
Article in English | MEDLINE | ID: mdl-31792064

ABSTRACT

Development and homeostasis rely upon concerted regulatory pathways to establish the specialized cell types needed for tissue function. Once a cell type is specified, the processes that restrict and maintain cell fate are equally important in ensuring tissue integrity. Over the past decade, several approaches to experimentally reprogram cell fate have emerged. Importantly, efforts to improve and understand these approaches have uncovered novel molecular determinants that reinforce lineage commitment and help resist cell fate changes. In this Review, we summarize recent studies that have provided insights into the various chromatin factors, post-transcriptional processes and features of genomic organization that safeguard cell identity in the context of reprogramming to pluripotency. We also highlight how these factors function in other experimental, physiological and pathological cell fate transitions, including direct lineage conversion, pluripotency-to-totipotency reversion and cancer.


Subject(s)
Cellular Reprogramming , Chromatin/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Chromatin/pathology , Humans , Neoplasms/pathology , Neoplastic Stem Cells/pathology
14.
Nat Cell Biol ; 21(11): 1449-1461, 2019 11.
Article in English | MEDLINE | ID: mdl-31659274

ABSTRACT

Development and differentiation are associated with profound changes to histone modifications, yet their in vivo function remains incompletely understood. Here, we generated mouse models expressing inducible histone H3 lysine-to-methionine (K-to-M) mutants, which globally inhibit methylation at specific sites. Mice expressing H3K36M developed severe anaemia with arrested erythropoiesis, a marked haematopoietic stem cell defect, and rapid lethality. By contrast, mice expressing H3K9M survived up to a year and showed expansion of multipotent progenitors, aberrant lymphopoiesis and thrombocytosis. Additionally, some H3K9M mice succumbed to aggressive T cell leukaemia/lymphoma, while H3K36M mice exhibited differentiation defects in testis and intestine. Mechanistically, induction of either mutant reduced corresponding histone trimethylation patterns genome-wide and altered chromatin accessibility as well as gene expression landscapes. Strikingly, discontinuation of transgene expression largely restored differentiation programmes. Our work shows that individual chromatin modifications are required at several specific stages of differentiation and introduces powerful tools to interrogate their roles in vivo.


Subject(s)
Epigenesis, Genetic , Histones/metabolism , Leukemia, T-Cell/genetics , Lysine/metabolism , Methionine/metabolism , Teratoma/genetics , Animals , Bone Marrow Transplantation , Cell Lineage/genetics , Disease Models, Animal , Doxycycline/pharmacology , Erythroid Cells/metabolism , Erythroid Cells/pathology , Female , Granulocytes/metabolism , Granulocytes/pathology , Histones/genetics , Leukemia, T-Cell/chemically induced , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Male , Methylation , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/pathology , Mutation , Signal Transduction , Survival Analysis , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Teratoma/chemically induced , Teratoma/metabolism , Teratoma/pathology
15.
Cell Stem Cell ; 25(5): 622-638.e13, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31588046

ABSTRACT

Post-transcriptional mechanisms have the potential to influence complex changes in gene expression, yet their role in cell fate transitions remains largely unexplored. Here, we show that suppression of the RNA helicase DDX6 endows human and mouse primed embryonic stem cells (ESCs) with a differentiation-resistant, "hyper-pluripotent" state, which readily reprograms to a naive state resembling the preimplantation embryo. We further demonstrate that DDX6 plays a key role in adult progenitors where it controls the balance between self-renewal and differentiation in a context-dependent manner. Mechanistically, DDX6 mediates the translational suppression of target mRNAs in P-bodies. Upon loss of DDX6 activity, P-bodies dissolve and release mRNAs encoding fate-instructive transcription and chromatin factors that re-enter the ribosome pool. Increased translation of these targets impacts cell fate by rewiring the enhancer, heterochromatin, and DNA methylation landscapes of undifferentiated cell types. Collectively, our data establish a link between P-body homeostasis, chromatin organization, and stem cell potency.


Subject(s)
Cell Differentiation/genetics , Cell Plasticity/genetics , DEAD-box RNA Helicases/metabolism , Induced Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , Ribonucleoproteins/metabolism , Animals , Cell Line , Chromatin Assembly and Disassembly/genetics , DEAD-box RNA Helicases/genetics , DNA Methylation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation/genetics , Gene Ontology , Homeostasis/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/enzymology , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mice, Inbred C57BL , Nanog Homeobox Protein/metabolism , Organoids/cytology , Organoids/diagnostic imaging , Organoids/metabolism , Protein Biosynthesis/genetics , Proteins/metabolism , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , RNA-Seq , Ribonucleoproteins/genetics , Ribosomes/metabolism
16.
Nat Cell Biol ; 21(7): 824-834, 2019 07.
Article in English | MEDLINE | ID: mdl-31235934

ABSTRACT

How pluripotent stem cells differentiate into the main germ layers is a key question of developmental biology. Here, we show that the chromatin-related factor Whsc1 (also known as Nsd2 and MMSET) has a dual role in pluripotency exit and germ layer specification of embryonic stem cells. On induction of differentiation, a proportion of Whsc1-depleted embryonic stem cells remain entrapped in a pluripotent state and fail to form mesendoderm, although they are still capable of generating neuroectoderm. These functions of Whsc1 are independent of its methyltransferase activity. Whsc1 binds to enhancers of the mesendodermal regulators Gata4, T (Brachyury), Gata6 and Foxa2, together with Brd4, and activates the expression of these genes. Depleting each of these regulators also delays pluripotency exit, suggesting that they mediate the effects observed with Whsc1. Our data indicate that Whsc1 links silencing of the pluripotency regulatory network with activation of mesendoderm lineages.


Subject(s)
Cell Differentiation/physiology , Endoderm/cytology , Histone-Lysine N-Methyltransferase/metabolism , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation/genetics , Cell Lineage , Embryonic Stem Cells/cytology , Germ Layers/cytology , Mice , Neural Plate/cytology , Nuclear Proteins/metabolism , Transcription Factors/metabolism
17.
Elife ; 82019 03 12.
Article in English | MEDLINE | ID: mdl-30860479

ABSTRACT

Forced transcription factor expression can transdifferentiate somatic cells into other specialised cell types or reprogram them into induced pluripotent stem cells (iPSCs) with variable efficiency. To better understand the heterogeneity of these processes, we used single-cell RNA sequencing to follow the transdifferentation of murine pre-B cells into macrophages as well as their reprogramming into iPSCs. Even in these highly efficient systems, there was substantial variation in the speed and path of fate conversion. We predicted and validated that these differences are inversely coupled and arise in the starting cell population, with Mychigh large pre-BII cells transdifferentiating slowly but reprogramming efficiently and Myclow small pre-BII cells transdifferentiating rapidly but failing to reprogram. Strikingly, differences in Myc activity predict the efficiency of reprogramming across a wide range of somatic cell types. These results illustrate how single cell expression and computational analyses can identify the origins of heterogeneity in cell fate conversion processes.


Subject(s)
Cell Lineage , Cell Transdifferentiation , Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Precursor Cells, B-Lymphoid/cytology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA-Seq , Signal Transduction , Single-Cell Analysis , Transcriptome
19.
Trends Cell Biol ; 28(12): 971-973, 2018 12.
Article in English | MEDLINE | ID: mdl-30463679

ABSTRACT

Cell fate transitions involve rapid changes in gene expression patterns, yet the role of post-translational modifications in these processes remains underexplored. A recent study identifies SUMOylation as a guardian of cell identity that acts during differentiation and reprogramming by reinforcing active enhancers and maintaining silenced heterochromatin in a context-specific manner.


Subject(s)
Chromatin , Sumoylation , Cell Differentiation , Cell Plasticity , Protein Processing, Post-Translational
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL