Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
1.
Nature ; 631(8019): 216-223, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38839954

ABSTRACT

Transcription is the primary regulatory step in gene expression. Divergent transcription initiation from promoters and enhancers produces stable RNAs from genes and unstable RNAs from enhancers1,2. Nascent RNA capture and sequencing assays simultaneously measure gene and enhancer activity in cell populations3. However, fundamental questions about the temporal regulation of transcription and enhancer-gene coordination remain unanswered, primarily because of the absence of a single-cell perspective on active transcription. In this study, we present scGRO-seq-a new single-cell nascent RNA sequencing assay that uses click chemistry-and unveil coordinated transcription throughout the genome. We demonstrate the episodic nature of transcription and the co-transcription of functionally related genes. scGRO-seq can estimate burst size and frequency by directly quantifying transcribing RNA polymerases in individual cells and can leverage replication-dependent non-polyadenylated histone gene transcription to elucidate cell cycle dynamics. The single-nucleotide spatial and temporal resolution of scGRO-seq enables the identification of networks of enhancers and genes. Our results suggest that the bursting of transcription at super-enhancers precedes bursting from associated genes. By imparting insights into the dynamic nature of global transcription and the origin and propagation of transcription signals, we demonstrate the ability of scGRO-seq to investigate the mechanisms of transcription regulation and the role of enhancers in gene expression.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Promoter Regions, Genetic , RNA , Sequence Analysis, RNA , Single-Cell Gene Expression Analysis , Transcription, Genetic , Animals , Humans , Mice , Cell Cycle/genetics , Click Chemistry/methods , DNA-Directed RNA Polymerases/analysis , DNA-Directed RNA Polymerases/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Histones/metabolism , Promoter Regions, Genetic/genetics , RNA/analysis , RNA/biosynthesis , RNA/genetics , Sequence Analysis, RNA/methods , Single-Cell Gene Expression Analysis/methods , Time Factors
2.
Genome Biol ; 25(1): 23, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38229106

ABSTRACT

Sequence-specific RNA-binding proteins (RBPs) play central roles in splicing decisions. Here, we describe a modular splicing architecture that leverages in vitro-derived RNA affinity models for 79 human RBPs and the annotated human genome to produce improved models of RBP binding and activity. Binding and activity are modeled by separate Motif and Aggregator components that can be mixed and matched, enforcing sparsity to improve interpretability. Training a new Adjusted Motif (AM) architecture on the splicing task not only yields better splicing predictions but also improves prediction of RBP-binding sites in vivo and of splicing activity, assessed using independent data.


Subject(s)
RNA Splicing , RNA-Binding Proteins , Humans , Binding Sites , RNA-Binding Proteins/metabolism , RNA/genetics , Protein Binding
4.
bioRxiv ; 2023 Sep 19.
Article in English | MEDLINE | ID: mdl-37745427

ABSTRACT

Transcription is the primary regulatory step in gene expression. Divergent transcription initiation from promoters and enhancers produces stable RNAs from genes and unstable RNAs from enhancers1-5. Nascent RNA capture and sequencing assays simultaneously measure gene and enhancer activity in cell populations6-9. However, fundamental questions in the temporal regulation of transcription and enhancer-gene synchrony remain unanswered primarily due to the absence of a single-cell perspective on active transcription. In this study, we present scGRO-seq - a novel single-cell nascent RNA sequencing assay using click-chemistry - and unveil the coordinated transcription throughout the genome. scGRO-seq demonstrates the episodic nature of transcription, and estimates burst size and frequency by directly quantifying transcribing RNA polymerases in individual cells. It reveals the co-transcription of functionally related genes and leverages the replication-dependent non-polyadenylated histone genes transcription to elucidate cell-cycle dynamics. The single-nucleotide spatial and temporal resolution of scGRO-seq identifies networks of enhancers and genes and indicates that the bursting of transcription at super-enhancers precedes the burst from associated genes. By imparting insights into the dynamic nature of transcription and the origin and propagation of transcription signals, scGRO-seq demonstrates its unique ability to investigate the mechanisms of transcription regulation and the role of enhancers in gene expression.

5.
Science ; 380(6642): eabn7625, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37079685

ABSTRACT

RNA surveillance pathways detect and degrade defective transcripts to ensure RNA fidelity. We found that disrupted nuclear RNA surveillance is oncogenic. Cyclin-dependent kinase 13 (CDK13) is mutated in melanoma, and patient-mutated CDK13 accelerates zebrafish melanoma. CDK13 mutation causes aberrant RNA stabilization. CDK13 is required for ZC3H14 phosphorylation, which is necessary and sufficient to promote nuclear RNA degradation. Mutant CDK13 fails to activate nuclear RNA surveillance, causing aberrant protein-coding transcripts to be stabilized and translated. Forced aberrant RNA expression accelerates melanoma in zebrafish. We found recurrent mutations in genes encoding nuclear RNA surveillance components in many malignancies, establishing nuclear RNA surveillance as a tumor-suppressive pathway. Activating nuclear RNA surveillance is crucial to avoid accumulation of aberrant RNAs and their ensuing consequences in development and disease.


Subject(s)
CDC2 Protein Kinase , Carcinogens , Melanoma , RNA Stability , RNA, Nuclear , Skin Neoplasms , Animals , CDC2 Protein Kinase/genetics , Melanoma/genetics , Mutation , RNA, Nuclear/genetics , Skin Neoplasms/genetics , Zebrafish , Humans
6.
Proc Natl Acad Sci U S A ; 120(11): e2300605120, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36881621

ABSTRACT

CRISPR-Cas9 introduces targeted DNA breaks that engage competing DNA repair pathways, producing a spectrum of imprecise insertion/deletion mutations (indels) and precise templated mutations (precise edits). The relative frequencies of these pathways are thought to primarily depend on genomic sequence and cell state contexts, limiting control over mutational outcomes. Here, we report that engineered Cas9 nucleases that create different DNA break structures engage competing repair pathways at dramatically altered frequencies. We accordingly designed a Cas9 variant (vCas9) that produces breaks which suppress otherwise dominant nonhomologous end-joining (NHEJ) repair. Instead, breaks created by vCas9 are predominantly repaired by pathways utilizing homologous sequences, specifically microhomology-mediated end-joining (MMEJ) and homology-directed repair (HDR). Consequently, vCas9 enables efficient precise editing through HDR or MMEJ while suppressing indels caused by NHEJ in dividing and nondividing cells. These findings establish a paradigm of targeted nucleases custom-designed for specific mutational applications.


Subject(s)
CRISPR-Cas Systems , INDEL Mutation , CRISPR-Cas Systems/genetics , Mutation , Culture , DNA End-Joining Repair/genetics , Endonucleases/genetics
7.
Mol Cell ; 82(23): 4410-4427.e12, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36356583

ABSTRACT

Gene expression heterogeneity underlies cell states and contributes to developmental robustness. While heterogeneity can arise from stochastic transcriptional processes, the extent to which it is regulated is unclear. Here, we characterize the regulatory program underlying heterogeneity in murine embryonic stem cell (mESC) states. We identify differentially active and transcribed enhancers (DATEs) across states. DATEs regulate differentially expressed genes and are distinguished by co-binding of transcription factors Klf4 and Zfp281. In contrast to other factors that interact in a positive feedback network stabilizing mESC cell-type identity, Klf4 and Zfp281 drive opposing transcriptional and chromatin programs. Abrogation of factor binding to DATEs dampens variation in gene expression, and factor loss alters kinetics of switching between states. These results show antagonism between factors at enhancers results in gene expression heterogeneity and formation of cell states, with implications for the generation of diverse cell types during development.


Subject(s)
Embryonic Stem Cells , Transcription Factors , Animals , Mice , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/metabolism , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , Transcription Factors/genetics , Transcription Factors/metabolism
8.
RNA ; 28(1): 52-57, 2022 01.
Article in English | MEDLINE | ID: mdl-34772787

ABSTRACT

Macroscopic membraneless organelles containing RNA such as the nucleoli, germ granules, and the Cajal body have been known for decades. These biomolecular condensates are liquid-like bodies that can be formed by a phase transition. Recent evidence has revealed the presence of similar microscopic condensates associated with the transcription of genes. This brief article summarizes thoughts about the importance of condensates in the regulation of transcription and how RNA molecules, as components of such condensates, control the synthesis of RNA. Models and experimental data suggest that RNAs from enhancers facilitate the formation of a condensate that stabilizes the binding of transcription factors and accounts for a burst of transcription at the promoter. Termination of this burst is pictured as a nonequilibrium feedback loop where additional RNA destabilizes the condensate.


Subject(s)
Biomolecular Condensates/chemistry , DNA/chemistry , RNA-Binding Proteins/chemistry , RNA/chemistry , Transcription Factors/chemistry , Transcription, Genetic , Binding Sites , Biomolecular Condensates/metabolism , Cell Compartmentation , Cell Nucleolus/chemistry , Cell Nucleolus/metabolism , Coiled Bodies/chemistry , Coiled Bodies/metabolism , DNA/metabolism , Eukaryotic Cells/chemistry , Eukaryotic Cells/metabolism , Feedback, Physiological , Germ Cell Ribonucleoprotein Granules/chemistry , Germ Cell Ribonucleoprotein Granules/metabolism , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Promoter Regions, Genetic , Protein Binding , RNA/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism
9.
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
10.
Science ; 368(6497): 1386-1392, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32554597

ABSTRACT

The nucleus contains diverse phase-separated condensates that compartmentalize and concentrate biomolecules with distinct physicochemical properties. Here, we investigated whether condensates concentrate small-molecule cancer therapeutics such that their pharmacodynamic properties are altered. We found that antineoplastic drugs become concentrated in specific protein condensates in vitro and that this occurs through physicochemical properties independent of the drug target. This behavior was also observed in tumor cells, where drug partitioning influenced drug activity. Altering the properties of the condensate was found to affect the concentration and activity of drugs. These results suggest that selective partitioning and concentration of small molecules within condensates contributes to drug pharmacodynamics and that further understanding of this phenomenon may facilitate advances in disease therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Nucleus/metabolism , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Neoplasms/metabolism , Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Proc Natl Acad Sci U S A ; 117(12): 6942-6950, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32139605

ABSTRACT

Pluripotent embryonic stem cells (ESCs) contain the potential to form a diverse array of cells with distinct gene expression states, namely the cells of the adult vertebrate. Classically, diversity has been attributed to cells sensing their position with respect to external morphogen gradients. However, an alternative is that diversity arises in part from cooption of fluctuations in the gene regulatory network. Here we find ESCs exhibit intrinsic heterogeneity in the absence of external gradients by forming interconverting cell states. States vary in developmental gene expression programs and display distinct activity of microRNAs (miRNAs). Notably, miRNAs act on neighborhoods of pluripotency genes to increase variation of target genes and cell states. Loss of miRNAs that vary across states reduces target variation and delays state transitions, suggesting variable miRNAs organize and propagate variation to promote state transitions. Together these findings provide insight into how a gene regulatory network can coopt variation intrinsic to cell systems to form robust gene expression states. Interactions between intrinsic heterogeneity and environmental signals may help achieve developmental outcomes.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental , Gene Regulatory Networks , MicroRNAs/genetics , Animals , Argonaute Proteins/physiology , Embryonic Stem Cells/cytology , Gene Expression Profiling , Mice , Mice, Knockout , Nanog Homeobox Protein/physiology , RNA-Binding Proteins/physiology , SOXB1 Transcription Factors/physiology , Signal Transduction
12.
Mol Cell ; 78(1): 85-95.e8, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32032531

ABSTRACT

Imprinted genes with parental-biased allelic expression are frequently co-regulated and enriched in common biological pathways. Here, we functionally characterize a large cluster of microRNAs (miRNAs) expressed from the maternally inherited allele ("maternally expressed") to explore the molecular and cellular consequences of imprinted miRNA activity. Using an induced neuron (iN) culture system, we show that maternally expressed miRNAs from the miR-379/410 cluster direct the RNA-induced silencing complex (RISC) to transcriptional and developmental regulators, including paternally expressed transcripts like Plagl1. Maternal deletion of this imprinted miRNA cluster resulted in increased protein levels of several targets and upregulation of a broader transcriptional program regulating synaptic transmission and neuronal function. A subset of the transcriptional changes resulting from miR-379/410 deletion can be attributed to de-repression of Plagl1. These data suggest maternally expressed miRNAs antagonize paternally driven gene programs in neurons.


Subject(s)
Genomic Imprinting , MicroRNAs/metabolism , Neurons/metabolism , Animals , Argonaute Proteins/metabolism , Cell Line , Cells, Cultured , Embryonic Stem Cells/metabolism , Excitatory Postsynaptic Potentials , Gene Deletion , Mice , MicroRNAs/genetics , Neurogenesis/genetics , Neurons/physiology , RNA-Induced Silencing Complex/metabolism , Synaptic Transmission/genetics , Transcription, Genetic
13.
Mol Cell ; 75(3): 549-561.e7, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398323

ABSTRACT

Enhancers are DNA elements that are bound by transcription factors (TFs), which recruit coactivators and the transcriptional machinery to genes. Phase-separated condensates of TFs and coactivators have been implicated in assembling the transcription machinery at particular enhancers, yet the role of DNA sequence in this process has not been explored. We show that DNA sequences encoding TF binding site number, density, and affinity above sharply defined thresholds drive condensation of TFs and coactivators. A combination of specific structured (TF-DNA) and weak multivalent (TF-coactivator) interactions allows for condensates to form at particular genomic loci determined by the DNA sequence and the complement of expressed TFs. DNA features found to drive condensation promote enhancer activity and transcription in cells. Our study provides a framework to understand how the genome can scaffold transcriptional condensates at specific loci and how the universal phenomenon of phase separation might regulate this process.


Subject(s)
Chromatin/genetics , Enhancer Elements, Genetic , Transcription Factors/genetics , Transcription, Genetic , Animals , Base Sequence/genetics , Binding Sites/genetics , DNA/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genomics , Mice , Mouse Embryonic Stem Cells
14.
Nature ; 572(7770): 543-548, 2019 08.
Article in English | MEDLINE | ID: mdl-31391587

ABSTRACT

The synthesis of pre-mRNA by RNA polymerase II (Pol II) involves the formation of a transcription initiation complex, and a transition to an elongation complex1-4. The large subunit of Pol II contains an intrinsically disordered C-terminal domain that is phosphorylated by cyclin-dependent kinases during the transition from initiation to elongation, thus influencing the interaction of the C-terminal domain with different components of the initiation or the RNA-splicing apparatus5,6. Recent observations suggest that this model provides only a partial picture of the effects of phosphorylation of the C-terminal domain7-12. Both the transcription-initiation machinery and the splicing machinery can form phase-separated condensates that contain large numbers of component molecules: hundreds of molecules of Pol II and mediator are concentrated in condensates at super-enhancers7,8, and large numbers of splicing factors are concentrated in nuclear speckles, some of which occur at highly active transcription sites9-12. Here we investigate whether the phosphorylation of the Pol II C-terminal domain regulates the incorporation of Pol II into phase-separated condensates that are associated with transcription initiation and splicing. We find that the hypophosphorylated C-terminal domain of Pol II is incorporated into mediator condensates and that phosphorylation by regulatory cyclin-dependent kinases reduces this incorporation. We also find that the hyperphosphorylated C-terminal domain is preferentially incorporated into condensates that are formed by splicing factors. These results suggest that phosphorylation of the Pol II C-terminal domain drives an exchange from condensates that are involved in transcription initiation to those that are involved in RNA processing, and implicates phosphorylation as a mechanism that regulates condensate preference.


Subject(s)
Mediator Complex/chemistry , Mediator Complex/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , RNA Splicing , Transcription, Genetic , Animals , Cell Line , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Humans , Mediator Complex/genetics , Mice , Phosphorylation , Protein Domains , RNA Polymerase II/genetics , RNA Splicing Factors/chemistry , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism
15.
RNA ; 25(10): 1291-1297, 2019 10.
Article in English | MEDLINE | ID: mdl-31289130

ABSTRACT

Argonaute (Ago) proteins interact with various binding partners and play a pivotal role in microRNA (miRNA)-mediated silencing pathways. By utilizing immunoprecipitation followed by mass spectrometry to determine cytoplasmic Ago2 protein complexes in mouse embryonic stem cells (mESCs), we identified a putative RNA-binding protein FAM120A (also known as OSSA/C9ORF10) as an Ago2 interacting protein. Individual nucleotide resolution cross-linking and immunoprecipitation (iCLIP) analysis revealed that FAM120A binds to homopolymeric tracts in 3'-UTRs of about 2000 mRNAs, particularly poly(G) sequences. Comparison of FAM120A iCLIP and Ago2 iCLIP reveals that greater than one-third of mRNAs bound by Ago2 in mESCs are co-bound by FAM120A. Furthermore, such FAM120A-bound Ago2 target genes are not subject to Ago2-mediated target degradation. Reporter assays suggest that the 3'-UTRs of several FAM120A-bound miRNA target genes are less sensitive to Ago2-mediated target repression than those of FAM120A-unbound miRNA targets and FAM120A modulates them via its G-rich target sites. These findings suggest that Ago2 may exist in multiple protein complexes with varying degrees of functionality.


Subject(s)
Argonaute Proteins/metabolism , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , 3' Untranslated Regions , Animals , Cells, Cultured , Embryonic Stem Cells/metabolism , Immunoprecipitation , Mice
16.
Nat Med ; 25(4): 583-590, 2019 04.
Article in English | MEDLINE | ID: mdl-30804514

ABSTRACT

MicroRNAs (miRNAs) are post-transcriptional regulators of gene expression. Heterozygous loss-of-function point mutations of miRNA genes are associated with several human congenital disorders1-5, but neomorphic (gain-of-new-function) mutations in miRNAs due to nucleotide substitutions have not been reported. Here we describe a neomorphic seed region mutation in the chondrocyte-specific, super-enhancer-associated MIR140 gene encoding microRNA-140 (miR-140) in a novel autosomal dominant human skeletal dysplasia. Mice with the corresponding single nucleotide substitution show skeletal abnormalities similar to those of the patients but distinct from those of miR-140-null mice6. This mutant miRNA gene yields abundant mutant miR-140-5p expression without miRNA-processing defects. In chondrocytes, the mutation causes widespread derepression of wild-type miR-140-5p targets and repression of mutant miR-140-5p targets, indicating that the mutation produces both loss-of-function and gain-of-function effects. Furthermore, the mutant miR-140-5p seed competes with the conserved RNA-binding protein Ybx1 for overlapping binding sites. This finding may explain the potent target repression and robust in vivo effect by this mutant miRNA even in the absence of evolutionary selection of miRNA-target RNA interactions, which contributes to the strong regulatory effects of conserved miRNAs7,8. Our study presents the first case of a pathogenic gain-of-function miRNA mutation and provides molecular insight into neomorphic actions of emerging and/or mutant miRNAs.


Subject(s)
Bone Diseases, Developmental/genetics , Gain of Function Mutation/genetics , MicroRNAs/genetics , Animals , Base Sequence , Chondrocytes/metabolism , Female , Homozygote , Humans , Male , Mice, Inbred C57BL , Mice, Mutant Strains , MicroRNAs/metabolism , Pedigree , Phenotype , Transcriptome/genetics
17.
Proc Natl Acad Sci U S A ; 115(47): E11053-E11060, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30404915

ABSTRACT

A hallmark of biological systems is that particular functions and outcomes are realized in specific contexts, such as when particular signals are received. One mechanism for mediating specificity is described by Fisher's "lock and key" metaphor, exemplified by enzymes that bind selectively to a particular substrate via specific finely tuned interactions. Another mechanism, more prevalent in multicellular organisms, relies on multivalent weak cooperative interactions. Its importance has recently been illustrated by the recognition that liquid-liquid phase transitions underlie the formation of membraneless condensates that perform specific cellular functions. Based on computer simulations of an evolutionary model, we report that the latter mechanism likely became evolutionarily prominent when a large number of tasks had to be performed specifically for organisms to function properly. We find that the emergence of weak cooperative interactions for mediating specificity results in organisms that can evolve to accomplish new tasks with fewer, and likely less lethal, mutations. We argue that this makes the system more capable of undergoing evolutionary changes robustly, and thus this mechanism has been repeatedly positively selected in increasingly complex organisms. Specificity mediated by weak cooperative interactions results in some useful cross-reactivity for related tasks, but at the same time increases susceptibility to misregulation that might lead to pathologies.


Subject(s)
Biological Evolution , Cell Physiological Phenomena , Computer Simulation , Models, Biological , Animals , Humans , Protein Domains/physiology , Proteins/metabolism
18.
Nature ; 564(7734): 141-145, 2018 12.
Article in English | MEDLINE | ID: mdl-30487607

ABSTRACT

Mutations that attenuate homologous recombination (HR)-mediated repair promote tumorigenesis and sensitize cells to chemotherapeutics that cause replication fork collapse, a phenotype known as 'BRCAness'1. BRCAness tumours arise from loss-of-function mutations in 22 genes1. Of these genes, all but one (CDK12) function directly in the HR repair pathway1. CDK12 phosphorylates serine 2 of the RNA polymerase II C-terminal domain heptapeptide repeat2-7, a modification that regulates transcription elongation, splicing, and cleavage and polyadenylation8,9. Genome-wide expression studies suggest that depletion of CDK12 abrogates the expression of several HR genes relatively specifically, thereby blunting HR repair3-7,10,11. This observation suggests that the mutational status of CDK12 may predict sensitivity to targeted treatments against BRCAness, such as PARP1 inhibitors, and that CDK12 inhibitors may induce sensitization of HR-competent tumours to these treatments6,7,10,11. Despite growing clinical interest, the mechanism by which CDK12 regulates HR genes remains unknown. Here we show that CDK12 globally suppresses intronic polyadenylation events in mouse embryonic stem cells, enabling the production of full-length gene products. Many HR genes harbour more intronic polyadenylation sites than other expressed genes, and these sites are particularly sensitive to loss of CDK12. The cumulative effect of these sites accounts for the enhanced sensitivity of HR gene expression to CDK12 loss, and we find that this mechanism is conserved in human tumours that contain loss-of-function CDK12 mutations. This work clarifies the function of CDK12 and underscores its potential both as a chemotherapeutic target and as a tumour biomarker.


Subject(s)
Cyclin-Dependent Kinases/metabolism , DNA Repair/genetics , Introns/genetics , Polyadenylation/genetics , Animals , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/deficiency , Cyclin-Dependent Kinases/genetics , DNA Damage , Homologous Recombination/genetics , Humans , Loss of Function Mutation , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Mouse Embryonic Stem Cells/metabolism , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Serine/metabolism , Transcription Elongation, Genetic
19.
Science ; 361(6400)2018 07 27.
Article in English | MEDLINE | ID: mdl-29930091

ABSTRACT

Super-enhancers (SEs) are clusters of enhancers that cooperatively assemble a high density of the transcriptional apparatus to drive robust expression of genes with prominent roles in cell identity. Here we demonstrate that the SE-enriched transcriptional coactivators BRD4 and MED1 form nuclear puncta at SEs that exhibit properties of liquid-like condensates and are disrupted by chemicals that perturb condensates. The intrinsically disordered regions (IDRs) of BRD4 and MED1 can form phase-separated droplets, and MED1-IDR droplets can compartmentalize and concentrate the transcription apparatus from nuclear extracts. These results support the idea that coactivators form phase-separated condensates at SEs that compartmentalize and concentrate the transcription apparatus, suggest a role for coactivator IDRs in this process, and offer insights into mechanisms involved in the control of key cell-identity genes.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Intrinsically Disordered Proteins/metabolism , Mediator Complex Subunit 1/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Conserved Sequence , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic/drug effects , Fluorescence Recovery After Photobleaching , Gene Expression Regulation/drug effects , Glycols/pharmacology , HEK293 Cells , Humans , Immunoprecipitation , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Mediator Complex Subunit 1/chemistry , Mediator Complex Subunit 1/genetics , Mice , Molecular Imaging , NIH 3T3 Cells , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Serine/chemistry , Serine/genetics , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
20.
Nat Genet ; 50(5): 657-661, 2018 05.
Article in English | MEDLINE | ID: mdl-29662165

ABSTRACT

RNA interference (RNAi) is a major, powerful platform for gene perturbations, but is restricted by off-target mechanisms. Communication between RNAs, small RNAs, and RNA-binding proteins (RBPs) is a pervasive feature of cellular RNA networks. We present a crosstalk scenario, designated as crosstalk with endogenous RBPs' (ceRBP), in which small interfering RNAs or microRNAs with seed sequences that overlap RBP motifs have extended biological effects by perturbing endogenous RBP activity. Systematic analysis of small interfering RNA (siRNA) off-target data and genome-wide RNAi cancer lethality screens using 501 human cancer cell lines, a cancer dependency map, identified that seed-to-RBP crosstalk is widespread, contributes to off-target activity, and affects RNAi performance. Specifically, deconvolution of the interactions between gene knockdown and seed-mediated silencing effects in the cancer dependency map showed widespread contributions of seed-to-RBP crosstalk to growth-phenotype modulation. These findings suggest a novel aspect of microRNA biology and offer a basis for improvement of RNAi agents and RNAi-based functional genomics.


Subject(s)
RNA Interference , RNA-Binding Proteins/genetics , Cell Line, Tumor , Genomics/methods , Humans , MicroRNAs/genetics , Neoplasms/genetics , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...