Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cell ; 169(4): 651-663.e14, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475894

ABSTRACT

The liver plays a pivotal role in metabolism and xenobiotic detoxification, processes that must be particularly efficient when animals are active and feed. A major question is how the liver adapts to these diurnal changes in physiology. Here, we show that, in mice, liver mass, hepatocyte size, and protein levels follow a daily rhythm, whose amplitude depends on both feeding-fasting and light-dark cycles. Correlative evidence suggests that the daily oscillation in global protein accumulation depends on a similar fluctuation in ribosome number. Whereas rRNA genes are transcribed at similar rates throughout the day, some newly synthesized rRNAs are polyadenylated and degraded in the nucleus in a robustly diurnal fashion with a phase opposite to that of ribosomal protein synthesis. Based on studies with cultured fibroblasts, we propose that rRNAs not packaged into complete ribosomal subunits are polyadenylated by the poly(A) polymerase PAPD5 and degraded by the nuclear exosome.


Subject(s)
Liver/cytology , Liver/physiology , Ribosomes/metabolism , Animals , Cell Nucleus/metabolism , Cell Size , Circadian Rhythm , Exosomes/metabolism , Hepatocytes/cytology , Hepatocytes/physiology , Male , Mice , Mice, Inbred C57BL , Photoperiod , RNA Processing, Post-Transcriptional , RNA, Ribosomal/genetics , Ribosomal Proteins/genetics , Ribosomes/chemistry
2.
EMBO J ; 43(12): 2506-2525, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38689024

ABSTRACT

Many microRNAs (miRNAs) are expressed with high spatiotemporal specificity during organismal development, with some being limited to rare cell types, often embedded in complex tissues. Yet, most miRNA profiling efforts remain at the tissue and organ levels. To overcome challenges in accessing the microRNomes from tissue-embedded cells, we had previously developed mime-seq (miRNome by methylation-dependent sequencing), a technique in which cell-specific miRNA methylation in C. elegans and Drosophila enabled chemo-selective sequencing without the need for cell sorting or biochemical purification. Here, we present mime-seq 2.0 for profiling miRNAs from specific mouse cell types. We engineered a chimeric RNA methyltransferase that is tethered to Argonaute protein and efficiently methylates miRNAs at their 3'-terminal 2'-OH in mouse and human cell lines. We also generated a transgenic mouse for conditional expression of this methyltransferase, which can be used to direct methylation of miRNAs in a cell type of choice. We validated the use of this mouse model by profiling miRNAs from B cells and bone marrow plasma cells.


Subject(s)
MicroRNAs , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Mice , Humans , Mice, Transgenic , Methylation , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Sequence Analysis, RNA/methods , Methyltransferases/genetics , Methyltransferases/metabolism , Cell Line , B-Lymphocytes/metabolism
3.
Nat Methods ; 15(4): 283-289, 2018 04.
Article in English | MEDLINE | ID: mdl-29481550

ABSTRACT

MicroRNAs (miRNAs) play an essential role in the post-transcriptional regulation of animal development and physiology. However, in vivo studies aimed at linking miRNA function to the biology of distinct cell types within complex tissues remain challenging, partly because in vivo miRNA-profiling methods lack cellular resolution. We report microRNome by methylation-dependent sequencing (mime-seq), an in vivo enzymatic small-RNA-tagging approach that enables high-throughput sequencing of tissue- and cell-type-specific miRNAs in animals. The method combines cell-type-specific 3'-terminal 2'-O-methylation of animal miRNAs by a genetically encoded, plant-specific methyltransferase (HEN1), with chemoselective small-RNA cloning and high-throughput sequencing. We show that mime-seq uncovers the miRNomes of specific cells within Caenorhabditis elegans and Drosophila at unprecedented specificity and sensitivity, enabling miRNA profiling with single-cell resolution in whole animals. Mime-seq overcomes current challenges in cell-type-specific small-RNA profiling and provides novel entry points for understanding the function of miRNAs in spatially restricted physiological settings.


Subject(s)
Caenorhabditis elegans/genetics , Drosophila/genetics , MicroRNAs/genetics , Sequence Analysis, RNA/methods , Animals , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cloning, Molecular , Neurons/metabolism
4.
Proc Natl Acad Sci U S A ; 115(8): E1916-E1925, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29432155

ABSTRACT

The mammalian circadian clock coordinates physiology with environmental cycles through the regulation of daily oscillations of gene expression. Thousands of transcripts exhibit rhythmic accumulations across mouse tissues, as determined by the balance of their synthesis and degradation. While diurnally rhythmic transcription regulation is well studied and often thought to be the main factor generating rhythmic mRNA accumulation, the extent of rhythmic posttranscriptional regulation is debated, and the kinetic parameters (e.g., half-lives), as well as the underlying regulators (e.g., mRNA-binding proteins) are relatively unexplored. Here, we developed a quantitative model for cyclic accumulations of pre-mRNA and mRNA from total RNA-seq data, and applied it to mouse liver. This allowed us to identify that about 20% of mRNA rhythms were driven by rhythmic mRNA degradation, and another 15% of mRNAs regulated by both rhythmic transcription and mRNA degradation. The method could also estimate mRNA half-lives and processing times in intact mouse liver. We then showed that, depending on mRNA half-life, rhythmic mRNA degradation can either amplify or tune phases of mRNA rhythms. By comparing mRNA rhythms in wild-type and Bmal1-/- animals, we found that the rhythmic degradation of many transcripts did not depend on a functional BMAL1. Interestingly clock-dependent and -independent degradation rhythms peaked at distinct times of day. We further predicted mRNA-binding proteins (mRBPs) that were implicated in the posttranscriptional regulation of mRNAs, either through stabilizing or destabilizing activities. Together, our results demonstrate how posttranscriptional regulation temporally shapes rhythmic mRNA accumulation in mouse liver.


Subject(s)
Circadian Clocks , Gene Expression Regulation , Liver/metabolism , Mice/genetics , RNA, Messenger/genetics , Animals , Male , Mice/metabolism , Mice, Inbred C57BL , Promoter Regions, Genetic , RNA, Messenger/metabolism , Transcription, Genetic
5.
Proc Natl Acad Sci U S A ; 112(47): E6579-88, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26554015

ABSTRACT

Diurnal oscillations of gene expression are a hallmark of rhythmic physiology across most living organisms. Such oscillations are controlled by the interplay between the circadian clock and feeding rhythms. Although rhythmic mRNA accumulation has been extensively studied, comparatively less is known about their transcription and translation. Here, we quantified simultaneously temporal transcription, accumulation, and translation of mouse liver mRNAs under physiological light-dark conditions and ad libitum or night-restricted feeding in WT and brain and muscle Arnt-like 1 (Bmal1)-deficient animals. We found that rhythmic transcription predominantly drives rhythmic mRNA accumulation and translation for a majority of genes. Comparison of wild-type and Bmal1 KO mice shows that circadian clock and feeding rhythms have broad impact on rhythmic gene expression, Bmal1 deletion affecting surprisingly both transcriptional and posttranscriptional levels. Translation efficiency is differentially regulated during the diurnal cycle for genes with 5'-Terminal Oligo Pyrimidine tract (5'-TOP) sequences and for genes involved in mitochondrial activity, many harboring a Translation Initiator of Short 5'-UTR (TISU) motif. The increased translation efficiency of 5'-TOP and TISU genes is mainly driven by feeding rhythms but Bmal1 deletion also affects amplitude and phase of translation, including TISU genes. Together this study emphasizes the complex interconnections between circadian and feeding rhythms at several steps ultimately determining rhythmic gene expression and translation.


Subject(s)
Circadian Rhythm/genetics , Feeding Behavior , Protein Biosynthesis , Transcription, Genetic , 5' Untranslated Regions/genetics , ARNTL Transcription Factors/metabolism , Adenylate Kinase/metabolism , Animals , Gene Deletion , Mechanistic Target of Rapamycin Complex 1 , Mice, Knockout , Models, Genetic , Multiprotein Complexes , Nucleotide Motifs/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Ribosomes/metabolism , TOR Serine-Threonine Kinases
6.
Proc Natl Acad Sci U S A ; 111(1): 167-72, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24344304

ABSTRACT

Diurnal oscillations of gene expression controlled by the circadian clock underlie rhythmic physiology across most living organisms. Although such rhythms have been extensively studied at the level of transcription and mRNA accumulation, little is known about the accumulation patterns of proteins. Here, we quantified temporal profiles in the murine hepatic proteome under physiological light-dark conditions using stable isotope labeling by amino acids quantitative MS. Our analysis identified over 5,000 proteins, of which several hundred showed robust diurnal oscillations with peak phases enriched in the morning and during the night and related to core hepatic physiological functions. Combined mathematical modeling of temporal protein and mRNA profiles indicated that proteins accumulate with reduced amplitudes and significant delays, consistent with protein half-life data. Moreover, a group comprising about one-half of the rhythmic proteins showed no corresponding rhythmic mRNAs, indicating significant translational or posttranslational diurnal control. Such rhythms were highly enriched in secreted proteins accumulating tightly during the night. Also, these rhythms persisted in clock-deficient animals subjected to rhythmic feeding, suggesting that food-related entrainment signals influence rhythms in circulating plasma factors.


Subject(s)
Circadian Clocks , Gene Expression Regulation , Liver/metabolism , Plasma/metabolism , Proteome , Albumins/metabolism , Animals , Circadian Rhythm , Cryptochromes/genetics , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Theoretical , Protein Processing, Post-Translational , RNA, Messenger/metabolism , alpha 1-Antitrypsin/metabolism
7.
Biophys J ; 107(10): 2403-16, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25418309

ABSTRACT

Biochemical reaction networks are subjected to large fluctuations attributable to small molecule numbers, yet underlie reliable biological functions. Thus, it is important to understand how regularity can emerge from noise. Here, we study the stochastic dynamics of a self-repressing gene with arbitrarily long or short response time. We find that when the mRNA and protein half-lives are approximately equal to the gene response time, fluctuations can induce relatively regular oscillations in the protein concentration. To gain insight into this phenomenon at the crossroads of determinism and stochasticity, we use an intermediate theoretical approach, based on a moment-closure approximation of the master equation, which allows us to take into account the binary character of gene activity. We thereby obtain differential equations that describe how nonlinearity can feed-back fluctuations into the mean-field equations to trigger oscillations. Finally, our results suggest that the self-repressing Hes1 gene circuit exploits this phenomenon to generate robust oscillations, inasmuch as its time constants satisfy precisely the conditions we have identified.


Subject(s)
Models, Biological , Repressor Proteins/genetics , Repressor Proteins/metabolism , Feedback, Physiological , Gene Expression Regulation , Kinetics , Stochastic Processes
8.
Dev Cell ; 2024 May 21.
Article in English | MEDLINE | ID: mdl-38788714

ABSTRACT

The salamander limb correctly regenerates missing limb segments because connective tissue cells have segment-specific identities, termed "positional information". How positional information is molecularly encoded at the chromatin level has been unknown. Here, we performed genome-wide chromatin profiling in mature and regenerating axolotl limb connective tissue cells. We find segment-specific levels of histone H3K27me3 as the major positional mark, especially at limb homeoprotein gene loci but not their upstream regulators, constituting an intrinsic segment information code. During regeneration, regeneration-specific regulatory elements became active prior to the re-appearance of developmental regulatory elements. In the hand, the permissive chromatin state of the homeoprotein gene HoxA13 engages with the regeneration program bypassing the upper limb program. Comparison of regeneration regulatory elements with those found in other regenerative animals identified a core shared set of transcription factors, supporting an ancient, conserved regeneration program.

9.
Dev Cell ; 2024 May 17.
Article in English | MEDLINE | ID: mdl-38776925

ABSTRACT

During neural tube (NT) development, the notochord induces an organizer, the floorplate, which secretes Sonic Hedgehog (SHH) to pattern neural progenitors. Conversely, NT organoids (NTOs) from embryonic stem cells (ESCs) spontaneously form floorplates without the notochord, demonstrating that stem cells can self-organize without embryonic inducers. Here, we investigated floorplate self-organization in clonal mouse NTOs. Expression of the floorplate marker FOXA2 was initially spatially scattered before resolving into multiple clusters, which underwent competition and sorting, resulting in a stable "winning" floorplate. We identified that BMP signaling governed long-range cluster competition. FOXA2+ clusters expressed BMP4, suppressing FOXA2 in receiving cells while simultaneously expressing the BMP-inhibitor NOGGIN, promoting cluster persistence. Noggin mutation perturbed floorplate formation in NTOs and in the NT in vivo at mid/hindbrain regions, demonstrating how the floorplate can form autonomously without the notochord. Identifying the pathways governing organizer self-organization is critical for harnessing the developmental plasticity of stem cells in tissue engineering.

10.
Nat Commun ; 14(1): 8160, 2023 Dec 09.
Article in English | MEDLINE | ID: mdl-38071364

ABSTRACT

Polycomb Repressive Complexes 1 and 2 (PRC1, PRC2) are conserved epigenetic regulators that promote transcriptional gene silencing. PRC1 and PRC2 converge on shared targets, catalyzing repressive histone modifications. Additionally, a subset of PRC1/PRC2 targets engage in long-range interactions whose functions in gene silencing are poorly understood. Using a CRISPR screen in mouse embryonic stem cells, we found that the cohesin regulator PDS5A links transcriptional silencing by Polycomb and 3D genome organization. PDS5A deletion impairs cohesin unloading and results in derepression of a subset of endogenous PRC1/PRC2 target genes. Importantly, derepression is not linked to loss of Polycomb chromatin domains. Instead, PDS5A removal causes aberrant cohesin activity leading to ectopic insulation sites, which disrupt the formation of ultra-long Polycomb loops. We show that these loops are important for robust silencing at a subset of PRC1/PRC2 target genes and that maintenance of cohesin-dependent genome architecture is critical for Polycomb regulation.


Subject(s)
Cohesins , Nuclear Proteins , Polycomb-Group Proteins , Animals , Mice , Chromatin/genetics , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 2/genetics , Polycomb-Group Proteins/genetics , Nuclear Proteins/genetics
11.
Elife ; 122023 08 25.
Article in English | MEDLINE | ID: mdl-37622993

ABSTRACT

Adaptation of the functional proteome is essential to counter pathogens during infection, yet precisely timed degradation of these response proteins after pathogen clearance is likewise key to preventing autoimmunity. Interferon regulatory factor 1 (IRF1) plays an essential role as a transcription factor in driving the expression of immune response genes during infection. The striking difference in functional output with other IRFs is that IRF1 also drives the expression of various cell cycle inhibiting factors, making it an important tumor suppressor. Thus, it is critical to regulate the abundance of IRF1 to achieve a 'Goldilocks' zone in which there is sufficient IRF1 to prevent tumorigenesis, yet not too much which could drive excessive immune activation. Using genetic screening, we identified the E3 ligase receptor speckle type BTB/POZ protein (SPOP) to mediate IRF1 proteasomal turnover in human and mouse cells. We identified S/T-rich degrons in IRF1 required for its SPOP MATH domain-dependent turnover. In the absence of SPOP, elevated IRF1 protein levels functionally increased IRF1-dependent cellular responses, underpinning the biological significance of SPOP in curtailing IRF1 protein abundance.


Subject(s)
Gene Expression Regulation , Genes, Regulator , Humans , Animals , Mice , Interferon Regulatory Factor-1/genetics , Acclimatization , Immunologic Factors
12.
Nat Cell Biol ; 25(1): 42-55, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36604593

ABSTRACT

ZNF462 haploinsufficiency is linked to Weiss-Kruszka syndrome, a genetic disorder characterized by neurodevelopmental defects, including autism. Though conserved in vertebrates and essential for embryonic development, the molecular functions of ZNF462 remain unclear. We identified its murine homologue ZFP462 in a screen for mediators of epigenetic gene silencing. Here we show that ZFP462 safeguards neural lineage specification of mouse embryonic stem cells (ESCs) by targeting the H3K9-specific histone methyltransferase complex G9A/GLP to silence meso-endodermal genes. ZFP462 binds to transposable elements that are potential enhancers harbouring pluripotency and meso-endoderm transcription factor binding sites. Recruiting G9A/GLP, ZFP462 seeds heterochromatin, restricting transcription factor binding. Loss of ZFP462 in ESCs results in increased chromatin accessibility at target sites and ectopic expression of meso-endodermal genes. Taken together, ZFP462 confers lineage and locus specificity to the broadly expressed epigenetic regulator G9A/GLP. Our results suggest that aberrant activation of lineage non-specific genes in the neuronal lineage underlies ZNF462-associated neurodevelopmental pathology.


Subject(s)
Heterochromatin , Histone-Lysine N-Methyltransferase , Animals , Mice , Heterochromatin/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Chromatin , Embryonic Stem Cells , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , Nerve Tissue Proteins/genetics
13.
Sci Adv ; 7(42): eabh1434, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34652942

ABSTRACT

Muscle function requires unique structural and metabolic adaptations that can render muscle cells selectively vulnerable, with mutations in some ubiquitously expressed genes causing myopathies but sparing other tissues. We uncovered a muscle cell vulnerability by studying miR-1, a deeply conserved, muscle-specific microRNA whose ablation causes various muscle defects. Using Caenorhabditis elegans, we found that miR-1 represses multiple subunits of the ubiquitous vacuolar adenosine triphosphatase (V-ATPase) complex, which is essential for internal compartment acidification and metabolic signaling. V-ATPase subunits are predicted miR-1 targets in animals ranging from C. elegans to humans, and we experimentally validated this in Drosophila. Unexpectedly, up-regulation of V-ATPase subunits upon miR-1 deletion causes reduced V-ATPase function due to defects in complex assembly. These results reveal V-ATPase assembly as a conserved muscle cell vulnerability and support a previously unknown role for microRNAs in the regulation of protein complexes.

14.
J Sep Sci ; 33(9): 1325-30, 2010 May.
Article in English | MEDLINE | ID: mdl-20201046

ABSTRACT

A preparative high-speed counter-current chromatography method for isolation and purification of bufadienolides from ChanSu was developed by using a stepwise elution with two-phase solvent system composed of n-hexane/ethyl acetate/methanol/water at the ratios of 4:6:2:4 v/v, 4:6:2.5:4 v/v and 4:6:3.2:4 v/v. A total of 3.8 mg of gamabufotalin (1), 7.2 mg of arenobufagin (2), 3.4 mg of telocinobufagin (3), 5.3 mg of bufotalin (4), 8.5 mg of cinobufotalin (5) and 8 mg of bufalin (6) were obtained in one-step separation from 80 mg of the crude extract with purity of 92.7, 96.7, 87.2, 97.3, 94.9 and 99.4%, respectively. Their chemical structures were identified on the basis of (1)H-NMR and (13)C-NMR technology.


Subject(s)
Amphibian Venoms/chemistry , Bufanolides/analysis , Countercurrent Distribution/methods , Medicine, Chinese Traditional , Bufanolides/chemistry , Chromatography, High Pressure Liquid , Magnetic Resonance Spectroscopy
15.
Curr Biol ; 30(24): 5058-5065.e5, 2020 12 21.
Article in English | MEDLINE | ID: mdl-33125867

ABSTRACT

MicroRNAs (miRNAs) are a class of post-transcriptional repressors with diverse roles in animal development and physiology [1]. The Microprocessor complex, composed of Drosha and Pasha/DGCR8, is necessary for the biogenesis of all canonical miRNAs and essential for the early stages of animal embryogenesis [2-8]. However, the cause for this requirement is largely unknown. Animals often express hundreds of miRNAs, and it remains unclear whether the Microprocessor is required to produce one or few essential miRNAs or many individually non-essential miRNAs. Additionally, both Drosha and Pasha/DGCR8 bind and cleave a variety of non-miRNA substrates [9-15], and it is unknown whether these activities account for the Microprocessor's essential requirement. To distinguish between these possibilities, we developed a system in C. elegans to stringently deplete embryos of Microprocessor activity. Using a combination of auxin-inducible degradation (AID) and RNA interference (RNAi), we achieved Drosha and Pasha/DGCR8 depletion starting in the maternal germline, resulting in Microprocessor and miRNA-depleted embryos, which fail to undergo morphogenesis or form organs. Using a Microprocessor-bypass strategy, we show that this early embryonic arrest is rescued by the addition of just two miRNAs, one miR-35 and one miR-51 family member, resulting in morphologically normal larvae. Thus, just two out of ∼150 canonical miRNAs are sufficient for morphogenesis and organogenesis, and the processing of these miRNAs accounts for the essential requirement for Drosha and Pasha/DGCR8 during the early stages of C. elegans embryonic development. VIDEO ABSTRACT.


Subject(s)
Caenorhabditis elegans/growth & development , Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Organogenesis/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Gene Knockdown Techniques , Ribonuclease III/genetics , Ribonuclease III/metabolism
16.
Dev Cell ; 55(4): 483-499.e7, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33002421

ABSTRACT

Combinatorial action of transcription factors (TFs) with partially overlapping expression is a widespread strategy to generate novel gene-expression patterns and, thus, cellular diversity. Known mechanisms underlying combinatorial activity require co-expression of TFs within the same cell. Here, we describe the mechanism by which two TFs that are never co-expressed generate a new, intersectional expression pattern in C. elegans embryos: lineage-specific priming of a gene by a transiently expressed TF generates a unique intersection with a second TF acting on the same gene four cell divisions later; the second TF is expressed in multiple cells but only activates transcription in those where priming occurred. Early induction of active transcription is necessary and sufficient to establish a competent state, maintained by broadly expressed regulators in the absence of the initial trigger. We uncover additional cells diversified through this mechanism. Our findings define a mechanism for combinatorial TF activity with important implications for generation of cell-type diversity.


Subject(s)
Caenorhabditis elegans/metabolism , Transcription Factors/metabolism , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/metabolism , Cell Lineage , Embryo, Nonmammalian/metabolism , Gene Expression Regulation , Genetic Loci , Neurons/metabolism , Protein Binding , Time Factors , Transcription, Genetic
17.
Nat Commun ; 10(1): 1931, 2019 04 29.
Article in English | MEDLINE | ID: mdl-31036804

ABSTRACT

Polycomb group (PcG) proteins play critical roles in the epigenetic inheritance of cell fate. The Polycomb Repressive Complexes PRC1 and PRC2 catalyse distinct chromatin modifications to enforce gene silencing, but how transcriptional repression is propagated through mitotic cell divisions remains a key unresolved question. Using reversible tethering of PcG proteins to ectopic sites in mouse embryonic stem cells, here we show that PRC1 can trigger transcriptional repression and Polycomb-dependent chromatin modifications. We find that canonical PRC1 (cPRC1), but not variant PRC1, maintains gene silencing through cell division upon reversal of tethering. Propagation of gene repression is sustained by cis-acting histone modifications, PRC2-mediated H3K27me3 and cPRC1-mediated H2AK119ub1, promoting a sequence-independent feedback mechanism for PcG protein recruitment. Thus, the distinct PRC1 complexes present in vertebrates can differentially regulate epigenetic maintenance of gene silencing, potentially enabling dynamic heritable responses to complex stimuli. Our findings reveal how PcG repression is potentially inherited in vertebrates.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic , Gene Silencing , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 2/genetics , Protein Processing, Post-Translational , Animals , Cell Line , Chromatin/chemistry , Feedback, Physiological , Histones/genetics , Histones/metabolism , Inheritance Patterns , Mice , Mitosis , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/metabolism , Transcription, Genetic
18.
Cell Chem Biol ; 26(10): 1365-1379.e22, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31422906

ABSTRACT

Polycomb-directed repression of gene expression is frequently misregulated in human diseases. A quantitative and target-specific cellular assay was utilized to discover the first potent positive allosteric modulator (PAM) peptidomimetic, UNC4976, of nucleic acid binding by CBX7, a chromodomain methyl-lysine reader of Polycomb repressive complex 1. The PAM activity of UNC4976 resulted in enhanced efficacy across three orthogonal cellular assays by simultaneously antagonizing H3K27me3-specific recruitment of CBX7 to target genes while increasing non-specific binding to DNA and RNA. PAM activity thereby reequilibrates PRC1 away from H3K27me3 target regions. Together, our discovery and characterization of UNC4976 not only revealed the most cellularly potent PRC1-specific chemical probe to date, but also uncovers a potential mechanism of Polycomb regulation with implications for non-histone lysine methylated interaction partners.


Subject(s)
Drug Discovery , Peptidomimetics/pharmacology , Polycomb Repressive Complex 1/metabolism , Allosteric Regulation/drug effects , Animals , HEK293 Cells , HeLa Cells , Humans , Mice , Peptidomimetics/chemistry
19.
Cell Rep ; 20(7): 1729-1743, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28813682

ABSTRACT

Lysine acetylation is involved in various biological processes and is considered a key reversible post-translational modification in the regulation of gene expression, enzyme activity, and subcellular localization. This post-translational modification is therefore highly relevant in the context of circadian biology, but its characterization on the proteome-wide scale and its circadian clock dependence are still poorly described. Here, we provide a comprehensive and rhythmic acetylome map of the mouse liver. Rhythmic acetylated proteins showed subcellular localization-specific phases that correlated with the related metabolites in the regulated pathways. Mitochondrial proteins were over-represented among the rhythmically acetylated proteins and were highly correlated with SIRT3-dependent deacetylation. SIRT3 activity being nicotinamide adenine dinucleotide (NAD)+ level-dependent, we show that NAD+ is orchestrated by both feeding rhythms and the circadian clock through the NAD+ salvage pathway but also via the nicotinamide riboside pathway. Hence, the diurnal acetylome relies on a functional circadian clock and affects important diurnal metabolic pathways in the mouse liver.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Liver/enzymology , Mitochondrial Proteins/metabolism , Protein Processing, Post-Translational , Proteome/metabolism , ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/genetics , Acetylation , Animals , Cryptochromes/deficiency , Cryptochromes/genetics , Eating/physiology , Lysine , Metabolic Networks and Pathways/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/genetics , NAD/metabolism , Photoperiod , Proteome/genetics , Sirtuin 3/genetics , Sirtuin 3/metabolism
20.
Cell Metab ; 25(1): 102-117, 2017 01 10.
Article in English | MEDLINE | ID: mdl-27818260

ABSTRACT

Diurnal oscillations of gene expression controlled by the circadian clock and its connected feeding rhythm enable organisms to coordinate their physiologies with daily environmental cycles. While available techniques yielded crucial insights into regulation at the transcriptional level, much less is known about temporally controlled functions within the nucleus and their regulation at the protein level. Here, we quantified the temporal nuclear accumulation of proteins and phosphoproteins from mouse liver by SILAC proteomics. We identified around 5,000 nuclear proteins, over 500 of which showed a diurnal accumulation. Parallel analysis of the nuclear phosphoproteome enabled the inference of the temporal activity of kinases accounting for rhythmic phosphorylation. Many identified rhythmic proteins were parts of nuclear complexes involved in transcriptional regulation, ribosome biogenesis, DNA repair, and the cell cycle and its potentially associated diurnal rhythm of hepatocyte polyploidy. Taken together, these findings provide unprecedented insights into the diurnal regulatory landscape of the mouse liver nucleus.


Subject(s)
Cell Nucleus/metabolism , Circadian Rhythm , Liver/metabolism , Proteomics/methods , Animals , Circadian Clocks/genetics , Circadian Rhythm/genetics , DNA Repair , Gene Expression Regulation , Isotope Labeling , Mass Spectrometry , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Organelle Biogenesis , Phosphoproteins/metabolism , Phosphorylation , Polyploidy , Protein Kinases/metabolism , Proteome/metabolism , Ribosomes/metabolism , Time Factors , Transcription Factors/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL