Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(5): 114175, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38691456

ABSTRACT

Transcription factors (TFs) are important mediators of aberrant transcriptional programs in cancer cells. In this study, we focus on TF activity (TFa) as a biomarker for cell-line-selective anti-proliferative effects, in that high TFa predicts sensitivity to loss of function of a given gene (i.e., genetic dependencies [GDs]). Our linear-regression-based framework identifies 3,047 pan-cancer and 3,952 cancer-type-specific candidate TFa-GD associations from cell line data, which are then cross-examined for impact on survival in patient cohorts. One of the most prominent biomarkers is TEAD1 activity, whose associations with its predicted GDs are validated through experimental evidence as proof of concept. Overall, these TFa-GD associations represent an attractive resource for identifying innovative, biomarker-driven hypotheses for drug discovery programs in oncology.


Subject(s)
Neoplasms , Transcription Factors , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Cell Line, Tumor , TEA Domain Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Cell Proliferation
2.
ChemMedChem ; 18(6): e202200686, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36649575

ABSTRACT

The bromodomain and PHD-finger containing transcription factor (BPTF) is part of the nucleosome remodeling factor (NURF) complex and has been implicated in multiple cancer types. Here, we report the discovery of a potent and selective chemical probe targeting the bromodomain of BPTF with an attractive pharmacokinetic profile enabling cellular and in vivo experiments in mice. Microarray-based transcriptomics in presence of the probe in two lung cancer cell lines revealed only minor effects on the transcriptome. Profiling against a panel of cancer cell lines revealed that the antiproliferative effect does not correlate with BPTF dependency score in depletion screens. Both observations and the multi-domain architecture of BPTF suggest that depleting the protein by proteolysis targeting chimeras (PROTACs) could be a promising strategy to target cancer cell proliferation. We envision that the presented chemical probe and the related negative control will enable the research community to further explore scientific hypotheses with respect to BPTF bromodomain inhibition.


Subject(s)
Lung Neoplasms , Transcription Factors , Animals , Mice , Cell Proliferation , Gene Expression Regulation , Nuclear Proteins/metabolism , Transcription Factors/metabolism
3.
Oncoimmunology ; 11(1): 2019466, 2022.
Article in English | MEDLINE | ID: mdl-35154905

ABSTRACT

Antibody-mediated cancer immunotherapy targets inhibitory surface molecules, such as PD1, PD-L1, and CTLA-4, aiming to re-invigorate dysfunctional T cells. We purified and characterized tumor-infiltrating lymphocytes (TILs) and their patient-matched non-tumor counterparts from treatment-naïve NSCLC patient biopsies to evaluate the effect of PD1 expression on the functional and molecular profiles of tumor-resident T cells. We show that PD1+ CD8+ TILs have elevated expression of the transcriptional regulator ID3 and that the cytotoxic potential of CD8 T cells can be improved by knocking down ID3, defining it as a potential regulator of T cell effector function. PD1+ CD4+ memory TILs display transcriptional patterns consistent with both helper and regulator function, but can robustly facilitate B cell activation and expansion. Furthermore, we show that expanding ex vivo-prepared TILs in vitro broadly preserves their functionality with respect to tumor cell killing, B cell help, and TCR repertoire. Although purified PD1+ CD8+ TILs generally maintain an exhausted phenotype upon expansion in vitro, transcriptional analysis reveals a downregulation of markers of T-cell dysfunction, including the co-inhibitory molecules PD1 and CTLA-4 and transcription factors ID3, TOX and TOX2, while genes involved in cell cycle and DNA repair are upregulated. We find reduced expression of WNT signaling components to be a hallmark of PD1+ CD8+ exhausted T cells in vivo and in vitro and demonstrate that restoring WNT signaling, by pharmacological blockade of GSK3ß, can improve effector function. These data unveil novel targets for tumor immunotherapy and have promising implications for the development of a personalized TIL-based cell therapy for lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , CTLA-4 Antigen , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Humans , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Programmed Cell Death 1 Receptor/genetics
4.
Nat Biotechnol ; 39(2): 174-178, 2021 02.
Article in English | MEDLINE | ID: mdl-32719478

ABSTRACT

We developed a functional lineage tracing tool termed CaTCH (CRISPRa tracing of clones in heterogeneous cell populations). CaTCH combines precise clonal tracing of millions of cells with the ability to retrospectively isolate founding clones alive before and during selection, allowing functional experiments. Using CaTCH, we captured rare clones representing as little as 0.001% of a population and investigated the emergence of resistance to targeted melanoma therapy in vivo.


Subject(s)
CRISPR-Cas Systems/genetics , Cell Separation , Clone Cells/metabolism , Genes, Reporter , Animals , Cell Line , Female , Humans , Melanoma/pathology , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , raf Kinases/antagonists & inhibitors
5.
Elife ; 82019 03 25.
Article in English | MEDLINE | ID: mdl-30910006

ABSTRACT

Targeted cancer therapy is based on exploiting selective dependencies of tumor cells. By leveraging recent functional screening data of cancer cell lines we identify Werner syndrome helicase (WRN) as a novel specific vulnerability of microsatellite instability-high (MSI-H) cancer cells. MSI, caused by defective mismatch repair (MMR), occurs frequently in colorectal, endometrial and gastric cancers. We demonstrate that WRN inactivation selectively impairs the viability of MSI-H but not microsatellite stable (MSS) colorectal and endometrial cancer cell lines. In MSI-H cells, WRN loss results in severe genome integrity defects. ATP-binding deficient variants of WRN fail to rescue the viability phenotype of WRN-depleted MSI-H cancer cells. Reconstitution and depletion studies indicate that WRN dependence is not attributable to acute loss of MMR gene function but might arise during sustained MMR-deficiency. Our study suggests that pharmacological inhibition of WRN helicase function represents an opportunity to develop a novel targeted therapy for MSI-H cancers.


Subject(s)
Microsatellite Instability , Neoplasms/therapy , Werner Syndrome Helicase/antagonists & inhibitors , Cell Line, Tumor , Cell Survival , DNA Mismatch Repair , Humans , Models, Theoretical , Werner Syndrome Helicase/genetics
6.
Science ; 360(6390): 800-805, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29622725

ABSTRACT

Defining direct targets of transcription factors and regulatory pathways is key to understanding their roles in physiology and disease. We combined SLAM-seq [thiol(SH)-linked alkylation for the metabolic sequencing of RNA], a method for direct quantification of newly synthesized messenger RNAs (mRNAs), with pharmacological and chemical-genetic perturbation in order to define regulatory functions of two transcriptional hubs in cancer, BRD4 and MYC, and to interrogate direct responses to BET bromodomain inhibitors (BETis). We found that BRD4 acts as general coactivator of RNA polymerase II-dependent transcription, which is broadly repressed upon high-dose BETi treatment. At doses triggering selective effects in leukemia, BETis deregulate a small set of hypersensitive targets including MYC. In contrast to BRD4, MYC primarily acts as a selective transcriptional activator controlling metabolic processes such as ribosome biogenesis and de novo purine synthesis. Our study establishes a simple and scalable strategy to identify direct transcriptional targets of any gene or pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Genes, Regulator , Leukemia, Myeloid/drug therapy , Nuclear Proteins/metabolism , Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Antineoplastic Agents/therapeutic use , Cell Cycle Proteins , Dose-Response Relationship, Drug , Humans , Leukemia, Myeloid/genetics , Molecular Targeted Therapy , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Purines/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Ribosomes/metabolism , Sequence Analysis, RNA , Transcription Factors/genetics , Transcription, Genetic
7.
Genes Dev ; 30(1): 117-31, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26728557

ABSTRACT

The transcription cycle of RNA polymerase II (Pol II) is regulated at discrete transition points by cyclin-dependent kinases (CDKs). Positive transcription elongation factor b (P-TEFb), a complex of Cdk9 and cyclin T1, promotes release of paused Pol II into elongation, but the precise mechanisms and targets of Cdk9 action remain largely unknown. Here, by a chemical genetic strategy, we identified ∼ 100 putative substrates of human P-TEFb, which were enriched for proteins implicated in transcription and RNA catabolism. Among the RNA processing factors phosphorylated by Cdk9 was the 5'-to-3' "torpedo" exoribonuclease Xrn2, required in transcription termination by Pol II, which we validated as a bona fide P-TEFb substrate in vivo and in vitro. Phosphorylation by Cdk9 or phosphomimetic substitution of its target residue, Thr439, enhanced enzymatic activity of Xrn2 on synthetic substrates in vitro. Conversely, inhibition or depletion of Cdk9 or mutation of Xrn2-Thr439 to a nonphosphorylatable Ala residue caused phenotypes consistent with inefficient termination in human cells: impaired Xrn2 chromatin localization and increased readthrough transcription of endogenous genes. Therefore, in addition to its role in elongation, P-TEFb regulates termination by promoting chromatin recruitment and activation of a cotranscriptional RNA processing enzyme, Xrn2.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Exoribonucleases/genetics , Exoribonucleases/metabolism , Gene Expression Regulation/genetics , Positive Transcriptional Elongation Factor B/metabolism , Chromatin/metabolism , Enzyme Activation/genetics , Genetic Testing , HCT116 Cells , Humans , Phosphorylation , Protein Binding
8.
Nat Struct Mol Biol ; 22(8): 611-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26167880

ABSTRACT

Phosphorylation of the spliceosome is essential for RNA splicing, yet how and to what extent kinase signaling affects splicing have not been defined on a genome-wide basis. Using a chemical genetic approach, we show in Schizosaccharomyces pombe that the SR protein kinase Dsk1 is required for efficient splicing of introns with suboptimal splice sites. Systematic substrate mapping in fission yeast and human cells revealed that SRPKs target evolutionarily conserved spliceosomal proteins, including the branchpoint-binding protein Bpb1 (SF1 in humans), by using an RXXSP consensus motif for substrate recognition. Phosphorylation of SF1 increases SF1 binding to introns with nonconsensus splice sites in vitro, and mutation of such sites to consensus relieves the requirement for Dsk1 and phosphorylated Bpb1 in vivo. Modulation of splicing efficiency through kinase signaling pathways may allow tuning of gene expression in response to environmental and developmental cues.


Subject(s)
Introns/genetics , Protein Serine-Threonine Kinases/genetics , RNA Splicing , Schizosaccharomyces pombe Proteins/genetics , Amino Acid Motifs/genetics , Amino Acid Sequence , Blotting, Western , Mutation , Oligonucleotide Array Sequence Analysis , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , RNA Splice Sites/drug effects , RNA Splicing Factors , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Substrate Specificity
9.
PLoS Genet ; 11(3): e1005074, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25825871

ABSTRACT

Although numerous regulatory connections between pre-mRNA splicing and chromatin have been demonstrated, the precise mechanisms by which chromatin factors influence spliceosome assembly and/or catalysis remain unclear. To probe the genetic network of pre-mRNA splicing in the fission yeast Schizosaccharomyces pombe, we constructed an epistatic mini-array profile (E-MAP) and discovered many new connections between chromatin and splicing. Notably, the nucleosome remodeler SWI/SNF had strong genetic interactions with components of the U2 snRNP SF3 complex. Overexpression of SF3 components in ΔSWI/SNF cells led to inefficient splicing of many fission yeast introns, predominantly those with non-consensus splice sites. Deletion of SWI/SNF decreased recruitment of the splicing ATPase Prp2, suggesting that SWI/SNF promotes co-transcriptional spliceosome assembly prior to first step catalysis. Importantly, defects in SWI/SNF as well as SF3 overexpression each altered nucleosome occupancy along intron-containing genes, illustrating that the chromatin landscape both affects--and is affected by--co-transcriptional splicing.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Gene Regulatory Networks , Nucleosomes/genetics , RNA Splicing/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Spliceosomes/genetics , Transcription Factors/genetics , Adenosine Triphosphatases/genetics , Chromatin/genetics , Gene Expression Regulation, Fungal , Introns/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic , Schizosaccharomyces/genetics , Spliceosomes/metabolism , Transcription, Genetic
10.
Cell ; 151(3): 603-18, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23101628

ABSTRACT

Whereas proliferating cells enter M phase shortly after DNA replication, the first M phase of meiosis is preceded by an extended prophase in which homologous chromosomes undergo recombination. Exit from prophase I is controlled by the recombination checkpoint (RC), which, in yeast, represses the meiosis-specific transcription factor Ndt80 required for the expression of B-type cyclins and other M phase regulators. We show that an extended prophase I additionally requires the suppression of latent, mitotic cell-cycle controls by the anaphase-promoting complex (APC/C) and its meiosis-specific activator Ama1, which trigger the degradation of M phase regulators and Ndd1, a subunit of a mitotic transcription factor. ama1Δ mutants exit from prophase I prematurely and independently of the RC, which results in recombination defects and chromosome missegregation. Thus, control of prophase I by meiotic mechanisms depends on the suppression of the alternative, mitotic mechanisms by a meiosis-specific form of the APC/C.


Subject(s)
Cell Cycle Proteins/metabolism , Meiosis , Prophase , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Cdc20 Proteins , Chromosome Segregation , Chromosomes, Fungal/metabolism , DNA-Binding Proteins/metabolism , Metaphase , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Spindle Apparatus , Transcription Factors/metabolism
11.
Dev Cell ; 18(3): 397-409, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20230747

ABSTRACT

During meiosis, two rounds of chromosome segregation after a single round of DNA replication produce haploid gametes from diploid precursors. At meiosis I, maternal and paternal kinetochores are pulled toward opposite poles, and chiasmata holding bivalent chromosomes together are resolved by cleavage of cohesin's alpha-kleisin subunit (Rec8) along chromosome arms. This creates dyad chromosomes containing a pair of chromatids joined solely by cohesin at centromeres that had resisted cleavage. The discovery that centromeric Rec8 is protected from separase during meiosis I by shugoshin/MEI-S332 proteins that bind PP2A phosphatase suggests that phosphorylation either of separase or cohesin may be necessary for Rec8 cleavage. We show here that multiple phosphorylation sites within Rec8 as well as two different kinases, casein kinase 1delta/epsilon (CK1delta/epsilon) and Dbf4-dependent Cdc7 kinase (DDK), are required for Rec8 cleavage and meiosis I nuclear division. Rec8 with phosphomimetic mutations is no longer protected from separase at centromeres and is cleaved even when the two kinases are inhibited. Our data suggest that PP2A protects centromeric cohesion by opposing CK1delta/epsilon- and DDK-dependent phosphorylation of Rec8.


Subject(s)
Casein Kinase 1 epsilon/metabolism , Casein Kinase Idelta/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Endopeptidases/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Binding Sites , Casein Kinase 1 epsilon/genetics , Casein Kinase I/genetics , Casein Kinase I/metabolism , Casein Kinase Idelta/genetics , Cell Cycle Proteins/genetics , Centromere/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Endopeptidases/genetics , Genes, Fungal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Meiosis/genetics , Meiosis/physiology , Models, Biological , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Separase , Cohesins
12.
Cell ; 135(4): 662-78, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-19013276

ABSTRACT

Meiosis differs from mitosis in that DNA replication is followed by the segregation of homologous chromosomes but not sister chromatids. This depends on the formation of interhomolog connections through crossover recombination and on the attachment of sister kinetochores to microtubules emanating from the same spindle pole. We show that in yeast, the Dbf4-dependent Cdc7 kinase (DDK) provides a link between premeiotic S phase, recombination, and monopolar attachment. Independently from its established role in initiating DNA replication, DDK promotes double-strand break formation, the first step of recombination, and the recruitment of the monopolin complex to kinetochores, which is essential for monopolar attachment. DDK regulates monopolin localization together with the polo-kinase Cdc5 bound to Spo13, probably through phosphorylation of the monopolin subunit Lrs4. Thus, activation of DDK both initiates DNA replication and commits meiotic cells to reductional chromosome segregation in the first division of meiosis.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Meiosis , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Cell Cycle , Chromosomes/ultrastructure , DNA Replication , Gene Deletion , Kinetochores/metabolism , Kinetochores/ultrastructure , Microtubules/metabolism , Models, Biological , Models, Genetic , Nuclear Proteins/metabolism , Protein Kinases/metabolism , Saccharomyces cerevisiae
13.
J Cell Sci ; 120(Pt 7): 1245-55, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17356064

ABSTRACT

The assembly of mitotic chromosomes is controlled by condensin complexes. In vertebrates, condensin I binds to chromatin in prometaphase, confers rigidity to chromosomes and enables the release of cohesin complexes from chromosome arms, whereas condensin II associates with chromosomes in prophase and promotes their condensation. Both complexes are essential for chromosome segregation in anaphase. Although the association of condensins with chromatin is important for the assembly and segregation of mitotic chromosomes, it is poorly understood how this process is controlled. Here we show that the mitotic kinase Aurora B regulates the association of condensin I, but not the interaction of condensin II with chromatin. Quantitative time-lapse imaging of cells expressing GFP-tagged condensin subunits revealed that Aurora B is required for efficient loading of condensin I onto chromosomes in prometaphase and for maintenance of the complex on chromosomes in later stages of mitosis. The three non-SMC subunits of condensin I are Aurora B substrates in vitro and their mitosis-specific phosphorylation depends on Aurora B in vivo. Our data indicate that Aurora B contributes to chromosome rigidity and segregation by promoting the binding of condensin I to chromatin. We have also addressed how Aurora B might mediate the dissociation of cohesin from chromosome arms.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromosomes, Human/enzymology , DNA-Binding Proteins/metabolism , Mitosis , Multiprotein Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Adenosine Triphosphatases/chemistry , Aurora Kinase B , Aurora Kinases , Chromatin/metabolism , Chromosomes, Human/metabolism , DNA-Binding Proteins/chemistry , Fluorescent Dyes/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Indoles/pharmacology , Kinetics , Microscopy, Video , Multiprotein Complexes/chemistry , Phosphorylation , Protein Subunits/chemistry , Substrate Specificity , Sulfonamides/pharmacology
14.
Curr Biol ; 17(4): 304-15, 2007 Feb 20.
Article in English | MEDLINE | ID: mdl-17291761

ABSTRACT

BACKGROUND: The mitotic kinases, Cdk1, Aurora A/B, and Polo-like kinase 1 (Plk1) have been characterized extensively to further understanding of mitotic mechanisms and as potential targets for cancer therapy. Cdk1 and Aurora kinase studies have been facilitated by small-molecule inhibitors, but few if any potent Plk1 inhibitors have been identified. RESULTS: We describe the cellular effects of a novel compound, BI 2536, a potent and selective inhibitor of Plk1. The fact that BI 2536 blocks Plk1 activity fully and instantaneously enabled us to study controversial and unknown functions of Plk1. Cells treated with BI 2536 are delayed in prophase but eventually import Cdk1-cyclin B into the nucleus, enter prometaphase, and degrade cyclin A, although BI 2536 prevents degradation of the APC/C inhibitor Emi1. BI 2536-treated cells lack prophase microtubule asters and thus polymerize mitotic microtubules only after nuclear-envelope breakdown and form monopolar spindles that do not stably attach to kinetochores. Mad2 accumulates at kinetochores, and cells arrest with an activated spindle-assembly checkpoint. BI 2536 prevents Plk1's enrichment at kinetochores and centrosomes, and when added to metaphase cells, it induces detachment of microtubules from kinetochores and leads to spindle collapse. CONCLUSIONS: Our results suggest that Plk1's accumulation at centrosomes and kinetochores depends on its own activity and that this activity is required for maintaining centrosome and kinetochore function. Our data also show that Plk1 is not required for prophase entry, but delays transition to prometaphase, and that Emi1 destruction in prometaphase is not essential for APC/C-mediated cyclin A degradation.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Enzyme Inhibitors/pharmacology , F-Box Proteins/metabolism , Microtubules/drug effects , Mitosis/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Spindle Apparatus/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , HeLa Cells , Humans , Image Processing, Computer-Assisted , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microtubules/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Pteridines/metabolism , Spindle Apparatus/metabolism , Polo-Like Kinase 1
15.
Cell ; 127(5): 955-67, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17113138

ABSTRACT

Cohesin establishes sister-chromatid cohesion from S phase until mitosis or meiosis. To allow chromosome segregation, cohesion has to be dissolved. In vertebrate cells, this process is mediated in part by the protease separase, which destroys a small amount of cohesin, but most cohesin is removed from chromosomes without proteolysis. How this is achieved is poorly understood. Here, we show that the interaction between cohesin and chromatin is controlled by Wapl, a protein implicated in heterochromatin formation and tumorigenesis. Wapl is associated with cohesin throughout the cell cycle, and its depletion blocks cohesin dissociation from chromosomes during the early stages of mitosis and prevents the resolution of sister chromatids until anaphase, which occurs after a delay. Wapl depletion also increases the residence time of cohesin on chromatin in interphase. Our data indicate that Wapl is required to unlock cohesin from a particular state in which it is stably bound to chromatin.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Amino Acid Sequence , Carrier Proteins/chemistry , Chromatids/metabolism , Chromosomes, Human/genetics , Conserved Sequence , DNA-Binding Proteins , Evolution, Molecular , Humans , Interphase , Multiprotein Complexes/metabolism , Oncogene Proteins/chemistry , Phosphoproteins/metabolism , Phosphorylation , Prophase , Protein Binding , Protein Structure, Secondary , Proto-Oncogene Proteins , Repetitive Sequences, Amino Acid , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Telophase , Cohesins
16.
Nature ; 438(7071): 1176-80, 2005 Dec 22.
Article in English | MEDLINE | ID: mdl-16222244

ABSTRACT

Histones are subject to numerous post-translational modifications. Some of these 'epigenetic' marks recruit proteins that modulate chromatin structure. For example, heterochromatin protein 1 (HP1) binds to histone H3 when its lysine 9 residue has been tri-methylated by the methyltransferase Suv39h (refs 2-6). During mitosis, H3 is also phosphorylated by the kinase Aurora B. Although H3 phosphorylation is a hallmark of mitosis, its function remains mysterious. It has been proposed that histone phosphorylation controls the binding of proteins to chromatin, but any such mechanisms are unknown. Here we show that antibodies against mitotic chromosomal antigens that are associated with human autoimmune diseases specifically recognize H3 molecules that are modified by both tri-methylation of lysine 9 and phosphorylation of serine 10 (H3K9me3S10ph). The generation of H3K9me3S10ph depends on Suv39h and Aurora B, and occurs at pericentric heterochromatin during mitosis in different eukaryotes. Most HP1 typically dissociates from chromosomes during mitosis, but if phosphorylation of H3 serine 10 is inhibited, HP1 remains chromosome-bound throughout mitosis. H3 phosphorylation by Aurora B is therefore part of a 'methyl/phos switch' mechanism that displaces HP1 and perhaps other proteins from mitotic heterochromatin.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Heterochromatin/metabolism , Histones/metabolism , Phosphoserine/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Aurora Kinase B , Aurora Kinases , Cell Line , Chromobox Protein Homolog 5 , Chromosomes, Mammalian/metabolism , Epitopes , Histones/chemistry , Humans , Mice , Mitosis , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...