Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Nat Genet ; 53(8): 1221-1232, 2021 08.
Article in English | MEDLINE | ID: mdl-34294917

ABSTRACT

Driver mutations in genes encoding histone H3 proteins resulting in p.Lys27Met substitutions (H3-K27M) are frequent in pediatric midline brain tumors. However, the precise mechanisms by which H3-K27M causes tumor initiation remain unclear. Here, we use human hindbrain neural stem cells to model the consequences of H3.3-K27M on the epigenomic landscape in a relevant developmental context. Genome-wide mapping of epitope-tagged histone H3.3 revealed that both the wild type and the K27M mutant incorporate abundantly at pre-existing active enhancers and promoters, and to a lesser extent at Polycomb repressive complex 2 (PRC2)-bound regions. At active enhancers, H3.3-K27M leads to focal H3K27ac loss, decreased chromatin accessibility and reduced transcriptional expression of nearby neurodevelopmental genes. In addition, H3.3-K27M deposition at a subset of PRC2 target genes leads to increased PRC2 and PRC1 binding and augmented transcriptional repression that can be partially reversed by PRC2 inhibitors. Our work suggests that, rather than imposing de novo transcriptional circuits, H3.3-K27M drives tumorigenesis by locking initiating cells in their pre-existing, immature epigenomic state, via disruption of PRC2 and enhancer functions.


Subject(s)
Enhancer Elements, Genetic , Histones/metabolism , Neural Stem Cells/physiology , Polycomb Repressive Complex 2/genetics , Rhombencephalon/cytology , Animals , Brain Neoplasms/genetics , Cell Differentiation/genetics , Cell Line , Enhancer of Zeste Homolog 2 Protein/genetics , Epigenome , Gene Expression Regulation, Developmental , Glioma/genetics , Histones/genetics , Humans , Lysine/metabolism , Male , Mice, Inbred Strains , Mutation , Neural Stem Cells/transplantation , Oncogenes , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , Rhombencephalon/physiology
2.
Genes Dev ; 33(15-16): 936-959, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31123059

ABSTRACT

Changes in chromatin structure mediated by ATP-dependent nucleosome remodelers and histone modifying enzymes are integral to the process of gene regulation. Here, we review the roles of the SWI/SNF (switch/sucrose nonfermenting) and NuRD (nucleosome remodeling and deacetylase) and the Polycomb system in chromatin regulation and cancer. First, we discuss the basic molecular mechanism of nucleosome remodeling, and how this controls gene transcription. Next, we provide an overview of the functional organization and biochemical activities of SWI/SNF, NuRD, and Polycomb complexes. We describe how, in metazoans, the balance of these activities is central to the proper regulation of gene expression and cellular identity during development. Whereas SWI/SNF counteracts Polycomb, NuRD facilitates Polycomb repression on chromatin. Finally, we discuss how disruptions of this regulatory equilibrium contribute to oncogenesis, and how new insights into the biological functions of remodelers and Polycombs are opening avenues for therapeutic interventions on a broad range of cancer types.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Neoplasms/physiopathology , Polycomb-Group Proteins/metabolism , Transcription Factors/metabolism , Animals , Gene Expression Regulation, Developmental , Humans
3.
Nat Rev Cancer ; 19(5): 255-269, 2019 05.
Article in English | MEDLINE | ID: mdl-30962549

ABSTRACT

Recurrent chromosomal rearrangements leading to the generation of oncogenic fusion proteins are a common feature of many cancers. These aberrations are particularly prevalent in sarcomas and haematopoietic malignancies and frequently involve genes required for chromatin regulation and transcriptional control. In many cases, these fusion proteins are thought to be the primary driver of cancer development, altering chromatin dynamics to initiate oncogenic gene expression programmes. In recent years, mechanistic insights into the underlying molecular functions of a number of these oncogenic fusion proteins have been discovered. These insights have allowed the design of mechanistically anchored therapeutic approaches promising substantial treatment advances. In this Review, we discuss how our understanding of fusion protein function is informing therapeutic innovations and illuminating mechanisms of chromatin and transcriptional regulation in cancer and normal cells.


Subject(s)
Chromatin/genetics , Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Animals , Chromosome Aberrations , Gene Expression/genetics , Humans , Neoplasms/pathology , Transcription, Genetic/genetics
4.
Trends Mol Med ; 25(5): 362-365, 2019 05.
Article in English | MEDLINE | ID: mdl-30885427

ABSTRACT

EZH2 is an oncogene in non-Hodgkin lymphoma. Understanding the underlying pathogenic mechanisms will be essential to improve treatments for patients with EZH2 mutant lymphomas. Recently Donaldson-Collier and colleagues (Nat. Genet. 2019; published online January 28, https://doi.org/10.1038/s41588-018-0338-y) examined the effects of mutant EZH2 on the 3D architecture of the lymphoma genome, highlighting the potential relevance of chromatin folding dynamics.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Genomics , Lymphoma, Non-Hodgkin/genetics , Mutation , Chromatin/genetics , Chromatin/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Genomics/methods , Humans , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/pathology
5.
Elife ; 72018 11 15.
Article in English | MEDLINE | ID: mdl-30431433

ABSTRACT

Synovial sarcoma tumours contain a characteristic fusion protein, SS18-SSX, which drives disease development. Targeting oncogenic fusion proteins presents an attractive therapeutic opportunity. However, SS18-SSX has proven intractable for therapeutic intervention. Using a domain-focused CRISPR screen we identified the bromodomain of BRD9 as a critical functional dependency in synovial sarcoma. BRD9 is a component of SS18-SSX containing BAF complexes in synovial sarcoma cells; and integration of BRD9 into these complexes is critical for cell growth. Moreover BRD9 and SS18-SSX co-localize extensively on the synovial sarcoma genome. Remarkably, synovial sarcoma cells are highly sensitive to a novel small molecule degrader of BRD9, while other sarcoma subtypes are unaffected. Degradation of BRD9 induces downregulation of oncogenic transcriptional programs and inhibits tumour progression in vivo. We demonstrate that BRD9 supports oncogenic mechanisms underlying the SS18-SSX fusion in synovial sarcoma and highlight targeted degradation of BRD9 as a potential therapeutic opportunity in this disease.


Subject(s)
Gene Expression Regulation, Neoplastic , Proteolysis , Sarcoma, Synovial/genetics , Transcription Factors/metabolism , Disease Progression , HEK293 Cells , Humans , Protein Binding , Protein Domains , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sarcoma, Synovial/pathology , Transcription Factors/chemistry , Transcription, Genetic
6.
Angew Chem Int Ed Engl ; 56(21): 5738-5743, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28418626

ABSTRACT

The bromodomain-containing protein BRD9, a subunit of the human BAF (SWI/SNF) nucleosome remodeling complex, has emerged as an attractive therapeutic target in cancer. Despite the development of chemical probes targeting the BRD9 bromodomain, there is a limited understanding of BRD9 function beyond acetyl-lysine recognition. We have therefore created the first BRD9-directed chemical degraders, through iterative design and testing of heterobifunctional ligands that bridge the BRD9 bromodomain and the cereblon E3 ubiquitin ligase complex. Degraders of BRD9 exhibit markedly enhanced potency compared to parental ligands (10- to 100-fold). Parallel study of degraders with divergent BRD9-binding chemotypes in models of acute myeloid leukemia resolves bromodomain polypharmacology in this emerging drug class. Together, these findings reveal the tractability of non-BET bromodomain containing proteins to chemical degradation, and highlight lead compound dBRD9 as a tool for the study of BRD9.


Subject(s)
DNA-Binding Proteins/chemistry , Nuclear Proteins/chemistry , Transcription Factors/chemistry , Drug Delivery Systems , Humans , Ligands , Molecular Structure , Pyrroles/chemistry
7.
Cancer Cell ; 30(6): 863-878, 2016 Dec 12.
Article in English | MEDLINE | ID: mdl-27889185

ABSTRACT

The nucleoporin 98 gene (NUP98) is fused to a variety of partner genes in multiple hematopoietic malignancies. Here, we demonstrate that NUP98 fusion proteins, including NUP98-HOXA9 (NHA9), NUP98-HOXD13 (NHD13), NUP98-NSD1, NUP98-PHF23, and NUP98-TOP1 physically interact with mixed lineage leukemia 1 (MLL1) and the non-specific lethal (NSL) histone-modifying complexes. Chromatin immunoprecipitation sequencing illustrates that NHA9 and MLL1 co-localize on chromatin and are found associated with Hox gene promoter regions. Furthermore, MLL1 is required for the proliferation of NHA9 cells in vitro and in vivo. Inactivation of MLL1 leads to decreased expression of genes bound by NHA9 and MLL1 and reverses a gene expression signature found in NUP98-rearranged human leukemias. Our data reveal a molecular dependency on MLL1 function in NUP98-fusion-driven leukemogenesis.


Subject(s)
Chromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Homeodomain Proteins/genetics , Leukemia/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/metabolism , Animals , Cell Proliferation , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Nuclear Pore Complex Proteins/metabolism , Promoter Regions, Genetic , Tumor Cells, Cultured
8.
Mol Cell Proteomics ; 15(11): 3450-3460, 2016 11.
Article in English | MEDLINE | ID: mdl-27634302

ABSTRACT

Polycomb proteins assemble to form complexes with important roles in epigenetic regulation. The Polycomb Repressive Complex 2 (PRC2) modulates the di- and tri-methylation of lysine 27 on histone H3, each of which are associated with gene repression. Although three subunits, EZH1/2, SUZ12, and EED, form the catalytic core of PRC2, a wider group of proteins associate with low stoichiometry. This raises the question of whether dynamic variation of the PRC2 interactome results in alternative forms of the complex during differentiation. Here we compared the physical interactions of PRC2 in undifferentiated and differentiated states of NTERA2 pluripotent embryonic carcinoma cells. Label-free quantitative proteomics was used to assess endogenous immunoprecipitation of the EZH2 and SUZ12 subunits of PRC2. A high stringency data set reflecting the endogenous state of PRC2 was produced that included all previously reported core and associated PRC2 components, and several novel interacting proteins. Comparison of the interactomes obtained in undifferentiated and differentiated cells revealed candidate proteins that were enriched in complexes isolated from one of the two states. For example, SALL4 and ZNF281 associate with PRC2 in pluripotent cells, whereas PCL1 and SMAD3 preferentially associate with PRC2 in differentiating cells. Analysis of the mRNA and protein levels of these factors revealed that their association with PRC2 correlated with their cell state-specific expression. Taken together, we propose that dynamic changes to the PRC2 interactome during differentiation may contribute to directing its activity during cell fate transitions.


Subject(s)
Embryonal Carcinoma Stem Cells/cytology , Pluripotent Stem Cells/cytology , Polycomb Repressive Complex 2/metabolism , Proteomics/methods , Cell Differentiation , Cell Line, Tumor , Embryonal Carcinoma Stem Cells/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Histones/metabolism , Humans , Neoplasm Proteins , Pluripotent Stem Cells/metabolism , Protein Interaction Maps , Transcription Factors
9.
Cancer Cell ; 29(4): 464-476, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-27070701

ABSTRACT

The epigenome is a key determinant of transcriptional output. Perturbations within the epigenome are thought to be a key feature of many, perhaps all cancers, and it is now clear that epigenetic changes are instrumental in cancer development. The inherent reversibility of these changes makes them attractive targets for therapeutic manipulation, and a number of small molecules targeting chromatin-based mechanisms are currently in clinical trials. In this perspective we discuss how understanding the cancer epigenome is providing insights into disease pathogenesis and informing drug development. We also highlight additional opportunities to further unlock the therapeutic potential within the cancer epigenome.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epigenomics , Gene Expression Regulation, Neoplastic , Molecular Targeted Therapy , Neoplasms/genetics , Therapies, Investigational , Animals , Antineoplastic Agents/pharmacokinetics , Chromatin/drug effects , Chromatin/genetics , Chromosome Aberrations , Clinical Trials as Topic , DNA Methylation/drug effects , DNA, Neoplasm/drug effects , DNA, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Histone Code/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Histones/metabolism , Humans , Mice , Models, Genetic , Mutation , Neoplasm Proteins/metabolism , Neoplasms/prevention & control , Neoplasms/therapy , Oncogene Proteins/metabolism , Protein Processing, Post-Translational/drug effects , Transcription, Genetic/drug effects
11.
Genes Dev ; 29(21): 2231-43, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26494712

ABSTRACT

Polycomb-like proteins 1-3 (PCL1-3) are substoichiometric components of the Polycomb-repressive complex 2 (PRC2) that are essential for association of the complex with chromatin. However, it remains unclear why three proteins with such apparent functional redundancy exist in mammals. Here we characterize their divergent roles in both positively and negatively regulating cellular proliferation. We show that while PCL2 and PCL3 are E2F-regulated genes expressed in proliferating cells, PCL1 is a p53 target gene predominantly expressed in quiescent cells. Ectopic expression of any PCL protein recruits PRC2 to repress the INK4A gene; however, only PCL2 and PCL3 confer an INK4A-dependent proliferative advantage. Remarkably, PCL1 has evolved a PRC2- and chromatin-independent function to negatively regulate proliferation. We show that PCL1 binds to and stabilizes p53 to induce cellular quiescence. Moreover, depletion of PCL1 phenocopies the defects in maintaining cellular quiescence associated with p53 loss. This newly evolved function is achieved by the binding of the PCL1 N-terminal PHD domain to the C-terminal domain of p53 through two unique serine residues, which were acquired during recent vertebrate evolution. This study illustrates the functional bifurcation of PCL proteins, which act in both a chromatin-dependent and a chromatin-independent manner to regulate the INK4A and p53 pathways.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Polycomb-Group Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Proliferation/genetics , Cells, Cultured , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/genetics , E2F Transcription Factors/metabolism , Humans , Mice , Polycomb-Group Proteins/genetics , Protein Binding , Protein Stability , Protein Structure, Tertiary/genetics
12.
FEBS J ; 282(18): 3455-73, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26094870

ABSTRACT

The majority of women diagnosed with lymph node-negative breast cancer are unnecessarily treated with damaging chemotherapeutics after surgical resection. This highlights the importance of understanding and more accurately predicting patient prognosis. In the present study, we define the transcriptional networks regulating well-established prognostic gene expression signatures. We find that the same set of transcriptional regulators consistently lie upstream of both 'prognosis' and 'proliferation' gene signatures, suggesting that a central transcriptional network underpins a shared phenotype within these signatures. Strikingly, the master transcriptional regulators within this network predict recurrence risk for lymph node-negative breast cancer better than currently used multigene prognostic assays, particularly in estrogen receptor-positive patients. Simultaneous examination of p16(INK4A) expression, which predicts tumours that have bypassed cellular senescence, revealed that intermediate levels of p16(INK4A) correlate with an intact pRB pathway and improved survival. A combination of these master transcriptional regulators and p16(INK4A), termed the OncoMasTR score, stratifies tumours based on their proliferative and senescence capacity, facilitating a clearer delineation of lymph node-negative breast cancer patients at high risk of recurrence, and thus requiring chemotherapy. Furthermore, OncoMasTR accurately classifies over 60% of patients as 'low risk', an improvement on existing prognostic assays, which has the potential to reduce overtreatment in early-stage patients. Taken together, the present study provides new insights into the transcriptional regulation of cellular proliferation in breast cancer and provides an opportunity to enhance and streamline methods of predicting breast cancer prognosis.


Subject(s)
Breast Neoplasms/genetics , Gene Regulatory Networks , Adult , Aged , Aged, 80 and over , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Proliferation/genetics , Cells, Cultured , Cellular Senescence/genetics , Cohort Studies , Female , Genes, p16 , Humans , Lymphatic Metastasis/genetics , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mice , Middle Aged , Prognosis , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, Estrogen/metabolism , Risk Factors , Tissue Array Analysis
13.
Nat Struct Mol Biol ; 19(12): 1273-81, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23160351

ABSTRACT

Polycomb group proteins are repressive chromatin modifiers with essential roles in metazoan development, cellular differentiation and cell fate maintenance. How Polycomb proteins access active chromatin to confer transcriptional silencing during lineage transitions remains unclear. Here we show that the Polycomb repressive complex 2 (PRC2) component PHF19 binds trimethylated histone H3 Lys36 (H3K36me3), a mark of active chromatin, via its Tudor domain. PHF19 associates with the H3K36me3 demethylase NO66, and it is required to recruit the PRC2 complex and NO66 to stem cell genes during differentiation, leading to PRC2-mediated trimethylation of histone H3 Lys27 (H3K27), loss of H3K36me3 and transcriptional silencing. We propose a model whereby PHF19 functions during mouse embryonic stem cell differentiation to transiently bind the H3K36me3 mark via its Tudor domain, forming essential contact points that allow recruitment of PRC2 and H3K36me3 demethylase activity to active gene loci during their transition to a Polycomb-repressed state.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Histones/metabolism , Polycomb Repressive Complex 2/metabolism , Animals , Embryonic Stem Cells/cytology , Mice
14.
Semin Cell Dev Biol ; 20(7): 835-41, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19682593

ABSTRACT

Mammalian development is a highly complex and tightly regulated process. Transcription factors and chromatin remodelers, acting downstream of cell signalling pathways, are the key intrinsic factors which control gene expression. Recent advances in transcriptomics are allowing biologists to begin to unravel the complex biological roles played by these factors. This review focuses on how genome-wide gene expression and chromatin immunoprecipitation studies are expanding our understanding of the roles played by transcription factors and chromatin remodelers during cell fate decisions in development and differentiation.


Subject(s)
Chromatin Assembly and Disassembly , Gene Expression Profiling/methods , Transcription Factors/genetics , Animals , Cell Differentiation , Chromatin Immunoprecipitation , Gene Expression Regulation, Developmental , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...