Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
Molecules ; 28(24)2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38138487

ABSTRACT

Inhibition of the Embryonic Ectoderm Development (EED) subunit in Polycomb Repressive Complex 2 (PRC2) can inhibit tumor growth. In this paper, we selected six experimentally designed EED competitive Inhibitors of the triazolopyrimidine derivatives class. We investigated the difference in the binding mode of the natural substrate to the Inhibitors and the effects of differences in the parent nuclei, heads, and tails of the Inhibitors on the inhibitory capacity. The results showed that the binding free energy of this class of Inhibitors was close to or lower compared to the natural substrate, providing an energetic basis for competitive inhibition. For the Inhibitors, the presence of a strong negatively charged group at the 6-position of the parent nucleus or the 8'-position of the head would make the hydrogen atom on the head imino group prone to flip, resulting in the vertical movement of the parent nucleus, which significantly decreased the inhibitory ability. When the 6-position of the parent nucleus was a nonpolar group, the parent nucleus would move horizontally, slightly decreasing the inhibitory ability. When the 8'-position of the head was methylene, it formed an intramolecular hydrophobic interaction with the benzene ring on the tail, resulting in a significant increase in inhibition ability.


Subject(s)
Ectoderm , Molecular Dynamics Simulation , Ectoderm/metabolism , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism
2.
Biochem J ; 480(22): 1833-1844, 2023 11 29.
Article in English | MEDLINE | ID: mdl-37888776

ABSTRACT

Polycomb repressive complex 2 (PRC2) is central to polycomb repression as it trimethylates lysine 27 on histone H3 (H3K27me3). How PRC2 is recruited to its targets to deposit H3K27me3 remains an open question. Polycomb-like (PCL) proteins, a group of conserved PRC2 accessory proteins, can direct PRC2 to its targets. In this report, we demonstrate that a PCL protein named PHF1 forms phase-separated condensates at H3K27me3 loci that recruit PRC2. Combining cellular observation and biochemical reconstitution, we show that the N-terminal domains of PHF1 cooperatively mediate target recognition, the chromo-like domain recruits PRC2, and the intrinsically disordered region (IDR) drives phase separation. Moreover, we reveal that the condensates compartmentalize PRC2, DNA, and nucleosome arrays by phase separation. Luciferase reporter assays confirm that PHF1 phase separation promotes transcription repression, further supporting a role of the condensates in polycomb repression. Based on our findings, we propose that these condensates create favorable microenvironments at the target loci for PRC2 to function.


Subject(s)
Histones , Polycomb Repressive Complex 2 , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Histones/genetics , Histones/metabolism , DNA-Binding Proteins/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Nucleosomes
3.
Science ; 381(6664): 1331-1337, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37733873

ABSTRACT

Polycomb repressive complex 2 (PRC2) silences genes through trimethylation of histone H3K27. PRC2 associates with numerous precursor messenger RNAs (pre-mRNAs) and long noncoding RNAs (lncRNAs) with a binding preference for G-quadruplex RNA. In this work, we present a 3.3-Å-resolution cryo-electron microscopy structure of PRC2 bound to a G-quadruplex RNA. Notably, RNA mediates the dimerization of PRC2 by binding both protomers and inducing a protein interface composed of two copies of the catalytic subunit EZH2, thereby blocking nucleosome DNA interaction and histone H3 tail accessibility. Furthermore, an RNA-binding loop of EZH2 facilitates the handoff between RNA and DNA, another activity implicated in PRC2 regulation by RNA. We identified a gain-of-function mutation in this loop that activates PRC2 in zebrafish. Our results reveal mechanisms for RNA-mediated regulation of a chromatin-modifying enzyme.


Subject(s)
G-Quadruplexes , Polycomb Repressive Complex 2 , RNA Precursors , RNA, Long Noncoding , Animals , Cryoelectron Microscopy , Histones/genetics , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/genetics , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/genetics , Zebrafish/genetics , Zebrafish/growth & development , Gain of Function Mutation , Promoter Regions, Genetic , Protein Binding , Enhancer of Zeste Homolog 2 Protein/chemistry , Enhancer of Zeste Homolog 2 Protein/genetics , Crystallography, X-Ray , Protein Conformation , Protein Multimerization
4.
J Mol Biol ; 435(4): 167936, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36610636

ABSTRACT

Polycomb repressive complex 1 (PRC1) and PRC2 are responsible for epigenetic gene regulation. PRC1 ubiquitinates histone H2A (H2Aub), which subsequently promotes PRC2 to introduce the H3 lysine 27 tri-methyl (H3K27me3) repressive chromatin mark. Although this mechanism provides a link between the two key transcriptional repressors, PRC1 and PRC2, it is unknown how histone-tail dynamics contribute to this process. Here, we have examined the effect of H2A ubiquitination and linker-DNA on H3-tail dynamics and H3K27 methylation by PRC2. In naïve nucleosomes, the H3-tail dynamically contacts linker DNA in addition to core DNA, and the linker-DNA is as important for H3K27 methylation as H2A ubiquitination. H2A ubiquitination alters contacts between the H3-tail and DNA to improve the methyltransferase activity of the PRC2-AEBP2-JARID2 complex. Collectively, our data support a model in which H2A ubiquitination by PRC1 synergizes with linker-DNA to hold H3 histone tails poised for their methylation by PRC2-AEBP2-JARID2.


Subject(s)
Histones , Polycomb Repressive Complex 1 , Polycomb Repressive Complex 2 , Ubiquitination , DNA/chemistry , Histones/chemistry , Histones/genetics , Methylation , Polycomb Repressive Complex 1/chemistry , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/genetics
5.
Proc Natl Acad Sci U S A ; 119(22): e2201883119, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35617427

ABSTRACT

Polycomb-group proteins play critical roles in gene silencing through the deposition of histone H3 lysine 27 trimethylation (H3K27me3) and chromatin compaction. This process is essential for embryonic stem cell (ESC) pluripotency, differentiation, and development. Polycomb repressive complex 2 (PRC2) can both read and write H3K27me3, enabling progressive spreading of H3K27me3 on the linear genome. Long-range Polycomb-associated DNA contacts have also been described, but their regulation and role in gene silencing remain unclear. Here, we apply H3K27me3 HiChIP, a protein-directed chromosome conformation method, and optical reconstruction of chromatin architecture to profile long-range Polycomb-associated DNA loops that span tens to hundreds of megabases across multiple topological associated domains in mouse ESCs and human induced pluripotent stem cells. We find that H3K27me3 loop anchors are enriched for Polycomb nucleation points and coincide with key developmental genes. Genetic deletion of H3K27me3 loop anchors results in disruption of spatial contact between distant loci and altered H3K27me3 in cis, both locally and megabases away on the same chromosome. In mouse embryos, loop anchor deletion leads to ectopic activation of the partner gene, suggesting that Polycomb-associated loops control gene silencing during development. Further, we find that alterations in PRC2 occupancy resulting from an RNA binding­deficient EZH2 mutant are accompanied by loss of Polycomb-associated DNA looping. Together, these results suggest PRC2 uses RNA binding to enhance long-range chromosome folding and H3K27me3 spreading. Developmental gene loci have unique roles in Polycomb spreading, emerging as important architectural elements of the epigenome.


Subject(s)
Chromosomes , Gene Expression Regulation, Developmental , Gene Silencing , Histones , Polycomb Repressive Complex 2 , Animals , Chromatin Immunoprecipitation/methods , Chromosomes/chemistry , Chromosomes/metabolism , Embryo, Mammalian , Enhancer of Zeste Homolog 2 Protein/genetics , Histones/genetics , Histones/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Lysine/metabolism , Methylation , Mice , Nucleic Acid Conformation , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism
6.
Future Med Chem ; 14(9): 609-621, 2022 05.
Article in English | MEDLINE | ID: mdl-35348013

ABSTRACT

Background: The inhibition of PRC2, implicated in the pathogenesis of several tumors, can be a useful therapeutic strategy for cancer treatment. In the literature, two types of PRC2 modulators are reported: competitive inhibitors of S-adenosyl methionine binding to the catalytic subunit EZH2; and allosteric ligands that prevent the interaction of the trimethylated H3K27 lysine in histone 3 to the EED subunit. The lack of dual EZH2/EED modulators drove us to search for compounds capable of recognizing both domains. Materials & methods: This goal was pursued by combining pharmacophore- and docking-based virtual screening of the Multi-Target Ligand Chemotheca database. Prediction tools for absorption, distribution, metabolism and excretion and pan-assay interference compounds were also applied. Results: Finally, five 1,2,3-triazole derivatives were identified as promising dual EZH2/EED modulators. Conclusion: Our multistage screening protocol highlighted the great potential of Chemotheca for identifying polypharmacological agents.


Subject(s)
Neoplasms , Polycomb Repressive Complex 2 , Catalytic Domain , Histones/metabolism , Ligands , Neoplasms/drug therapy , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism
7.
J Med Chem ; 64(23): 17146-17183, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34807608

ABSTRACT

Aberrant activity of the histone methyltransferase polycomb repressive complex 2 (PRC2) has been linked to several cancers, with small-molecule inhibitors of the catalytic subunit of the PRC2 enhancer of zeste homologue 2 (EZH2) being recently approved for the treatment of epithelioid sarcoma (ES) and follicular lymphoma (FL). Compounds binding to the EED subunit of PRC2 have recently emerged as allosteric inhibitors of PRC2 methyltransferase activity. In contrast to orthosteric inhibitors that target EZH2, small molecules that bind to EED retain their efficacy in EZH2 inhibitor-resistant cell lines. In this paper we disclose the discovery of potent and orally bioavailable EED ligands with good solubilities. The solubility of the EED ligands was optimized through a variety of design tactics, with the resulting compounds exhibiting in vivo efficacy in EZH2-driven tumors.


Subject(s)
Enzyme Inhibitors/pharmacology , Polycomb Repressive Complex 2/antagonists & inhibitors , Allosteric Regulation , Animals , Catalytic Domain , Cell Line , Cell Proliferation/drug effects , Enhancer of Zeste Homolog 2 Protein/chemistry , Enhancer of Zeste Homolog 2 Protein/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Heterocyclic Compounds/chemistry , Humans , Ligands , Polycomb Repressive Complex 2/chemistry , Rats , Structure-Activity Relationship
8.
Biochem Soc Trans ; 49(6): 2639-2653, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34747969

ABSTRACT

Polycomb repressive complexes are a family of chromatin modifier enzymes which are critical for regulating gene expression and maintaining cell-type identity. The reversible chemical modifications of histone H3 and H2A by the Polycomb proteins are central to its ability to function as a gene silencer. PRC2 is both a reader and writer of the tri-methylation of histone H3 lysine 27 (H3K27me3) which serves as a marker for transcription repression, and heterochromatin boundaries. Over the last few years, several studies have provided key insights into the mechanisms regulating the recruitment and activation of PRC2 at Polycomb target genes. In this review, we highlight the recent structural studies which have elucidated the roles played by Polycomb cofactor proteins in mediating crosstalk between histone post-translational modifications and the recruitment of PRC2 and the stimulation of PRC2 methyltransferase activity.


Subject(s)
Chromatin/metabolism , Polycomb Repressive Complex 2/metabolism , Animals , Humans , Polycomb Repressive Complex 2/chemistry , Protein Binding , Protein Conformation , Protein Processing, Post-Translational , Structure-Activity Relationship
9.
Curr Top Med Chem ; 21(31): 2771-2777, 2021.
Article in English | MEDLINE | ID: mdl-34544341

ABSTRACT

The polycomb repressive complex 2 (PRC2) can methylate at lysine 27 of histone H3 at the trimethylation level (H3K27me3). This leads to gene silencing and is known to be dysregulated in many cancers. PRC2 is made up of three core subunits: EZH2, SUZ12, and EED. EED is essential for the regulation of PRC2 function by binding to H3K27me3. Targeting the allosteric site within EED offers new strategies to disrupt the PRC2 activity. In this minireview, we summarize some of the recent developments in small molecules that target EED and its interaction with other core proteins in the PRC2 complex.


Subject(s)
Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/metabolism , Polycomb Repressive Complex 2/antagonists & inhibitors , Allosteric Site , Animals , Histones/chemistry , Histones/metabolism , Humans , Methylation , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism
10.
Mol Cell Proteomics ; 20: 100139, 2021.
Article in English | MEDLINE | ID: mdl-34418567

ABSTRACT

Proteomics methodology has expanded to include protein structural analysis, primarily through cross-linking mass spectrometry (XL-MS) and hydrogen-deuterium exchange mass spectrometry (HX-MS). However, while the structural proteomics community has effective tools for primary data analysis, there is a need for structure modeling pipelines that are accessible to the proteomics specialist. Integrative structural biology requires the aggregation of multiple distinct types of data to generate models that satisfy all inputs. Here, we describe IMProv, an app in the Mass Spec Studio that combines XL-MS data with other structural data, such as cryo-EM densities and crystallographic structures, for integrative structure modeling on high-performance computing platforms. The resource provides an easily deployed bundle that includes the open-source Integrative Modeling Platform program (IMP) and its dependencies. IMProv also provides functionality to adjust cross-link distance restraints according to the underlying dynamics of cross-linked sites, as characterized by HX-MS. A dynamics-driven conditioning of restraint values can improve structure modeling precision, as illustrated by an integrative structure of the five-membered Polycomb Repressive Complex 2. IMProv is extensible to additional types of data.


Subject(s)
Models, Molecular , Proteomics/methods , Software , Mass Spectrometry , Polycomb Repressive Complex 2/chemistry , Protein Conformation
11.
Nat Rev Mol Cell Biol ; 22(12): 815-833, 2021 12.
Article in English | MEDLINE | ID: mdl-34400841

ABSTRACT

Precise control of gene expression is fundamental to cell function and development. Although ultimately gene expression relies on DNA-binding transcription factors to guide the activity of the transcription machinery to genes, it has also become clear that chromatin and histone post-translational modification have fundamental roles in gene regulation. Polycomb repressive complexes represent a paradigm of chromatin-based gene regulation in animals. The Polycomb repressive system comprises two central protein complexes, Polycomb repressive complex 1 (PRC1) and PRC2, which are essential for normal gene regulation and development. Our early understanding of Polycomb function relied on studies in simple model organisms, but more recently it has become apparent that this system has expanded and diverged in mammals. Detailed studies are now uncovering the molecular mechanisms that enable mammalian PRC1 and PRC2 to identify their target sites in the genome, communicate through feedback mechanisms to create Polycomb chromatin domains and control transcription to regulate gene expression. In this Review, we discuss and contextualize the emerging principles that define how this fascinating chromatin-based system regulates gene expression in mammals.


Subject(s)
Gene Expression Regulation/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/metabolism , Chromatin/chemistry , Chromatin/metabolism , Histones/metabolism , Humans , Methylation , Polycomb Repressive Complex 1/chemistry , Polycomb Repressive Complex 2/chemistry , Protein Processing, Post-Translational , Transcription, Genetic , Ubiquitination
12.
Nat Commun ; 12(1): 4592, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34321472

ABSTRACT

The polycomb repressive complex 2 (PRC2) is a histone methyltransferase that maintains cell identities. JARID2 is the only accessory subunit of PRC2 that known to trigger an allosteric activation of methyltransferase. Yet, this mechanism cannot be generalised to all PRC2 variants as, in vertebrates, JARID2 is mutually exclusive with most of the accessory subunits of PRC2. Here we provide functional and structural evidence that the vertebrate-specific PRC2 accessory subunit PALI1 emerged through a convergent evolution to mimic JARID2 at the molecular level. Mechanistically, PRC2 methylates PALI1 K1241, which then binds to the PRC2-regulatory subunit EED to allosterically activate PRC2. PALI1 K1241 is methylated in mouse and human cell lines and is essential for PALI1-induced allosteric activation of PRC2. High-resolution crystal structures revealed that PALI1 mimics the regulatory interactions formed between JARID2 and EED. Independently, PALI1 also facilitates DNA and nucleosome binding by PRC2. In acute myelogenous leukemia cells, overexpression of PALI1 leads to cell differentiation, with the phenotype altered by a separation-of-function PALI1 mutation, defective in allosteric activation and active in DNA binding. Collectively, we show that PALI1 facilitates catalysis and substrate binding by PRC2 and provide evidence that subunit-induced allosteric activation is a general property of holo-PRC2 complexes.


Subject(s)
Allosteric Regulation/physiology , DNA/metabolism , Nucleosomes/metabolism , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Animals , Catalysis , Cell Differentiation , Cell Line , Histones/metabolism , Humans , Mice , Polycomb Repressive Complex 2/genetics , Protein Binding
13.
Theranostics ; 11(14): 6873-6890, 2021.
Article in English | MEDLINE | ID: mdl-34093859

ABSTRACT

Rationale: Chemoresistance is a major obstacle in prostate cancer (PCa) treatment. We sought to understand the underlying mechanism of PCa chemoresistance and discover new treatments to overcome docetaxel resistance. Methods: We developed a novel phenotypic screening platform for the discovery of specific inhibitors of chemoresistant PCa cells. The mechanism of action of the lead compound was investigated using computational, molecular and cellular approaches. The in vivo toxicity and efficacy of the lead compound were evaluated in clinically-relevant animal models. Results: We identified LG1980 as a lead compound that demonstrates high selectivity and potency against chemoresistant PCa cells. Mechanistically, LG1980 binds embryonic ectoderm development (EED), disrupts the interaction between EED and enhancer of zeste homolog 2 (EZH2), thereby inducing the protein degradation of EZH2 and inhibiting the phosphorylation and activity of EZH2. Consequently, LG1980 targets a survival signaling cascade consisting of signal transducer and activator of transcription 3 (Stat3), S-phase kinase-associated protein 2 (SKP2), ATP binding cassette B 1 (ABCB1) and survivin. As a lead compound, LG1980 is well tolerated in mice and effectively suppresses the in vivo growth of chemoresistant PCa and synergistically enhances the efficacy of docetaxel in xenograft models. Conclusions: These results indicate that pharmacological inhibition of EED-EZH2 interaction is a novel strategy for the treatment of chemoresistant PCa. LG1980 and its analogues have the potential to be integrated into standard of care to improve clinical outcomes in PCa patients.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Drug Discovery/methods , Drug Resistance, Neoplasm , Enhancer of Zeste Homolog 2 Protein/metabolism , Polycomb Repressive Complex 2/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Line, Tumor , Cell Survival/drug effects , Docetaxel/pharmacology , Docetaxel/therapeutic use , Drug Synergism , Humans , Inhibitory Concentration 50 , Male , Mice , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , S-Phase Kinase-Associated Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Survivin/metabolism , Xenograft Model Antitumor Assays
14.
Nucleic Acids Res ; 49(11): 6238-6248, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34057467

ABSTRACT

Polycomb repressive complex 2 (PRC2) is an essential protein complex that silences gene expression via post-translational modifications of chromatin. This paper combined homology modeling, atomistic and coarse-grained molecular dynamics simulations, and single-molecule force spectroscopy experiments to characterize both its full-length structure and PRC2-DNA interactions. Using free energy calculations with a newly parameterized protein-DNA force field, we studied a total of three potential PRC2 conformations and their impact on DNA binding and bending. Consistent with cryo-EM studies, we found that EZH2, a core subunit of PRC2, provides the primary interface for DNA binding, and its curved surface can induce DNA bending. Our simulations also predicted the C2 domain of the SUZ12 subunit to contact DNA. Multiple PRC2 complexes bind with DNA cooperatively via allosteric communication through the DNA, leading to a hairpin-like looped configuration. Single-molecule experiments support PRC2-mediated DNA looping and the role of AEBP2 in regulating such loop formation. The impact of AEBP2 can be partly understood from its association with the C2 domain, blocking C2 from DNA binding. Our study suggests that accessory proteins may regulate the genomic location of PRC2 by interfering with its DNA interactions.


Subject(s)
DNA/chemistry , Polycomb Repressive Complex 2/chemistry , DNA/metabolism , Molecular Dynamics Simulation , Nucleic Acid Conformation , Polycomb Repressive Complex 2/metabolism , Protein Binding , Protein Conformation
15.
Chem Biol Interact ; 344: 109530, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34029540

ABSTRACT

PURPOSE: Targeting enhancer of zeste homolog 2 (EZH2) can represent a hopeful strategy for oncotherapy. Also, the use of PLGA-based nanoparticles as a novel and rate-controlling carrier system was of our concern. METHODS: Benzimidazole derivatives were synthesized, and their structures were clarified. In vitro antitumor activity was evaluated. Then, a modeling study was performed to investigate the ability of the most active compounds to recognize EZH2 active sites. Compound 30 (Drug) was selected to conduct pre-formulation studies and then it was incorporated into polymeric PLGA nanoparticles (NPs). NPs were then fully characterized to select an optimized formula (NP4) that subjected to further evaluation regarding antitumor activity and protein expression levels of EZH2 and EpCAM. RESULTS: The results showed the antitumor activity of some synthesized derivatives. Docking outcomes demonstrated that Compound 30 was able to identify EZH2 active sites. NP4 exhibited promising findings and proved to keep the antitumor activity of Compound 30. HEPG-2 was the most sensitive for both Drug and NP4. Protein analysis indicated that Drug and NP4 had targeted EZH2 and the downstream signaling pathway leading to the decline of EpCAM expression. CONCLUSIONS: Targeting EZH2 by Compound 30 has potential use in the treatment of cancer especially hepatocellular carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Drug Carriers/chemistry , Enhancer of Zeste Homolog 2 Protein/metabolism , Enzyme Inhibitors/pharmacology , Nanoparticles/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Benzimidazoles/chemical synthesis , Benzimidazoles/metabolism , Binding Sites , Cell Line, Tumor , Drug Liberation , Drug Screening Assays, Antitumor , Enhancer of Zeste Homolog 2 Protein/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Epithelial Cell Adhesion Molecule/metabolism , Humans , Molecular Docking Simulation , Molecular Structure , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Protein Binding , Solubility , Structure-Activity Relationship
16.
Trends Genet ; 37(6): 547-565, 2021 06.
Article in English | MEDLINE | ID: mdl-33494958

ABSTRACT

Modulation of chromatin structure and/or modification by Polycomb repressive complexes (PRCs) provides an important means to partition the genome into functionally distinct subdomains and to regulate the activity of the underlying genes. Both the enzymatic activity of PRC2 and its chromatin recruitment, spreading, and eviction are exquisitely regulated via interactions with cofactors and DNA elements (such as unmethylated CpG islands), histones, RNA (nascent mRNA and long noncoding RNA), and R-loops. PRC2-catalyzed histone H3 lysine 27 trimethylation (H3K27me3) is recognized by distinct classes of effectors such as canonical PRC1 and BAH module-containing proteins (notably BAHCC1 in human). These effectors mediate gene silencing by different mechanisms including phase separation-related chromatin compaction and histone deacetylation. We discuss recent advances in understanding the structural architecture of PRC2, the regulation of its activity and chromatin recruitment, and the molecular mechanisms underlying Polycomb-mediated gene silencing. Because PRC deregulation is intimately associated with the development of diseases, a better appreciation of Polycomb-based (epi)genomic regulation will have far-reaching implications in biology and medicine.


Subject(s)
Chromatin/genetics , Gene Silencing/physiology , Histones/metabolism , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/genetics , Alkynes , Animals , Chromatin/chemistry , Chromatin/metabolism , Histones/genetics , Humans , Lysine/metabolism , Polycomb Repressive Complex 2/metabolism , RNA, Long Noncoding/metabolism , Vertebrates
17.
J Mol Biol ; 433(6): 166827, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33460684

ABSTRACT

The regulation of chromatin biology ultimately depends on the manipulation of its smallest subunit, the nucleosome. The proteins that bind and operate on the nucleosome do so, while their substrate is part of a polymer embedded in the dense nuclear environment. Their molecular interactions must in some way be tuned to deal with this complexity. Due to the rapid increase in the number of high-resolution structures of nucleosome-protein complexes and the increasing understanding of the cellular chromatin structure, it is starting to become clearer how chromatin factors operate in this complex environment. In this review, we analyze the current literature on the interplay between nucleosome-protein interactions and higher-order chromatin structure. We examine in what way nucleosomes-protein interactions can affect and can be affected by chromatin organization at the oligonucleosomal level. In addition, we review the characteristics of nucleosome-protein interactions that can cause phase separation of chromatin. Throughout, we hope to illustrate the exciting challenges in characterizing nucleosome-protein interactions beyond the nucleosome.


Subject(s)
Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/chemistry , DNA/chemistry , Histones/chemistry , Nucleosomes/ultrastructure , Polycomb Repressive Complex 2/chemistry , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA/genetics , DNA/metabolism , Histones/genetics , Histones/metabolism , Humans , Molecular Dynamics Simulation , Nucleic Acid Conformation , Nucleosomes/chemistry , Nucleosomes/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Static Electricity
18.
Science ; 371(6527)2021 01 22.
Article in English | MEDLINE | ID: mdl-33479123

ABSTRACT

Polycomb repressive complexes 1 and 2 (PRC1 and PRC2) cooperate to determine cell identity by epigenetic gene expression regulation. However, the mechanism of PRC2 recruitment by means of recognition of PRC1-mediated H2AK119ub1 remains poorly understood. Our PRC2 cryo-electron microscopy structure with cofactors JARID2 and AEBP2 bound to a H2AK119ub1-containing nucleosome reveals a bridge helix in EZH2 that connects the SET domain, H3 tail, and nucleosomal DNA. JARID2 and AEBP2 each interact with one ubiquitin and the H2A-H2B surface. JARID2 stimulates PRC2 through interactions with both the polycomb protein EED and the H2AK119-ubiquitin, whereas AEBP2 has an additional scaffolding role. The presence of these cofactors partially overcomes the inhibitory effect that H3K4me3 and H3K36me3 exert on core PRC2 (in the absence of cofactors). Our results support a key role for JARID2 and AEBP2 in the cross-talk between histone modifications and PRC2 activity.


Subject(s)
Histone Code , Polycomb Repressive Complex 2/metabolism , Repressor Proteins/metabolism , Animals , Cryoelectron Microscopy , Gene Expression Regulation , Histones/metabolism , Humans , Nucleosomes/metabolism , PR-SET Domains , Polycomb Repressive Complex 2/chemistry , Ubiquitin/metabolism , Xenopus
19.
Subcell Biochem ; 96: 519-562, 2021.
Article in English | MEDLINE | ID: mdl-33252743

ABSTRACT

Polycomb Repressive Complex 2 (PRC2) is a major repressive chromatin complex formed by the Polycomb Group (PcG) proteins. PRC2 mediates trimethylation of histone H3 lysine 27 (H3K27me3), a hallmark of gene silencing. PRC2 is a key regulator of development, impacting many fundamental biological processes, like stem cell differentiation in mammals and vernalization in plants. Misregulation of PRC2 function is linked to a variety of human cancers and developmental disorders. In correlation with its diverse roles in development, PRC2 displays a high degree of compositional complexity and plasticity. Structural biology research over the past decade has shed light on the molecular mechanisms of the assembly, catalysis, allosteric activation, autoinhibition, chemical inhibition, dimerization and chromatin targeting of various developmentally regulated PRC2 complexes. In addition to these aspects, structure-function analysis is also discussed in connection with disease data in this chapter.


Subject(s)
Chromatin/genetics , Gene Silencing , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Animals , Chromatin/metabolism , Histones/metabolism , Humans , Methylation
20.
Mol Inform ; 40(2): e2000148, 2021 02.
Article in English | MEDLINE | ID: mdl-32833314

ABSTRACT

The Polycomb Repressive complex 2 (PRC2) maintains a repressive chromatin state and silences many genes, acting as methylase on histone tails. This enzyme was found overexpressed in many types of cancer. In this work, we have set up a Computer-Aided Drug Design approach based on the allosteric modulation of PRC2. In order to minimize the possible bias derived from using a single set of coordinates within the protein-ligand complex, a dynamic workflow was developed. In details, molecular dynamic was used as tool to identify the most significant ligand-protein interactions from several crystallized protein structures. The identified features were used for the creation of dynamic pharmacophore models and docking grid constraints for the design of new PRC2 allosteric modulators. Our protocol was retrospectively validated using a dataset of active and inactive compounds, and the results were compared to the classic approaches, through ROC curves and enrichment factor. Our approach suggested some important interaction features to be adopted for virtual screening performance improvement.


Subject(s)
Allosteric Site , Binding Sites , Drug Design , Molecular Docking Simulation , Molecular Dynamics Simulation , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/chemistry , Humans , Ligands , Protein Binding , ROC Curve
SELECTION OF CITATIONS
SEARCH DETAIL
...