Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Chembiochem ; 24(21): e202300490, 2023 11 02.
Article En | MEDLINE | ID: mdl-37581408

Nτ -methylation of His73 in actin by histidine methyltransferase SETD3 plays an important role in stabilising actin filaments in eukaryotes. Mutations in actin and overexpression of SETD3 have been related to human diseases, including cancer. Here, we investigated the importance of Trp79 in ß-actin on productive human SETD3 catalysis. Substitution of Trp79 in ß-actin peptides by its chemically diverse analogues reveals that the hydrophobic Trp79 binding pocket modulates the catalytic activity of SETD3, and that retaining a bulky and hydrophobic amino acid at position 79 is important for efficient His73 methylation by SETD3. Molecular dynamics simulations show that the Trp79 binding pocket of SETD3 is ideally shaped to accommodate large and hydrophobic Trp79, contributing to the favourable release of water molecules upon binding. Our results demonstrate that the distant Trp79 binding site plays an important role in efficient SETD3 catalysis, contributing to the identification of new SETD3 substrates and the development of chemical probes targeting the biomedically important SETD3.


Actins , Methyltransferases , Humans , Methyltransferases/metabolism , Actins/chemistry , Histone Methyltransferases/chemistry , Histone Methyltransferases/genetics , Histone Methyltransferases/metabolism , Histidine/chemistry , Methylation , Catalysis
2.
Protein Sci ; 31(5): e4305, 2022 05.
Article En | MEDLINE | ID: mdl-35481649

Actin histidine Nτ -methylation by histidine methyltransferase SETD3 plays an important role in human biology and diseases. Here, we report integrated synthetic, biocatalytic, biostructural, and computational analyses on human SETD3-catalyzed methylation of actin peptides possessing histidine and its structurally and chemically diverse mimics. Our enzyme assays supported by biostructural analyses demonstrate that SETD3 has a broader substrate scope beyond histidine, including N-nucleophiles on the aromatic and aliphatic side chains. Quantum mechanical/molecular mechanical molecular dynamics and free-energy simulations provide insight into binding geometries and the free energy barrier for the enzymatic methyl transfer to histidine mimics, further supporting experimental data that histidine is the superior SETD3 substrate over its analogs. This work demonstrates that human SETD3 has a potential to catalyze efficient methylation of several histidine mimics, overall providing mechanistic, biocatalytic, and functional insight into actin histidine methylation by SETD3.


Actins , Methyltransferases , Actins/chemistry , Actins/metabolism , Histidine/chemistry , Histone Methyltransferases/chemistry , Histone Methyltransferases/metabolism , Humans , Methylation , Methyltransferases/metabolism
3.
Interdiscip Sci ; 14(4): 929-936, 2022 Dec.
Article En | MEDLINE | ID: mdl-35419695

The SETD3 enzyme has been identified as the methyltransferase for the His73 methylation in ß-actin, and such methylation plays an important role in regulating the actin's biochemical properties and fine-tuning the protein's cellular roles. Further studies have demonstrated that SETD3 may be able to methylase some other residues, including lysine and methionine, that substitute His73 in the ß-actin peptide. The activity of SETD3 on the Met73 peptide is low without turnover. Interestingly, it has been shown that the N255V and N255A mutations of SETD3 can increase the activity by about 3-fold for the methionine methylation, while such mutations lead to a significant reduction of kcat for the His73 methylation. The detailed mechanism that leads to such increase of the activity for the Met73 methylation as a result of the mutations has not been understood. In this work, QM/MM molecular dynamics (MD) and potential of mean force (PMF) free energy simulations are undertaken for investigating structural, dynamic, and energetic properties involving the complex of SETD3 and Met73 peptide and to study the SETD3-catalyzed methionine methylation and the effects of the N255V mutation. It is demonstrated that the free energy barrier in the case of the methionine methylation in SETD3 is about 10 kcal/mol higher than that for the histidine methylation. Moreover, the free energy barrier for the methionine methylation in the N255V mutant is about 1 kcal/mol lower than that in the wild-type enzyme. These results agree with previous experimental observation. The origin of the free-energy barrier changes as a result of the H to M substitution on the ß-actin peptide and the N255V mutation of SETD3 is discussed based on the data obtained from the simulations.


Actins , Lysine , Methylation , Histone Methyltransferases/chemistry , Histone Methyltransferases/genetics , Histone Methyltransferases/metabolism , Actins/chemistry , Actins/genetics , Actins/metabolism , Histidine/chemistry , Histidine/metabolism , Methionine , Methyltransferases/metabolism , Racemethionine , Peptides , Catalysis
4.
mBio ; 12(6): e0135221, 2021 12 21.
Article En | MEDLINE | ID: mdl-34749530

The parasite Trypanosoma brucei periodically changes the expression of protective variant surface glycoproteins (VSGs) to evade its host's immune system in a process known as antigenic variation. One route to change VSG expression is the transcriptional activation of a previously silent VSG expression site (ES), a subtelomeric region containing the VSG genes. Homologous recombination of a different VSG from a large reservoir into the active ES represents another route. The conserved histone methyltransferase DOT1B is involved in transcriptional silencing of inactive ES and influences ES switching kinetics. The molecular machinery that enables DOT1B to execute these regulatory functions remains elusive, however. To better understand DOT1B-mediated regulatory processes, we purified DOT1B-associated proteins using complementary biochemical approaches. We identified several novel DOT1B interactors. One of these was the RNase H2 complex, previously shown to resolve RNA-DNA hybrids, maintain genome integrity, and play a role in antigenic variation. Our study revealed that DOT1B depletion results in an increase in RNA-DNA hybrids, accumulation of DNA damage, and ES switching events. Surprisingly, a similar pattern of VSG deregulation was observed in RNase H2 mutants. We propose that both proteins act together in resolving R-loops to ensure genome integrity and contribute to the tightly regulated process of antigenic variation. IMPORTANCE Trypanosoma brucei is a unicellular parasite that causes devastating diseases like sleeping sickness in humans and the "nagana" disease in cattle in Africa. Fundamental to the establishment and prolongation of a trypanosome infection is the parasite's ability to escape the mammalian host's immune system by antigenic variation, which relies on periodic changes of a protein surface coat. The exact mechanisms, however, which mediate these changes are still elusive. In this work, we describe a novel protein complex consisting of the histone methyltransferase DOT1B and RNase H2 which is involved in antigenic variation.


Histone Methyltransferases/metabolism , Protozoan Proteins/metabolism , Ribonuclease H/metabolism , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism , Trypanosomiasis/parasitology , Antigenic Variation , Genome, Protozoan , Genomic Instability , Histone Methyltransferases/chemistry , Histone Methyltransferases/genetics , Humans , Protein Binding , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , R-Loop Structures , Ribonuclease H/chemistry , Ribonuclease H/genetics , Trypanosoma brucei brucei/chemistry , Trypanosoma brucei brucei/enzymology
5.
BMC Plant Biol ; 21(1): 373, 2021 Aug 13.
Article En | MEDLINE | ID: mdl-34388969

BACKGROUND: Flowering is an important inflection point in the transformation from vegetative to reproductive growth, and premature bolting severely decreases crop yield and quality. RESULTS: In this study, a stable early-bolting mutant, ebm3, was identified in an ethyl methanesulfonate (EMS)-mutagenized population of a Chinese cabbage doubled haploid (DH) line 'FT'. Compared with 'FT', ebm3 showed early bolting under natural cultivation in autumn, and curled leaves. Genetic analysis showed that the early-bolting phenotype was controlled by a single recessive nuclear gene. Modified MutMap sequencing, genotyping analyses and allelism test provide strong evidence that BrEBM3 (BraA04g017190.3 C), encoding the histone methyltransferase CURLY LEAF (CLF), was the strongly candidate gene of the emb3. A C to T base substitution in the 14th exon of BrEBM3 resulted in an amino acid change (S to F) and the early-bolting phenotype of emb3. The mutation occurred in the SET domain (Suppressor of protein-effect variegation 3-9, Enhancer-of-zeste, Trithorax), which catalyzes site- and state-specific lysine methylation in histones. Tissue-specific expression analysis showed that BrEBM3 was highly expressed in the flower and bud. Promoter activity assay confirmed that BrEBM3 promoter was active in inflorescences. Subcellular localization analysis revealed that BrEBM3 localized in the nucleus. Transcriptomic studies supported that BrEBM3 mutation might repress H3K27me3 deposition and activate expression of the AGAMOUS (AG) and AGAMOUS-like (AGL) loci, resulting in early flowering. CONCLUSIONS: Our study revealed that an EMS-induced early-bolting mutant ebm3 in Chinese cabbage was caused by a nonsynonymous mutation in BraA04g017190.3 C, encoding the histone methyltransferase CLF. These results improve our knowledge of the genetic and genomic resources of bolting and flowering, and may be beneficial to the genetic improvement of Chinese cabbage.


Amino Acid Substitution , Brassica rapa/enzymology , Histone Methyltransferases/metabolism , Plant Proteins/metabolism , Amino Acids/metabolism , Brassica rapa/genetics , Brassica rapa/growth & development , Flowers/enzymology , Flowers/genetics , Flowers/growth & development , Histone Methyltransferases/chemistry , Histone Methyltransferases/genetics , Mutation , Plant Proteins/genetics , Transcriptome
6.
J Biol Chem ; 295(32): 10901-10910, 2020 08 07.
Article En | MEDLINE | ID: mdl-32503840

Most characterized protein methylation events encompass arginine and lysine N-methylation, and only a few cases of protein methionine thiomethylation have been reported. Newly discovered oncohistone mutations include lysine-to-methionine substitutions at positions 27 and 36 of histone H3.3. In these instances, the methionine substitution localizes to the active-site pocket of the corresponding histone lysine methyltransferase, thereby inhibiting the respective transmethylation activity. SET domain-containing 3 (SETD3) is a protein (i.e. actin) histidine methyltransferase. Here, we generated an actin variant in which the histidine target of SETD3 was substituted with methionine. As for previously characterized histone SET domain proteins, the methionine substitution substantially (76-fold) increased binding affinity for SETD3 and inhibited SETD3 activity on histidine. Unexpectedly, SETD3 was active on the substituted methionine, generating S-methylmethionine in the context of actin peptide. The ternary structure of SETD3 in complex with the methionine-containing actin peptide at 1.9 Å resolution revealed that the hydrophobic thioether side chain is packed by the aromatic rings of Tyr312 and Trp273, as well as the hydrocarbon side chain of Ile310 Our results suggest that placing methionine properly in the active site-within close proximity to and in line with the incoming methyl group of SAM-would allow some SET domain proteins to selectively methylate methionine in proteins.


Histone Methyltransferases/metabolism , Methionine/metabolism , Histone Methyltransferases/chemistry , Humans , Methylation , Protein Binding , Protein Processing, Post-Translational , Protein Structure, Tertiary
7.
Nucleic Acids Res ; 48(4): 1652-1668, 2020 02 28.
Article En | MEDLINE | ID: mdl-31930303

The excision of mutagenic DNA adducts by the nucleotide excision repair (NER) pathway is essential for genome stability, which is key to avoiding genetic diseases, premature aging, cancer and neurologic disorders. Due to the need to process an extraordinarily high damage density embedded in the nucleosome landscape of chromatin, NER activity provides a unique functional caliper to understand how histone modifiers modulate DNA damage responses. At least three distinct lysine methyltransferases (KMTs) targeting histones have been shown to facilitate the detection of ultraviolet (UV) light-induced DNA lesions in the difficult to access DNA wrapped around histones in nucleosomes. By methylating core histones, these KMTs generate docking sites for DNA damage recognition factors before the chromatin structure is ultimately relaxed and the offending lesions are effectively excised. In view of their function in priming nucleosomes for DNA repair, mutations of genes coding for these KMTs are expected to cause the accumulation of DNA damage promoting cancer and other chronic diseases. Research on the question of how KMTs modulate DNA repair might pave the way to the development of pharmacologic agents for novel therapeutic strategies.


Chromatin/genetics , DNA Damage/genetics , Histone Methyltransferases/genetics , Histones/genetics , Chromatin/radiation effects , DNA Damage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , Genomic Instability/genetics , Genomic Instability/radiation effects , Histone Methyltransferases/chemistry , Methylation/radiation effects , Nucleosomes/genetics , Nucleosomes/radiation effects , Saccharomyces cerevisiae/genetics , Signal Transduction/radiation effects , Ultraviolet Rays
8.
J Biol Chem ; 295(9): 2582-2589, 2020 02 28.
Article En | MEDLINE | ID: mdl-31911441

Most characterized SET domain (SETD) proteins are protein lysine methyltransferases, but SETD3 was recently demonstrated to be a protein (i.e. actin) histidine-N3 methyltransferase. Human SETD3 shares a high structural homology with two known protein lysine methyltransferases-human SETD6 and the plant LSMT-but differs in the residues constituting the active site. In the SETD3 active site, Asn255 engages in a unique hydrogen-bonding interaction with the target histidine of actin that likely contributes to its >1300-fold greater catalytic efficiency (kcat/Km ) on histidine than on lysine. Here, we engineered active-site variants to switch the SETD3 target specificity from histidine to lysine. Substitution of Asn255 with phenylalanine (N255F), together with substitution of Trp273 with alanine (W273A), generated an active site mimicking that of known lysine methyltransferases. The doubly substituted SETD3 variant exhibited a 13-fold preference for lysine over histidine. We show, by means of X-ray crystallography, that the two target nitrogen atoms-the N3 atom of histidine and the terminal ϵ-amino nitrogen of lysine-occupy the same position and point toward and are within a short distance of the incoming methyl group of SAM for a direct methyl transfer during catalysis. In contrast, SETD3 and its Asn255 substituted derivatives did not methylate glutamine (another potentially methylated amino acid). However, the glutamine-containing peptide competed with the substrate peptide, and glutamine bound in the active site, but too far away from SAM to be methylated. Our results provide insight into the structural parameters defining the target amino acid specificity of SET enzymes.


Histone Methyltransferases/genetics , Histone-Lysine N-Methyltransferase/metabolism , Lysine/metabolism , Actins/metabolism , Amino Acid Substitution , Catalytic Domain , Histidine/metabolism , Histone Methyltransferases/chemistry , Histone Methyltransferases/metabolism , Histone-Lysine N-Methyltransferase/genetics , Humans , Hydrogen Bonding , Methylation , Protein Engineering , Substrate Specificity/genetics
9.
Mol Cell ; 76(5): 712-723.e4, 2019 12 05.
Article En | MEDLINE | ID: mdl-31733991

The COMPASS (complex of proteins associated with Set1) complex represents the prototype of the SET1/MLL family of methyltransferases that controls gene transcription by H3K4 methylation (H3K4me). Although H2B monoubiquitination (H2Bub) is well known as a prerequisite histone mark for COMPASS activity, how H2Bub activates COMPASS remains unclear. Here, we report the cryoelectron microscopy (cryo-EM) structures of an extended COMPASS catalytic module (CM) bound to the H2Bub and free nucleosome. The COMPASS CM clamps onto the nucleosome disk-face via an extensive interface to capture the flexible H3 N-terminal tail. The interface also sandwiches a critical Set1 arginine-rich motif (ARM) that autoinhibits COMPASS. Unexpectedly, without enhancing COMPASS-nucleosome interaction, H2Bub activates the enzymatic assembly by packing against Swd1 and alleviating the inhibitory effect of the Set1 ARM upon fastening it to the acidic patch. By delineating the spatial configuration of the COMPASS-H2Bub-nucleosome assembly, our studies establish the structural framework for understanding the long-studied H2Bub-H3K4me histone modification crosstalk.


Histone Methyltransferases/ultrastructure , Histones/ultrastructure , Chromatin/genetics , Cryoelectron Microscopy/methods , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Epigenomics/methods , Fungal Proteins/chemistry , Histone Methyltransferases/chemistry , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Histones/genetics , Kluyveromyces/genetics , Kluyveromyces/metabolism , Methyltransferases/metabolism , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Nucleosomes/metabolism , Protein Subunits , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitination
10.
Proc Natl Acad Sci U S A ; 116(44): 22152-22157, 2019 10 29.
Article En | MEDLINE | ID: mdl-31611394

A lysine-to-methionine mutation at lysine 27 of histone 3 (H3K27M) has been shown to promote oncogenesis in a subset of pediatric gliomas. While there is evidence that this "oncohistone" mutation acts by inhibiting the histone methyltransferase PRC2, the details of this proposed mechanism nevertheless continue to be debated. Recent evidence suggests that PRC2 must simultaneously bind both H3K27M and H3K27me3 to experience competitive inhibition of its methyltransferase activity. In this work, we used PRC2 inhibitor treatments in a transgenic H3K27M cell line to validate this dependence in a cellular context. We further used designer chromatin inhibitors to probe the geometric constraints of PRC2 engagement of H3K27M and H3K27me3 in a biochemical setting. We found that PRC2 binds to a bivalent inhibitor unit consisting of an H3K27M and an H3K27me3 nucleosome and exhibits a distance dependence in its affinity for such an inhibitor, which favors closer proximity of the 2 nucleosomes within a chromatin array. Together, our data precisely delineate fundamental aspects of the H3K27M inhibitor and support a model wherein PRC2 becomes trapped at H3K27M-H3K27me3 boundaries.


Histones/genetics , Polycomb Repressive Complex 2/physiology , Amino Acid Substitution , Binding Sites , Cell Line , Histone Methyltransferases/antagonists & inhibitors , Histone Methyltransferases/chemistry , Histones/chemistry , Histones/physiology , Humans , Models, Molecular , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism
11.
Curr Opin Struct Biol ; 59: 98-106, 2019 12.
Article En | MEDLINE | ID: mdl-31229920

Covalent modifications of histone proteins regulate a wide variety of cellular processes. Methylation of histone H3K79 and H3K4 is associated with active transcription and is catalyzed by Dot1L and Set1, respectively. Both Dot1L and Set1 are activated by prior ubiquitination of histone H2B on K120 in a process termed 'histone crosstalk'. Recent structures of Dot1L bound to a ubiquitinated nucleosome revealed how Dot1L is activated by ubiquitin and how Dot1L distorts the nucleosome to access its substrate. Structures of Dot1L-interacting proteins have provided insight into how Dot1L is recruited to sites of active transcription. Cryo-EM and crystallographic studies of the complex of proteins associated with Set1 (COMPASS), uncovered the architecture of COMPASS and how Set1 is activated upon complex assembly.


Histone Methyltransferases/metabolism , Histones/metabolism , Ubiquitin/metabolism , Binding Sites , Enzyme Activation , Histone Methyltransferases/chemistry , Histones/chemistry , Models, Molecular , Molecular Conformation , Protein Binding , Structure-Activity Relationship , Ubiquitin/chemistry
12.
Elife ; 82019 02 20.
Article En | MEDLINE | ID: mdl-30785395

SETD3 is a member of the SET (Su(var)3-9, Enhancer of zeste, and Trithorax) domain protein superfamily and plays important roles in hypoxic pulmonary hypertension, muscle differentiation, and carcinogenesis. Previously, we identified SETD3 as the actin-specific methyltransferase that methylates the N3 of His73 on ß-actin (Kwiatkowski et al., 2018). Here, we present two structures of S-adenosyl-L-homocysteine-bound SETD3 in complex with either an unmodified ß-actin peptide or its His-methylated variant. Structural analyses, supported by biochemical experiments and enzyme activity assays, indicate that the recognition and methylation of ß-actin by SETD3 are highly sequence specific, and that both SETD3 and ß-actin adopt pronounced conformational changes upon binding to each other. In conclusion, this study is the first to show a catalytic mechanism of SETD3-mediated histidine methylation on ß-actin, which not only throws light on the protein histidine methylation phenomenon but also facilitates the design of small molecule inhibitors of SETD3.


Actins/chemistry , Histone Methyltransferases/chemistry , Protein Conformation , S-Adenosylhomocysteine/chemistry , Actins/genetics , Animals , Binding Sites , Histidine/chemistry , Histidine/genetics , Histone Methyltransferases/genetics , Humans , Methylation , Mice , Peptides/chemistry , Protein Binding
13.
Eur J Med Chem ; 166: 351-368, 2019 Mar 15.
Article En | MEDLINE | ID: mdl-30735901

With each newly disclosed resistance mechanism, management of cancer with previously established targets have become a "failure" oriented approach. Molecular targets such as kinases did initially provide a ray of hope against cancer but with decades of struggle between novel therapeutic agents and more sophisticated resistance mechanisms, they seem to have saturated as anti-cancer targets. Now, with more exhaustive molecular recognition techniques and approaches, epigenetic targets have accessed the centre stage as anti-cancer targets. Accordingly, several classes of epigenetic enzymes are being studied for this role and histone methyltransferases form one such class. They include a class of epigenetic enzymes which transfer methyl group from histone proteins and maintain genetic homeostasis. In cancer, several reports have deduced upregulation of different members of this family according to the tumor environment, establishing them as one of the novel anti-cancer targets. This compilation provides an updated information on several members of histone methyltransferases family as epigenetic targets for developing novel anti-cancer agents.


Epigenesis, Genetic , Histone Methyltransferases/metabolism , Animals , Cell Proliferation/genetics , Histone Methyltransferases/chemistry , Humans
14.
Nucleic Acids Res ; 47(1): 122-133, 2019 01 10.
Article En | MEDLINE | ID: mdl-30329085

Transposons impart dynamism to the genomes they inhabit and their movements frequently rewire the control of nearby genes. Occasionally, their proteins are domesticated when they evolve a new function. SETMAR is a protein methylase with a sequence-specific DNA binding domain. It began to evolve about 50 million years ago when an Hsmar1 transposon integrated downstream of a SET-domain methylase gene. Here we show that the DNA-binding domain of the transposase targets the enzyme to transposon-end remnants and that this is capable of regulating gene expression, dependent on the methylase activity. When SETMAR was modestly overexpressed in human cells, almost 1500 genes changed expression by more than 2-fold (65% up- and 35% down-regulated). These genes were enriched for the KEGG Pathways in Cancer and include several transcription factors important for development and differentiation. Expression of a similar level of a methylase-deficient SETMAR changed the expression of many fewer genes, 77% of which were down-regulated with no significant enrichment of KEGG Pathways. Our data is consistent with a model in which SETMAR is part of an anthropoid primate-specific regulatory network centered on the subset of genes containing a transposon end.


DNA-Binding Proteins/genetics , Histone Methyltransferases/genetics , Histone-Lysine N-Methyltransferase/genetics , Transcriptome/genetics , Animals , DNA Transposable Elements/genetics , DNA-Binding Proteins/chemistry , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks/genetics , Histone Methyltransferases/chemistry , Histones/genetics , Humans , Methylation , Neoplasms/genetics , Primates/genetics , Signal Transduction/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
15.
Nucleic Acids Res ; 46(21): 11129-11143, 2018 11 30.
Article En | MEDLINE | ID: mdl-30325428

H2B ubiquitylation (H2Bub)-dependent H3K4 methylation is mediated by the multisubunit Set1 complex (Set1C) in yeast, but precisely how Set1C subunits contribute to this histone modification remains unclear. Here, using reconstituted Set1Cs and recombinant H2Bub chromatin, we identified Set1C subunits and domains involved in the H2Bub-dependent H3K4 methylation process, showing that the Spp1 PHDL domain, in conjunction with the Set1 n-SET domain, interacts with Swd1/Swd3 and that this interaction is essential for H2Bub-dependent H3K4 methylation. Importantly, Set1C containing an Spp1-Swd1 fusion bypasses the requirement for H2Bub for H3K4 methylation, suggesting that the role of H2Bub is to induce allosteric rearrangements of the subunit-interaction network within the active site of Set1C that are necessary for methylation activity. Moreover, the interaction between the Set1 N-terminal region and Swd1 renders the Spp1-lacking Set1C competent for H2Bub-dependent H3K4 methylation. Collectively, our results suggest that H2Bub induces conformational changes in Set1C that support H3K4 methylation activity.


Histone-Lysine N-Methyltransferase/chemistry , Histones/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Catalytic Domain , Chromatin/chemistry , Histone Methyltransferases/chemistry , Methylation , Protein Binding , Protein Multimerization , Protein Processing, Post-Translational , Recombinant Fusion Proteins/chemistry , Saccharomyces cerevisiae/genetics , Ubiquitination
16.
Cell ; 174(5): 1117-1126.e12, 2018 08 23.
Article En | MEDLINE | ID: mdl-30100186

The methylation of histone 3 lysine 4 (H3K4) is carried out by an evolutionarily conserved family of methyltransferases referred to as complex of proteins associated with Set1 (COMPASS). The activity of the catalytic SET domain (su(var)3-9, enhancer-of-zeste, and trithorax) is endowed through forming a complex with a set of core proteins that are widely shared from yeast to humans. We obtained cryo-electron microscopy (cryo-EM) maps of the yeast Set1/COMPASS core complex at overall 4.0- to 4.4-Å resolution, providing insights into its structural organization and conformational dynamics. The Cps50 C-terminal tail weaves within the complex to provide a central scaffold for assembly. The SET domain, snugly positioned at the junction of the Y-shaped complex, is extensively contacted by Cps60 (Bre2), Cps50 (Swd1), and Cps30 (Swd3). The mobile SET-I motif of the SET domain is engaged by Cps30, explaining its key role in COMPASS catalytic activity toward higher H3K4 methylation states.


Fungal Proteins/chemistry , Histone Methyltransferases/chemistry , Histones/chemistry , Animals , Catalytic Domain , Chaetomium/chemistry , Chromatin/chemistry , Cryoelectron Microscopy , DNA-Binding Proteins/chemistry , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/chemistry , Humans , Insecta , Intracellular Signaling Peptides and Proteins , Methylation , Protein Subunits , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Software
17.
Nature ; 560(7719): 504-508, 2018 08.
Article En | MEDLINE | ID: mdl-30051891

Histone H3 lysine 9 methylation (H3K9me) mediates heterochromatic gene silencing and is important for genome stability and the regulation of gene expression1-4. The establishment and epigenetic maintenance of heterochromatin involve the recruitment of H3K9 methyltransferases to specific sites on DNA, followed by the recognition of pre-existing H3K9me by the methyltransferase and methylation of proximal histone H35-11. This positive feedback loop must be tightly regulated to prevent deleterious epigenetic gene silencing. Extrinsic anti-silencing mechanisms involving histone demethylation or boundary elements help to limit the spread of inappropriate H3K9me12-15. However, how H3K9 methyltransferase activity is locally restricted or prevented from initiating random H3K9me-which would lead to aberrant gene silencing and epigenetic instability-is not fully understood. Here we reveal an autoinhibited conformation in the conserved H3K9 methyltransferase Clr4 (also known as Suv39h) of the fission yeast Schizosaccharomyces pombe that has a critical role in preventing aberrant heterochromatin formation. Biochemical and X-ray crystallographic data show that an internal loop in Clr4 inhibits the catalytic activity of this enzyme by blocking the histone H3K9 substrate-binding pocket, and that automethylation of specific lysines in this loop promotes a conformational switch that enhances the H3K9me activity of Clr4. Mutations that are predicted to disrupt this regulation lead to aberrant H3K9me, loss of heterochromatin domains and inhibition of growth, demonstrating the importance of the intrinsic inhibition and auto-activation of Clr4 in regulating the deposition of H3K9me and in preventing epigenetic instability. Conservation of the Clr4 autoregulatory loop in other H3K9 methyltransferases and the automethylation of a corresponding lysine in the human SUV39H2 homologue16 suggest that the mechanism described here is broadly conserved.


Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Epigenesis, Genetic , Histone Methyltransferases/chemistry , Histone Methyltransferases/metabolism , Histones/metabolism , Methyltransferases/chemistry , Methyltransferases/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces/genetics , Evolution, Molecular , Gene Silencing , Heterochromatin/chemistry , Heterochromatin/genetics , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Humans , Methylation , Protein Conformation
...