Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Science ; 381(6660): eadd8673, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37616371

ABSTRACT

Chromatin inheritance entails de novo nucleosome assembly after DNA replication by chromatin assembly factor-1 (CAF-1). Yet direct knowledge about CAF-1's histone binding mode and nucleosome assembly process is lacking. In this work, we report the crystal structure of human CAF-1 in the absence of histones and the cryo-electron microscopy structure of CAF-1 in complex with histones H3 and H4. One histone H3-H4 heterodimer is bound by one CAF-1 complex mainly through the p60 subunit and the acidic domain of the p150 subunit. We also observed a dimeric CAF-1-H3-H4 supercomplex in which two H3-H4 heterodimers are poised for tetramer assembly and discovered that CAF-1 facilitates right-handed DNA wrapping of H3-H4 tetramers. These findings signify the involvement of DNA in H3-H4 tetramer formation and suggest a right-handed nucleosome precursor in chromatin replication.


Subject(s)
Chromatin Assembly Factor-1 , Histones , Nucleosomes , Humans , Chromatin , Chromatin Assembly Factor-1/chemistry , Cryoelectron Microscopy , Histones/chemistry , Crystallography, X-Ray , Protein Domains
2.
Nature ; 616(7955): 176-182, 2023 04.
Article in English | MEDLINE | ID: mdl-36991118

ABSTRACT

Repression of gene expression by protein complexes of the Polycomb group is a fundamental mechanism that governs embryonic development and cell-type specification1-3. The Polycomb repressive deubiquitinase (PR-DUB) complex removes the ubiquitin moiety from monoubiquitinated histone H2A K119 (H2AK119ub1) on the nucleosome4, counteracting the ubiquitin E3 ligase activity of Polycomb repressive complex 1 (PRC1)5 to facilitate the correct silencing of genes by Polycomb proteins and safeguard active genes from inadvertent silencing by PRC1 (refs. 6-9). The intricate biological function of PR-DUB requires accurate targeting of H2AK119ub1, but PR-DUB can deubiquitinate monoubiquitinated free histones and peptide substrates indiscriminately; the basis for its exquisite nucleosome-dependent substrate specificity therefore remains unclear. Here we report the cryo-electron microscopy structure of human PR-DUB, composed of BAP1 and ASXL1, in complex with the chromatosome. We find that ASXL1 directs the binding of the positively charged C-terminal extension of BAP1 to nucleosomal DNA and histones H3-H4 near the dyad, an addition to its role in forming the ubiquitin-binding cleft. Furthermore, a conserved loop segment of the catalytic domain of BAP1 is situated near the H2A-H2B acidic patch. This distinct nucleosome-binding mode displaces the C-terminal tail of H2A from the nucleosome surface, and endows PR-DUB with the specificity for H2AK119ub1.


Subject(s)
Deubiquitinating Enzymes , Histones , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Humans , Cryoelectron Microscopy , Histones/chemistry , Histones/metabolism , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/metabolism , Polycomb Repressive Complex 1/chemistry , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 1/ultrastructure , Polycomb-Group Proteins/chemistry , Polycomb-Group Proteins/metabolism , Polycomb-Group Proteins/ultrastructure , Ubiquitin/metabolism , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/ultrastructure , Ubiquitination , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Repressor Proteins/ultrastructure , Catalytic Domain , Deubiquitinating Enzymes/classification , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/ultrastructure , Substrate Specificity , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/ultrastructure
3.
Nat Commun ; 13(1): 5703, 2022 09 28.
Article in English | MEDLINE | ID: mdl-36171202

ABSTRACT

RNA polymerase II (Pol II) apparatuses are compartmentalized into transcriptional clusters. Whether protein factors control these clusters remains unknown. In this study, we find that the ATPase-associated with diverse cellular activities (AAA + ) ATPase RUVBL2 co-occupies promoters with Pol II and various transcription factors. RUVBL2 interacts with unphosphorylated Pol II in chromatin to promote RPB1 carboxy-terminal domain (CTD) clustering and transcription initiation. Rapid depletion of RUVBL2 leads to a decrease in the number of Pol II clusters and inhibits nascent RNA synthesis, and tethering RUVBL2 to an active promoter enhances Pol II clustering at the promoter. We also identify target genes that are directly linked to the RUVBL2-Pol II axis. Many of these genes are hallmarks of cancers and encode proteins with diverse cellular functions. Our results demonstrate an emerging activity for RUVBL2 in regulating Pol II cluster formation in the nucleus.


Subject(s)
RNA Polymerase II , Transcription Factors , Adenosine Triphosphatases/metabolism , Chromatin/genetics , Cluster Analysis , RNA , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Transcription, Genetic
4.
Genes Dev ; 36(7-8): 408-413, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35393344

ABSTRACT

Chaperones influence histone conformation and intermolecular interaction in multiprotein complexes, and the structures obtained with full-length histones often provide more accurate and comprehensive views. Here, our structure of the Hat1-Hat2 acetyltransferase complex bound to Asf1-H3-H4 shows that the core domains of H3 and H4 are involved in binding Hat1 and Hat2, and the N-terminal tail of H3 makes extensive interaction with Hat2. These findings expand the knowledge about histone-protein interaction and implicate a function of Hat2/RbAp46/48, which is a versatile histone chaperone found in many chromatin-associated complexes, in the passing of histones between chaperones.


Subject(s)
Histone Acetyltransferases , Histones , Acetylation , Cell Cycle Proteins/metabolism , Chromatin , Histone Acetyltransferases/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Molecular Chaperones/genetics
5.
Science ; 375(6584): 1053-1058, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35143257

ABSTRACT

Bivalent genes are ready for activation upon the arrival of developmental cues. Here, we report that BEND3 is a CpG island (CGI)-binding protein that is enriched at regulatory elements. The cocrystal structure of BEND3 in complex with its target DNA reveals the structural basis for its DNA methylation-sensitive binding property. Mouse embryos ablated of Bend3 died at the pregastrulation stage. Bend3 null embryonic stem cells (ESCs) exhibited severe defects in differentiation, during which hundreds of CGI-containing bivalent genes were prematurely activated. BEND3 is required for the stable association of polycomb repressive complex 2 (PRC2) at bivalent genes that are highly occupied by BEND3, which suggests a reining function of BEND3 in maintaining high levels of H3K27me3 at these bivalent genes in ESCs to prevent their premature activation in the forthcoming developmental stage.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Repressor Proteins/metabolism , Transcriptional Activation , Animals , Chromatin/metabolism , CpG Islands , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , DNA Methylation , Embryonic Stem Cells/cytology , Enhancer Elements, Genetic , Gene Deletion , Gene Expression Regulation, Developmental , Histones/metabolism , Methylation , Mice , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Domains , RNA-Seq , Repressor Proteins/chemistry , Repressor Proteins/genetics , Up-Regulation
6.
Genes Dev ; 35(23-24): 1610-1624, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34819355

ABSTRACT

Chromosomal duplication requires de novo assembly of nucleosomes from newly synthesized histones, and the process involves a dynamic network of interactions between histones and histone chaperones. sNASP and ASF1 are two major histone H3-H4 chaperones found in distinct and common complexes, yet how sNASP binds H3-H4 in the presence and absence of ASF1 remains unclear. Here we show that, in the presence of ASF1, sNASP principally recognizes a partially unfolded Nα region of histone H3, and in the absence of ASF1, an additional sNASP binding site becomes available in the core domain of the H3-H4 complex. Our study also implicates a critical role of the C-terminal tail of H4 in the transfer of H3-H4 between sNASP and ASF1 and the coiled-coil domain of sNASP in nucleosome assembly. These findings provide mechanistic insights into coordinated histone binding and transfer by histone chaperones.


Subject(s)
Histone Chaperones , Histones , Binding Sites , Cell Cycle Proteins/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Nucleosomes , Protein Binding
7.
Mol Cell ; 78(3): 423-433.e5, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32220645

ABSTRACT

A commencing and critical step in miRNA biogenesis involves processing of pri-miRNAs in the nucleus by Microprocessor. An important, but not completely understood, question is how Drosha, the catalytic subunit of Microprocessor, binds pri-miRNAs and correctly specifies cleavage sites. Here we report the cryoelectron microscopy structures of the Drosha-DGCR8 complex with and without a pri-miRNA. The RNA-bound structure provides direct visualization of the tertiary structure of pri-miRNA and shows that a helix hairpin in the extended PAZ domain and the mobile basic (MB) helix in the RNase IIIa domain of Drosha coordinate to recognize the single-stranded to double-stranded junction of RNA, whereas the dsRNA binding domain makes extensive contacts with the RNA stem. Furthermore, the RNA-free structure reveals an autoinhibitory conformation of the PAZ helix hairpin. These findings provide mechanistic insights into pri-miRNA cleavage site selection and conformational dynamics governing pri-miRNA recognition by the catalytic component of Microprocessor.


Subject(s)
MicroRNAs/chemistry , MicroRNAs/metabolism , Ribonuclease III/chemistry , Ribonuclease III/metabolism , Animals , Cryoelectron Microscopy , Humans , Models, Molecular , Protein Conformation , Protein Domains , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonuclease III/genetics , Spodoptera/cytology
8.
Structure ; 27(5): 837-845.e3, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30827843

ABSTRACT

The evolutionarily conserved Trithorax group protein Ash1 is a SET domain histone methyltransferase that mono- and dimethylates lysine 36 of histone H3 (H3K36). Ash1 forms a complex with Mrg15 and Nurf55, and the binding of Mrg15 greatly stimulates the catalytic activity of Ash1, yet the underlying molecular mechanisms remain unknown. Here we report the crystal structure of the tandem Mrg15-interacting and SET domains of human Ash1L in complex with Mrg15. Ash1L interacts with Mrg15 principally via a segment located N-terminal to the catalytic SET domain. Surprisingly, an autoinhibitory loop in the post-SET region of Ash1L is destabilized on Mrg15 binding despite no direct contact. Dynamics of the autoinhibitory loop can be attributed to subtle structural changes of the S-adenosylmethionine (SAM) binding pocket induced by Mrg15 binding, implicating a mechanism of conformational coupling between SAM and substrate binding sites. The findings broaden the understanding of regulation of H3K36 methyltransferases.


Subject(s)
DNA-Binding Proteins/chemistry , Histone-Lysine N-Methyltransferase/chemistry , Histones/chemistry , Transcription Factors/chemistry , Binding Sites , Catalytic Domain , Computer Simulation , Crystallography, X-Ray , Humans , Hydrogen Bonding , Methylation , Protein Binding
9.
BMC Biol ; 16(1): 110, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30285846

ABSTRACT

BACKGROUND: H3.3 is an ancient and conserved H3 variant and plays essential roles in transcriptional regulation. HIRA complex, which is composed of HIRA, UBN1 or UBN2, and Cabin1, is a H3.3 specific chaperone complex. However, it still remains largely uncharacterized how HIRA complex specifically recognizes and deposits H3.3 to the chromatin, such as promoters and enhancers. RESULTS: In this study, we demonstrate that the UBN1 or UBN2 subunit is mainly responsible for specific recognition and direct binding of H3.3 by the HIRA complex. While the HIRA subunit can enhance the binding affinity of UBN1 toward H3.3, Cabin1 subunit cannot. We also demonstrate that both Ala87 and Gly90 residues of H3.3 are required and sufficient for the specific recognition and binding by UBN1. ChIP-seq studies reveal that two independent HIRA complexes (UBN1-HIRA and UBN2-HIRA) can cooperatively deposit H3.3 to cis-regulatory regions, including active promoters and active enhancers in mouse embryonic stem (mES) cells. Importantly, disruption of histone chaperone activities of UBN1 and UBN2 by FID/AAA mutation results in the defect of H3.3 deposition at promoters of developmental genes involved in neural differentiation, and subsequently causes the failure of activation of these genes during neural differentiation of mES cells. CONCLUSION: Together, our results provide novel insights into the mechanism by which the HIRA complex specifically recognizes and deposits H3.3 at promoters and enhancers of developmental genes, which plays a critical role in neural differentiation of mES cells.


Subject(s)
Gene Expression Regulation , Histones/genetics , Mouse Embryonic Stem Cells/metabolism , Nuclear Proteins/genetics , Regulatory Sequences, Nucleic Acid , Adaptor Proteins, Signal Transducing , Animals , Calcineurin/genetics , Calcineurin/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Histone Chaperones/genetics , Histone Chaperones/metabolism , Histones/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Genes Dev ; 30(21): 2391-2403, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27881601

ABSTRACT

Assembly of the spliceosomal small nuclear ribonucleoparticle (snRNP) core requires the participation of the multisubunit SMN (survival of motor neuron) complex, which contains SMN and several Gemin proteins. The SMN and Gemin2 subunits directly bind Sm proteins, and Gemin5 is required for snRNP biogenesis and has been implicated in snRNA recognition. The RNA sequence required for snRNP assembly includes the Sm site and an adjacent 3' stem-loop, but a precise understanding of Gemin5's RNA-binding specificity is lacking. Here we show that the N-terminal half of Gemin5, which is composed of two juxtaposed seven-bladed WD40 repeat domains, recognizes the Sm site. The tandem WD40 repeat domains are rigidly held together to form a contiguous RNA-binding surface. RNA-contacting residues are located mostly on loops between ß strands on the apical surface of the WD40 domains. Structural and biochemical analyses show that base-stacking interactions involving four aromatic residues and hydrogen bonding by a pair of arginines are crucial for specific recognition of the Sm sequence. We also show that an adenine immediately 5' to the Sm site is required for efficient binding and that Gemin5 can bind short RNA oligos in an alternative mode. Our results provide mechanistic understandings of Gemin5's snRNA-binding specificity as well as valuable insights into the molecular mechanism of RNA binding by WD40 repeat proteins in general.


Subject(s)
Models, Molecular , RNA, Small Nuclear/metabolism , SMN Complex Proteins/chemistry , SMN Complex Proteins/metabolism , WD40 Repeats/physiology , Crystallization , Guanosine/analogs & derivatives , Guanosine/metabolism , Humans , Protein Binding , Protein Structure, Tertiary , RNA, Small Nuclear/chemistry
11.
Nat Struct Mol Biol ; 19(12): 1287-92, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23142979

ABSTRACT

Mammalian histone H3.3 is a variant of the canonical H3.1 essential for genome reprogramming in fertilized eggs and maintenance of chromatin structure in neuronal cells. An H3.3-specific histone chaperone, DAXX, directs the deposition of H3.3 onto pericentric and telomeric heterochromatin. H3.3 differs from H3.1 by only five amino acids, yet DAXX can distinguish the two with high precision. By a combination of structural, biochemical and cell-based targeting analyses, we show that Ala87 and Gly90 are the principal determinants of human H3.3 specificity. DAXX uses a shallow hydrophobic pocket to accommodate the small hydrophobic Ala87 of H3.3, whereas a polar binding environment in DAXX prefers Gly90 in H3.3 over the hydrophobic Met90 in H3.1. An H3.3-H4 heterodimer is bound by the histone-binding domain of DAXX, which makes extensive contacts with both H3.3 and H4.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Histones/chemistry , Molecular Chaperones/chemistry , Nuclear Proteins/chemistry , Co-Repressor Proteins , Dimerization , Humans , Models, Molecular
12.
Article in English | MEDLINE | ID: mdl-21301083

ABSTRACT

Streptococcus mutans is one of the pathogenic species involved in dental caries, especially in the initiation and development stages. Here, the crystal structure of SMU.595, a putative dihydroorotate dehydrogenase (DHOD) from S. mutans, is reported at 2.4 Šresolution. DHOD is a flavin mononucleotide-containing enzyme which catalyzes the oxidation of L-dihydroorotate to orotate, which is the fourth step and the only redox reaction in the de novo biosynthesis of pyrimidine nucleotides. The reductive lysine-methylation procedure was applied in order to improve the diffraction qualities of the crystals. Analysis of the S. mutans DHOD crystal structure shows that this enzyme is a class 1A DHOD and also suggests potential sites that could be exploited for the design of highly specific inhibitors using the structure-based chemotherapeutic design technique.


Subject(s)
Oxidoreductases Acting on CH-CH Group Donors/chemistry , Streptococcus mutans/enzymology , Amino Acid Sequence , Binding Sites/genetics , Catalysis , Conserved Sequence , Crystallography, X-Ray/methods , Dental Caries/microbiology , Dihydroorotate Dehydrogenase , Dimerization , Escherichia coli/genetics , Flavin Mononucleotide/chemistry , Flavin Mononucleotide/metabolism , Histidine/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Lysine/metabolism , Methylation , Models, Molecular , Molecular Sequence Data , Orotic Acid/chemistry , Oxidation-Reduction , Oxidoreductases Acting on CH-CH Group Donors/classification , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Protein Conformation , Protein Folding , Protein Structure, Quaternary , Protein Structure, Secondary/genetics , Protein Subunits/chemistry , Protein Subunits/metabolism , Pyrimidines/biosynthesis , Pyrimidines/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , X-Ray Diffraction
13.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 66(Pt 5): 498-502, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20445243

ABSTRACT

Orotate phosphoribosyltransferase (OPRTase) catalyzes the OMP-forming step in de novo pyrimidine-nucleotide biosynthesis. Here, the crystal structure of OPRTase from the caries pathogen Streptococcus mutans is reported at 2.4 A resolution. S. mutans OPRTase forms a symmetric dimer and each monomer binds two sulfates at the active sites. The structural symmetry of the sulfate-binding sites and the missing loops in this structure are consistent with a symmetric catalysis mechanism.


Subject(s)
Orotate Phosphoribosyltransferase/chemistry , Streptococcus mutans/enzymology , Binding Sites , Crystallography, X-Ray , Hydrogen Bonding , Models, Molecular , Protein Structure, Quaternary , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...