Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
ACS Chem Biol ; 17(9): 2572-2582, 2022 09 16.
Article in English | MEDLINE | ID: mdl-35973051

ABSTRACT

Targeting the lysine deacetylase activity of class I histone deacetylases (HDACs) is potentially beneficial for the treatment of several diseases including human immunodeficiency virus (HIV) infection, Alzheimer's disease, and various cancers. It is therefore important to understand the function and mechanism of action of these enzymes. Class I HDACs act as catalytic components of seven large, multiprotein corepressor complexes. Different HDAC corepressor complexes have specific, nonredundant roles in the cell. It is likely that their specific functions are at least partly influenced by the substrate specificity of the complexes. To address this, we developed chemical tools to probe the specificity of HDAC complexes. We assessed a library of acetyl-lysine-containing substrate peptides and hydroxamic acid-containing inhibitor peptides against the full range of class I HDAC corepressor complexes. The results suggest that site-specific HDAC corepressor complex activity is driven in part by the recognition of the primary amino acid sequence surrounding a particular lysine position in the histone tail.


Subject(s)
Hydroxamic Acids , Peptide Library , Co-Repressor Proteins/metabolism , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Hydroxamic Acids/chemistry , Lysine , Peptides/chemistry
2.
Nucleic Acids Res ; 48(22): 12972-12982, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33264408

ABSTRACT

Class I histone deacetylase complexes play essential roles in many nuclear processes. Whilst they contain a common catalytic subunit, they have diverse modes of action determined by associated factors in the distinct complexes. The deacetylase module from the NuRD complex contains three protein domains that control the recruitment of chromatin to the deacetylase enzyme, HDAC1/2. Using biochemical approaches and cryo-electron microscopy, we have determined how three chromatin-binding domains (MTA1-BAH, MBD2/3 and RBBP4/7) are assembled in relation to the core complex so as to facilitate interaction of the complex with the genome. We observe a striking arrangement of the BAH domains suggesting a potential mechanism for binding to di-nucleosomes. We also find that the WD40 domains from RBBP4 are linked to the core with surprising flexibility that is likely important for chromatin engagement. A single MBD2 protein binds asymmetrically to the dimerisation interface of the complex. This symmetry mismatch explains the stoichiometry of the complex. Finally, our structures suggest how the holo-NuRD might assemble on a di-nucleosome substrate.


Subject(s)
Chromatin/genetics , DNA-Binding Proteins/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Repressor Proteins/genetics , Retinoblastoma-Binding Protein 4/genetics , Trans-Activators/genetics , Amino Acid Sequence/genetics , Cryoelectron Microscopy , DNA-Binding Proteins/ultrastructure , Histone Deacetylase 1/genetics , Histone Deacetylase 1/ultrastructure , Histone Deacetylases/genetics , Histone Deacetylases/ultrastructure , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/ultrastructure , Nucleosomes/genetics , Nucleosomes/ultrastructure , Protein Binding/genetics , Protein Domains/genetics , Repressor Proteins/ultrastructure , Retinoblastoma-Binding Protein 4/ultrastructure , Trans-Activators/ultrastructure
3.
Nat Commun ; 11(1): 3252, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32591534

ABSTRACT

MiDAC is one of seven distinct, large multi-protein complexes that recruit class I histone deacetylases to the genome to regulate gene expression. Despite implications of involvement in cell cycle regulation and in several cancers, surprisingly little is known about the function or structure of MiDAC. Here we show that MiDAC is important for chromosome alignment during mitosis in cancer cell lines. Mice lacking the MiDAC proteins, DNTTIP1 or MIDEAS, die with identical phenotypes during late embryogenesis due to perturbations in gene expression that result in heart malformation and haematopoietic failure. This suggests that MiDAC has an essential and unique function that cannot be compensated by other HDAC complexes. Consistent with this, the cryoEM structure of MiDAC reveals a unique and distinctive mode of assembly. Four copies of HDAC1 are positioned at the periphery with outward-facing active sites suggesting that the complex may target multiple nucleosomes implying a processive deacetylase function.


Subject(s)
Embryonic Development , Histone Deacetylases/metabolism , Multiprotein Complexes/metabolism , Amino Acid Sequence , Animals , Cell Line , Chromatin/metabolism , Chromosomes, Mammalian/metabolism , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Gene Regulatory Networks , Heterozygote , Homozygote , Humans , Mice, Inbred C57BL , Mice, Knockout , Mitosis , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Nuclear Proteins/metabolism , Protein Domains , Protein Multimerization
4.
Elife ; 92020 06 05.
Article in English | MEDLINE | ID: mdl-32501215

ABSTRACT

Histone acetylation regulates chromatin structure and gene expression and is removed by histone deacetylases (HDACs). HDACs are commonly found in various protein complexes to confer distinct cellular functions, but how the multi-subunit complexes influence deacetylase activities and site-selectivities in chromatin is poorly understood. Previously we reported the results of studies on the HDAC1 containing CoREST complex and acetylated nucleosome substrates which revealed a notable preference for deacetylation of histone H3 acetyl-Lys9 vs. acetyl-Lys14 (Wu et al, 2018). Here we analyze the enzymatic properties of five class I HDAC complexes: CoREST, NuRD, Sin3B, MiDAC and SMRT with site-specific acetylated nucleosome substrates. Our results demonstrate that these HDAC complexes show a wide variety of deacetylase rates in a site-selective manner. A Gly13 in the histone H3 tail is responsible for a sharp reduction in deacetylase activity of the CoREST complex for H3K14ac. These studies provide a framework for connecting enzymatic and biological functions of specific HDAC complexes.


Subject(s)
Histone Deacetylases/metabolism , Histones/metabolism , Nucleosomes/metabolism , Acetylation , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Histone Deacetylases/genetics , Histones/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nucleosomes/genetics
5.
Chem Commun (Camb) ; 56(32): 4476-4479, 2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32201871

ABSTRACT

We have identified a proteolysis targeting chimera (PROTAC) of class I HDACs 1, 2 and 3. The most active degrader consists of a benzamide HDAC inhibitor, an alkyl linker, and the von Hippel-Lindau E3 ligand. Our PROTAC increased histone acetylation levels and compromised colon cancer HCT116 cell viability, establishing a degradation strategy as an alternative to class I HDAC inhibition.


Subject(s)
Co-Repressor Proteins , Histone Deacetylases , Animals , Humans , Mice , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Co-Repressor Proteins/metabolism , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histone Demethylases/antagonists & inhibitors , Proteolysis
6.
Hum Mol Genet ; 28(15): 2501-2513, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31067316

ABSTRACT

Craniosynostosis, the premature ossification of cranial sutures, is a developmental disorder of the skull vault, occurring in approximately 1 in 2250 births. The causes are heterogeneous, with a monogenic basis identified in ~25% of patients. Using whole-genome sequencing, we identified a novel, de novo variant in BCL11B, c.7C>A, encoding an R3S substitution (p.R3S), in a male patient with coronal suture synostosis. BCL11B is a transcription factor that interacts directly with the nucleosome remodelling and deacetylation complex (NuRD) and polycomb-related complex 2 (PRC2) through the invariant proteins RBBP4 and RBBP7. The p.R3S substitution occurs within a conserved amino-terminal motif (RRKQxxP) of BCL11B and reduces interaction with both transcriptional complexes. Equilibrium binding studies and molecular dynamics simulations show that the p.R3S substitution disrupts ionic coordination between BCL11B and the RBBP4-MTA1 complex, a subassembly of the NuRD complex, and increases the conformational flexibility of Arg-4, Lys-5 and Gln-6 of BCL11B. These alterations collectively reduce the affinity of BCL11B p.R3S for the RBBP4-MTA1 complex by nearly an order of magnitude. We generated a mouse model of the BCL11B p.R3S substitution using a CRISPR-Cas9-based approach, and we report herein that these mice exhibit craniosynostosis of the coronal suture, as well as other cranial sutures. This finding provides strong evidence that the BCL11B p.R3S substitution is causally associated with craniosynostosis and confirms an important role for BCL11B in the maintenance of cranial suture patency.


Subject(s)
Chromatin Assembly and Disassembly , Cranial Sutures/growth & development , Craniosynostoses/metabolism , Mutation, Missense , Nucleosomes/metabolism , Osteogenesis , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Cranial Sutures/metabolism , Craniosynostoses/genetics , Craniosynostoses/physiopathology , DNA Mutational Analysis , Disease Models, Animal , Humans , Infant , Male , Mice , Protein Binding , Protein Conformation , Repressor Proteins/metabolism , Repressor Proteins/physiology , Retinoblastoma-Binding Protein 4/metabolism , Trans-Activators/metabolism , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology , White People , Whole Genome Sequencing
7.
Org Lett ; 21(9): 3178-3182, 2019 05 03.
Article in English | MEDLINE | ID: mdl-30998366

ABSTRACT

Syntheses of Fmoc amino acids having zinc-binding groups were prepared and incorporated into substrate inhibitor H3K27 peptides using Fmoc/tBu solid-phase peptide synthesis (SPPS). Peptide 11, prepared using Fmoc-Asu(NHOtBu)-OH, is a potent inhibitor (IC50 = 390 nM) of the core NuRD corepressor complex (HDAC1-MTA1-RBBP4). The Fmoc amino acids have the potential to facilitate the rapid preparation of substrate peptidomimetic inhibitor (SPI) libraries in the search for selective HDAC inhibitors.


Subject(s)
Amino Acids/chemistry , Fluorenes/chemistry , Histone Deacetylase Inhibitors/chemical synthesis , Peptidomimetics/chemical synthesis , Zinc/chemistry , Chelating Agents/chemistry , Coordination Complexes/chemistry , Nickel/chemistry , Solid-Phase Synthesis Techniques , Stereoisomerism
8.
Nat Commun ; 9(1): 3798, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30228260

ABSTRACT

Transcriptional regulation by chromatin is a highly dynamic process directed through the recruitment and coordinated action of epigenetic modifiers and readers of these modifications. Using an unbiased proteomic approach to find interactors of H3K36me3, a modification enriched on active chromatin, here we identify PWWP2A and HDAC2 among the top interactors. PWWP2A and its paralog PWWP2B form a stable complex with NuRD subunits MTA1/2/3:HDAC1/2:RBBP4/7, but not with MBD2/3, p66α/ß, and CHD3/4. PWWP2A competes with MBD3 for binding to MTA1, thus defining a new variant NuRD complex that is mutually exclusive with the MBD2/3 containing NuRD. In mESCs, PWWP2A/B is most enriched at highly transcribed genes. Loss of PWWP2A/B leads to increases in histone acetylation predominantly at highly expressed genes, accompanied by decreases in Pol II elongation. Collectively, these findings suggest a role for PWWP2A/B in regulating transcription through the fine-tuning of histone acetylation dynamics at actively transcribed genes.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , RNA Polymerase II/metabolism , Transcription Elongation, Genetic , Acetylation , Animals , Chromosomal Proteins, Non-Histone/genetics , DNA Methylation/physiology , DNA-Binding Proteins/metabolism , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Humans , Mice , Mouse Embryonic Stem Cells , Nucleosomes/metabolism , Proteomics
9.
Trends Pharmacol Sci ; 38(4): 363-377, 2017 04.
Article in English | MEDLINE | ID: mdl-28139258

ABSTRACT

Histone deacetylase (HDAC) inhibitors are proven anticancer therapeutics and have potential in the treatment of many other diseases including HIV infection, Alzheimer's disease, and Friedreich's ataxia. A problem with the currently available HDAC inhibitors is that they have limited specificity and target multiple deacetylases. Designing isoform-selective inhibitors has proven challenging due to similarities in the structure and chemistry of HDAC active sites. However, the fact that HDACs 1, 2, and 3 are recruited to several large multi-subunit complexes, each with particular biological functions, raises the possibility of specifically inhibiting individual complexes. This may be assisted by recent structural and functional information about the assembly of these complexes. Here, we review the available structural information and discuss potential targeting strategies.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/physiology , Allosteric Regulation , Animals , Drug Discovery , Histone Deacetylases/chemistry , Humans , Inositol Phosphates/pharmacology
10.
Elife ; 5: e13941, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27098840

ABSTRACT

The NuRD complex is a multi-protein transcriptional corepressor that couples histone deacetylase and ATP-dependent chromatin remodelling activities. The complex regulates the higher-order structure of chromatin, and has important roles in the regulation of gene expression, DNA damage repair and cell differentiation. HDACs 1 and 2 are recruited by the MTA1 corepressor to form the catalytic core of the complex. The histone chaperone protein RBBP4, has previously been shown to bind to the carboxy-terminal tail of MTA1. We show that MTA1 recruits a second copy of RBBP4. The crystal structure reveals an extensive interface between MTA1 and RBBP4. An EM structure, supported by SAXS and crosslinking, reveals the architecture of the dimeric HDAC1:MTA1:RBBP4 assembly which forms the core of the NuRD complex. We find evidence that in this complex RBBP4 mediates interaction with histone H3 tails, but not histone H4, suggesting a mechanism for recruitment of the NuRD complex to chromatin.


Subject(s)
Chromatin/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/chemistry , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Crystallography, X-Ray , Histone Deacetylase 1 , Histone Deacetylase 2 , Histone Deacetylases , Protein Conformation , Repressor Proteins , Retinoblastoma-Binding Protein 4 , Trans-Activators
11.
Nat Commun ; 7: 11262, 2016 04 25.
Article in English | MEDLINE | ID: mdl-27109927

ABSTRACT

Histone deacetylases (HDACs) 1, 2 and 3 form the catalytic subunit of several large transcriptional repression complexes. Unexpectedly, the enzymatic activity of HDACs in these complexes has been shown to be regulated by inositol phosphates, which bind in a pocket sandwiched between the HDAC and co-repressor proteins. However, the actual mechanism of activation remains poorly understood. Here we have elucidated the stereochemical requirements for binding and activation by inositol phosphates, demonstrating that activation requires three adjacent phosphate groups and that other positions on the inositol ring can tolerate bulky substituents. We also demonstrate that there is allosteric communication between the inositol-binding site and the active site. The crystal structure of the HDAC1:MTA1 complex bound to a novel peptide-based inhibitor and to inositol hexaphosphate suggests a molecular basis of substrate recognition, and an entropically driven allosteric mechanism of activation.


Subject(s)
Histone Deacetylase 1/metabolism , Histone Deacetylases/metabolism , Inositol Phosphates/metabolism , Multiprotein Complexes/metabolism , Allosteric Regulation , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Enzyme Activation/drug effects , HEK293 Cells , Histone Deacetylase 1/chemistry , Histone Deacetylase 1/genetics , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , Humans , Inositol Phosphates/chemistry , Molecular Docking Simulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Protein Binding , Protein Domains
12.
Nucleic Acids Res ; 43(4): 2033-44, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25653165

ABSTRACT

Recent proteomic studies have identified a novel histone deacetylase complex that is upregulated during mitosis and is associated with cyclin A. This complex is conserved from nematodes to man and contains histone deacetylases 1 and 2, the MIDEAS corepressor protein and a protein called DNTTIP1 whose function was hitherto poorly understood. Here, we report the structures of two domains from DNTTIP1. The amino-terminal region forms a tight dimerization domain with a novel structural fold that interacts with and mediates assembly of the HDAC1:MIDEAS complex. The carboxy-terminal domain of DNTTIP1 has a structure related to the SKI/SNO/DAC domain, despite lacking obvious sequence homology. We show that this domain in DNTTIP1 mediates interaction with both DNA and nucleosomes. Thus, DNTTIP1 acts as a dimeric chromatin binding module in the HDAC1:MIDEAS corepressor complex.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Histone Deacetylase 1/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Cell Cycle , Co-Repressor Proteins/metabolism , DNA/metabolism , DNA-Binding Proteins , HEK293 Cells , Histone Deacetylase 2/metabolism , Humans , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Tertiary , Transcription Factors
13.
Structure ; 22(11): 1571-81, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25450766

ABSTRACT

Trafficking of leukocytes in immune surveillance and inflammatory responses is activated by chemokines engaging their receptors. Sulfation of tyrosine residues in peptides derived from the eosinophil chemokine receptor CCR3 dramatically enhances binding to cognate chemokines. We report the structural basis of this recognition and affinity enhancement. We describe the structure of a CC chemokine (CCL11/eotaxin-1) bound to a fragment of a chemokine receptor: residues 8­23 of CCR3, including two sulfotyrosine residues. We also show that intact CCR3 is sulfated and sulfation enhances receptor activity. The CCR3 sulfotyrosine residues form hydrophobic, salt bridge and cation-p interactions with residues that are highly conserved in CC chemokines. However, the orientation of the chemokine relative to the receptor N terminus differs substantially from those observed for two CXC chemokines, suggesting that initial binding of the receptor sulfotyrosine residues guides subsequent steps in receptor activation, thereby influencing the receptor conformational changes and signaling.


Subject(s)
Chemokine CCL11/chemistry , Chemokine CCL11/genetics , Receptors, CCR3/chemistry , Receptors, CCR3/metabolism , Tyrosine/analogs & derivatives , Amino Acid Sequence , Binding Sites , Chemokine CCL11/metabolism , Conserved Sequence , HEK293 Cells , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Structure, Secondary , Tyrosine/metabolism
14.
J Vis Exp ; (92): e51897, 2014 Oct 16.
Article in English | MEDLINE | ID: mdl-25349981

ABSTRACT

The expression and purification of large amounts of recombinant protein complexes is an essential requirement for structural biology studies. For over two decades, prokaryotic expression systems such as E. coli have dominated the scientific literature over costly and less efficient eukaryotic cell lines. Despite the clear advantage in terms of yields and costs of expressing recombinant proteins in bacteria, the absence of specific co-factors, chaperones and post-translational modifications may cause loss of function, mis-folding and can disrupt protein-protein interactions of certain eukaryotic multi-subunit complexes, surface receptors and secreted proteins. The use of mammalian cell expression systems can address these drawbacks since they provide a eukaryotic expression environment. However, low protein yields and high costs of such methods have until recently limited their use for structural biology. Here we describe a simple and accessible method for expressing and purifying milligram quantities of protein by performing transient transfections of suspension grown HEK (Human Embryonic Kidney) 293 F cells.


Subject(s)
Biotechnology/methods , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , HEK293 Cells , Humans , Molecular Biology/methods , Recombinant Proteins/genetics , Transfection/methods
15.
Cancer Metastasis Rev ; 33(4): 857-67, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25352341

ABSTRACT

Gene expression is controlled through the recruitment of large coregulator complexes to specific gene loci to regulate chromatin structure by modifying epigenetic marks on DNA and histones. Metastasis-associated protein 1 (MTA1) is an essential component of the nucleosome remodelling and deacetylase (NuRD) complex that acts as a scaffold protein to assemble enzymatic activity and nucleosome targeting proteins. MTA1 consists of four characterised domains, a number of interaction motifs, and regions that are predicted to be intrinsically disordered. The ELM2-SANT domain is one of the best-characterised regions of MTA1, which recruits histone deacetylase 1 (HDAC1) and activates the enzyme in the presence of inositol phosphate. MTA1 is highly upregulated in several types of aggressive tumours and is therefore a possible target for cancer therapy. In this review, we summarise the structure and function of the four domains of MTA1 and discuss the possible functions of less well-characterised regions of the protein.


Subject(s)
Epigenesis, Genetic , Histone Deacetylases/genetics , Neoplasms/genetics , Repressor Proteins/genetics , Transcriptional Activation/genetics , Chromatin Assembly and Disassembly/genetics , Gene Expression Regulation, Neoplastic , Histone Deacetylase 1/genetics , Histone Deacetylases/chemistry , Histones/genetics , Humans , Neoplasm Metastasis , Neoplasms/pathology , Neoplasms/therapy , Repressor Proteins/chemistry , Structure-Activity Relationship , Trans-Activators
16.
Proc Natl Acad Sci U S A ; 111(27): 9840-5, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-24958871

ABSTRACT

Histone deacetylases 1 and 2 (HDAC1/2) form the core catalytic components of corepressor complexes that modulate gene expression. In most cell types, deletion of both Hdac1 and Hdac2 is required to generate a discernible phenotype, suggesting their activity is largely redundant. We have therefore generated an ES cell line in which Hdac1 and Hdac2 can be inactivated simultaneously. Loss of HDAC1/2 resulted in a 60% reduction in total HDAC activity and a loss of cell viability. Cell death is dependent upon cell cycle progression, because differentiated, nonproliferating cells retain their viability. Furthermore, we observe increased mitotic defects, chromatin bridges, and micronuclei, suggesting HDAC1/2 are necessary for accurate chromosome segregation. Consistent with a critical role in the regulation of gene expression, microarray analysis of Hdac1/2-deleted cells reveals 1,708 differentially expressed genes. Significantly for the maintenance of stem cell self-renewal, we detected a reduction in the expression of the pluripotent transcription factors, Oct4, Nanog, Esrrb, and Rex1. HDAC1/2 activity is regulated through binding of an inositol tetraphosphate molecule (IP4) sandwiched between the HDAC and its cognate corepressor. This raises the important question of whether IP4 regulates the activity of the complex in cells. By rescuing the viability of double-knockout cells, we demonstrate for the first time (to our knowledge) that mutations that abolish IP4 binding reduce the activity of HDAC1/2 in vivo. Our data indicate that HDAC1/2 have essential and pleiotropic roles in cellular proliferation and regulate stem cell self-renewal by maintaining expression of key pluripotent transcription factors.


Subject(s)
Cell Division/physiology , Embryonic Stem Cells/enzymology , Histone Deacetylase 1/physiology , Histone Deacetylase 2/physiology , Pluripotent Stem Cells/enzymology , Acetylation , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Histones/metabolism , Mice , Mice, Knockout , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism
17.
J Mol Endocrinol ; 51(3): T23-36, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24203923

ABSTRACT

Nuclear receptors are transcription factors that regulate gene expression through the ligand-controlled recruitment of a diverse group of proteins known as coregulators. Most nuclear receptor coregulators function in large multi-protein complexes that modify chromatin and thereby regulate the transcription of target genes. Structural and functional studies are beginning to reveal how these complexes are assembled bringing together multiple functionalities that mediate: recruitment to specific genomic loci through interaction with transcription factors; recruitment of enzymatic activities that either modify or remodel chromatin and targeting the complexes to their chromatin substrate. These activities are regulated by post-translational modifications, alternative splicing and small signalling molecules. This review focuses on our current understanding of coregulator complexes and aims to highlight the common principles that are beginning to emerge.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Chromatin/metabolism , Humans , Models, Biological , Repressor Proteins/metabolism , Transcription Factors/metabolism
18.
Mol Cell ; 51(1): 57-67, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23791785

ABSTRACT

Class I histone deacetylases (HDAC1, HDAC2, and HDAC3) are recruited by cognate corepressor proteins into specific transcriptional repression complexes that target HDAC activity to chromatin resulting in chromatin condensation and transcriptional silencing. We previously reported the structure of HDAC3 in complex with the SMRT corepressor. This structure revealed the presence of inositol-tetraphosphate [Ins(1,4,5,6)P4] at the interface of the two proteins. It was previously unclear whether the role of Ins(1,4,5,6)P4 is to act as a structural cofactor or a regulator of HDAC3 activity. Here we report the structure of HDAC1 in complex with MTA1 from the NuRD complex. The ELM2-SANT domains from MTA1 wrap completely around HDAC1 occupying both sides of the active site such that the adjacent BAH domain is ideally positioned to recruit nucleosomes to the active site of the enzyme. Functional assays of both the HDAC1 and HDAC3 complexes reveal that Ins(1,4,5,6)P4 is a bona fide conserved regulator of class I HDAC complexes.


Subject(s)
Histone Deacetylase 1/chemistry , Histone Deacetylases/chemistry , Inositol Phosphates/physiology , Repressor Proteins/chemistry , Amino Acid Sequence , Dimerization , HEK293 Cells , Histone Deacetylase 1/metabolism , Histone Deacetylase 1/physiology , Histone Deacetylases/metabolism , Histone Deacetylases/physiology , Humans , Inositol Phosphates/chemistry , Models, Molecular , Molecular Sequence Data , Protein Folding , Protein Structure, Tertiary , Repressor Proteins/metabolism , Repressor Proteins/physiology , Substrate Specificity , Trans-Activators
19.
Genes Dev ; 25(12): 1262-74, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21685362

ABSTRACT

We previously identified the E3 ubiquitin ligase IDOL as a sterol-dependent regulator of the LDL receptor (LDLR). The molecular pathway underlying IDOL action, however, remains to be determined. Here we report the identification and biochemical and structural characterization of an E2-E3 ubiquitin ligase complex for LDLR degradation. We identified the UBE2D family (UBE2D1-4) as E2 partners for IDOL that support both autoubiquitination and IDOL-dependent ubiquitination of the LDLR in a cell-free system. NMR chemical shift mapping and a 2.1 Å crystal structure of the IDOL RING domain-UBE2D1 complex revealed key interactions between the dimeric IDOL protein and the E2 enzyme. Analysis of the IDOL-UBE2D1 interface also defined the stereochemical basis for the selectivity of IDOL for UBE2Ds over other E2 ligases. Structure-based mutations that inhibit IDOL dimerization or IDOL-UBE2D interaction block IDOL-dependent LDLR ubiquitination and degradation. Furthermore, expression of a dominant-negative UBE2D enzyme inhibits the ability of IDOL to degrade the LDLR in cells. These results identify the IDOL-UBE2D complex as an important determinant of LDLR activity, and provide insight into molecular mechanisms underlying the regulation of cholesterol uptake.


Subject(s)
Receptors, LDL/metabolism , Sterols/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Amino Acid Sequence , HEK293 Cells , Humans , Iron/metabolism , Models, Molecular , Protein Structure, Tertiary , Stereoisomerism , Substrate Specificity , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Protein Ligases/chemistry
20.
Biochemistry ; 50(9): 1524-34, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21235238

ABSTRACT

The interactions of chemokines with their G protein-coupled receptors play critical roles in the control of leukocyte trafficking in normal homeostasis and in inflammatory responses. Tyrosine sulfation is a common post-translational modification in the amino-terminal regions of chemokine receptors. However, tyrosine sulfation of chemokine receptors is commonly incomplete or heterogeneous. To investigate the possibility that differential sulfation of two adjacent tyrosine residues could bias the responses of chemokine receptor CCR3 to different chemokines, we have studied the binding of three chemokines (eotaxin-1/CCL11, eotaxin-2/CCL24, and eotaxin-3/CCL26) to an N-terminal CCR3-derived peptide in each of its four possible sulfation states. Whereas the nonsulfated peptide binds to the three chemokines with approximately equal affinity, sulfation of Tyr-16 gives rise to 9-16-fold selectivity for eotaxin-1 over the other two chemokines. Subsequent sulfation of Tyr-17 contributes additively to the affinity for eotaxin-1 and eotaxin-2 but cooperatively to the affinity for eotaxin-3. The doubly sulfated peptide selectively binds to both eotaxin-1 and eotaxin-3 approximately 10-fold more tightly than to eotaxin-2. Nuclear magnetic resonance chemical shift mapping indicates that these variations in affinity probably result from only subtle differences in the chemokine surfaces interacting with these receptor peptides. These data support the proposal that variations in sulfation states or levels may regulate the responsiveness of chemokine receptors to their cognate chemokines.


Subject(s)
Chemokines, CC/metabolism , Chemokines/metabolism , Peptide Fragments/metabolism , Receptors, CCR3/chemistry , Tyrosine/metabolism , Chemokines, CC/chemistry , Peptide Fragments/chemistry , Protein Binding , Receptors, CCR3/metabolism , Sulfates/chemistry , Sulfates/metabolism , Tyrosine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...