Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Brain ; 2024 May 16.
Article in English | MEDLINE | ID: mdl-38753057

ABSTRACT

Deubiquitination is critical for the proper functioning of numerous biological pathways such as DNA repair, cell cycle progression, transcription, signal transduction, and autophagy. Accordingly, pathogenic variants in deubiquitinating enzymes (DUBs) have been implicated in neurodevelopmental disorders (ND) and congenital abnormalities. ATXN7L3 is a component of the DUB module of the SAGA complex, and two other related DUB modules, and serves as an obligate adaptor protein of 3 ubiquitin-specific proteases (USP22, USP27X or USP51). Through exome sequencing and GeneMatching, we identified nine individuals with heterozygous variants in ATXN7L3. The core phenotype included global motor and language developmental delay, hypotonia, and distinctive facial characteristics including hypertelorism, epicanthal folds, blepharoptosis, a small nose and mouth, and low-set posteriorly rotated ears. In order to assess pathogenicity, we investigated the effects of a recurrent nonsense variant [c.340C>T; p.(Arg114Ter)] in fibroblasts of an affected individual. ATXN7L3 protein levels were reduced, and deubiquitylation was impaired, as indicated by an increase in histone H2Bub1 levels. This is consistent with the previous observation of increased H2Bub1 levels in Atxn7l3-null mouse embryos, which have developmental delay and embryonic lethality. In conclusion, we present clinical information and biochemical characterization supporting ATXN7L3 variants in the pathogenesis of a rare syndromic ND.

2.
Cell Rep ; 42(9): 113099, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37682711

ABSTRACT

To understand the function of multisubunit complexes, it is of key importance to uncover the precise mechanisms that guide their assembly. Nascent proteins can find and bind their interaction partners during their translation, leading to co-translational assembly. Here, we demonstrate that the core modules of ATAC (ADA-two-A-containing) and SAGA (Spt-Ada-Gcn5-acetyltransferase), two lysine acetyl transferase-containing transcription co-activator complexes, assemble co-translationally in the cytoplasm of mammalian cells. In addition, a SAGA complex containing all of its modules forms in the cytoplasm and acetylates non-histone proteins. In contrast, ATAC complex subunits cannot be detected in the cytoplasm of mammalian cells. However, an endogenous ATAC complex containing two functional modules forms and functions in the nucleus. Thus, the two related co-activators, ATAC and SAGA, assemble using co-translational pathways, but their subcellular localization, cytoplasmic abundance, and functions are distinct.


Subject(s)
Histone Acetyltransferases , Saccharomyces cerevisiae Proteins , Animals , Histone Acetyltransferases/metabolism , Transcription Factors/metabolism , Chromatin , Cell Nucleus/metabolism , Fungal Proteins , Saccharomyces cerevisiae Proteins/metabolism , Mammals/metabolism
3.
bioRxiv ; 2023 Aug 03.
Article in English | MEDLINE | ID: mdl-37577620

ABSTRACT

To understand the function of multisubunit complexes it is of key importance to uncover the precise mechanisms that guide their assembly. Nascent proteins can find and bind their interaction partners during their translation, leading to co-translational assembly. Here we demonstrate that the core modules of ATAC (ADA-Two-A-Containing) and SAGA (Spt-Ada-Gcn5-acetyltransferase), two lysine acetyl transferase-containing transcription coactivator complexes, assemble co-translationally in the cytoplasm of mammalian cells. In addition, SAGA complex containing all of its modules forms in the cytoplasm and acetylates non-histones proteins. In contrast, fully assembled ATAC complex cannot be detected in the cytoplasm of mammalian cells. However, endogenous ATAC complex containing two functional modules forms and functions in the nucleus. Thus, the two related coactivators, ATAC and SAGA, assemble by using co-translational pathways, but their subcellular localization, cytoplasmic abundance and functions are distinct.

4.
Nat Struct Mol Biol ; 30(8): 1141-1152, 2023 08.
Article in English | MEDLINE | ID: mdl-37386215

ABSTRACT

Large heteromeric multiprotein complexes play pivotal roles at every step of gene expression in eukaryotic cells. Among them, the 20-subunit basal transcription factor TFIID nucleates the RNA polymerase II preinitiation complex at gene promoters. Here, by combining systematic RNA-immunoprecipitation (RIP) experiments, single-molecule imaging, proteomics and structure-function analyses, we show that human TFIID biogenesis occurs co-translationally. We discovered that all protein heterodimerization steps happen during protein synthesis. We identify TAF1-the largest protein in the complex-as a critical factor for TFIID assembly. TAF1 acts as a flexible scaffold that drives the co-translational recruitment of TFIID submodules preassembled in the cytoplasm. Altogether, our data suggest a multistep hierarchical model for TFIID biogenesis that culminates with the co-translational assembly of the complex onto the nascent TAF1 polypeptide. We envision that this assembly strategy could be shared with other large heteromeric protein complexes.


Subject(s)
TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Humans , Cell Nucleus/metabolism , Multiprotein Complexes/chemistry , Promoter Regions, Genetic , TATA-Binding Protein Associated Factors/chemistry , Transcription Factor TFIID/metabolism
5.
bioRxiv ; 2023 Apr 05.
Article in English | MEDLINE | ID: mdl-37066372

ABSTRACT

Large heteromeric multiprotein complexes play pivotal roles at every step of gene expression in eukaryotic cells. Among them, the 20-subunit basal transcription factor TFIID nucleates RNA polymerase II preinitiation complex at gene promoters. Here, by combining systematic RNA-immunoprecipitation (RIP) experiments, single-molecule imaging, proteomics and structure-function analyses, we show that TFIID biogenesis occurs co-translationally. We discovered that all protein heterodimerization steps happen during protein synthesis. We identify TAF1 - the largest protein in the complex - as a critical factor for TFIID assembly. TAF1 acts as a flexible scaffold that drives the co-translational recruitment of TFIID submodules preassembled in the cytoplasm. Altogether, our data suggest a multistep hierarchical model for TFIID biogenesis that culminates with the co-translational assembly of the complex onto the nascent TAF1 polypeptide. We envision that this assembly strategy could be shared with other large heteromeric protein complexes.

6.
Nucleic Acids Res ; 50(14): 7972-7990, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35871303

ABSTRACT

Coactivator complexes regulate chromatin accessibility and transcription. SAGA (Spt-Ada-Gcn5 Acetyltransferase) is an evolutionary conserved coactivator complex. The core module scaffolds the entire SAGA complex and adopts a histone octamer-like structure, which consists of six histone-fold domain (HFD)-containing proteins forming three histone-fold (HF) pairs, to which the double HFD-containing SUPT3H adds one HF pair. Spt3, the yeast ortholog of SUPT3H, interacts genetically and biochemically with the TATA binding protein (TBP) and contributes to global RNA polymerase II (Pol II) transcription. Here we demonstrate that (i) SAGA purified from human U2OS or mouse embryonic stem cells (mESC) can assemble without SUPT3H, (ii) SUPT3H is not essential for mESC survival, but required for their growth and self-renewal, and (iii) the loss of SUPT3H from mammalian cells affects the transcription of only a specific subset of genes. Accordingly, in the absence of SUPT3H no major change in TBP accumulation at gene promoters was observed. Thus, SUPT3H is not required for the assembly of SAGA, TBP recruitment, or overall Pol II transcription, but plays a role in mESC growth and self-renewal. Our data further suggest that yeast and mammalian SAGA complexes contribute to transcription regulation by distinct mechanisms.


Subject(s)
RNA Polymerase II , Trans-Activators , Transcription Factors , Animals , DNA-Binding Proteins/genetics , Histone Acetyltransferases/metabolism , Histones/genetics , Histones/metabolism , Humans , Mice , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic
7.
Blood Adv ; 6(1): 165-180, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34654054

ABSTRACT

Epigenetic histone modifiers are key regulators of cell fate decisions in normal and malignant hematopoiesis. Their enzymatic activities are of particular significance as putative therapeutic targets in leukemia. In contrast, less is known about the contextual role in which those enzymatic activities are exercised and specifically how different macromolecular complexes configure the same enzymatic activity with distinct molecular and cellular consequences. We focus on KAT2A, a lysine acetyltransferase responsible for histone H3 lysine 9 acetylation, which we recently identified as a dependence in acute myeloid leukemia stem cells and that participates in 2 distinct macromolecular complexes: Ada two-A-containing (ATAC) and Spt-Ada-Gcn5-Acetyltransferase (SAGA). Through analysis of human cord blood hematopoietic stem cells and progenitors, and of myeloid leukemia cells, we identify unique respective contributions of the ATAC complex to regulation of biosynthetic activity in undifferentiated self-renewing cells and of the SAGA complex to stabilization or correct progression of cell type-specific programs with putative preservation of cell identity. Cell type and stage-specific dependencies on ATAC and SAGA-regulated programs explain multilevel KAT2A requirements in leukemia and in erythroid lineage specification and development. Importantly, they set a paradigm against which lineage specification and identity can be explored across developmental stem cell systems.


Subject(s)
Histone Acetyltransferases , Leukemia, Myeloid, Acute , Acetylation , Hematopoiesis , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism
8.
J Biol Chem ; 297(5): 101288, 2021 11.
Article in English | MEDLINE | ID: mdl-34634302

ABSTRACT

The human general transcription factor TFIID is composed of the TATA-binding protein (TBP) and 13 TBP-associated factors (TAFs). In eukaryotic cells, TFIID is thought to nucleate RNA polymerase II (Pol II) preinitiation complex formation on all protein coding gene promoters and thus, be crucial for Pol II transcription. TFIID is composed of three lobes, named A, B, and C. A 5TAF core complex can be assembled in vitro constituting a building block for the further assembly of either lobe A or B in TFIID. Structural studies showed that TAF8 forms a histone fold pair with TAF10 in lobe B and participates in connecting lobe B to lobe C. To better understand the role of TAF8 in TFIID, we have investigated the requirement of the different regions of TAF8 for the in vitro assembly of lobe B and C and the importance of certain TAF8 regions for mouse embryonic stem cell (ESC) viability. We have identified a region of TAF8 distinct from the histone fold domain important for assembling with the 5TAF core complex in lobe B. We also delineated four more regions of TAF8 each individually required for interacting with TAF2 in lobe C. Moreover, CRISPR/Cas9-mediated gene editing indicated that the 5TAF core-interacting TAF8 domain and the proline-rich domain of TAF8 that interacts with TAF2 are both required for mouse embryonic stem cell survival. Thus, our study defines distinct TAF8 regions involved in connecting TFIID lobe B to lobe C that appear crucial for TFIID function and consequent ESC survival.


Subject(s)
Mouse Embryonic Stem Cells/metabolism , Protein Folding , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Cell Survival , Humans , Mice , Protein Domains , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
9.
Nat Struct Mol Biol ; 25(12): 1119-1127, 2018 12.
Article in English | MEDLINE | ID: mdl-30510221

ABSTRACT

TFIID is a cornerstone of eukaryotic gene regulation. Distinct TFIID complexes with unique subunit compositions exist and several TFIID subunits are shared with other complexes, thereby conveying precise cellular control of subunit allocation and functional assembly of this essential transcription factor. However, the molecular mechanisms that underlie the regulation of TFIID remain poorly understood. Here we use quantitative proteomics to examine TFIID submodules and assembly mechanisms in human cells. Structural and mutational analysis of the cytoplasmic TAF5-TAF6-TAF9 submodule identified novel interactions that are crucial for TFIID integrity and for allocation of TAF9 to TFIID or the Spt-Ada-Gcn5 acetyltransferase (SAGA) co-activator complex. We discover a key checkpoint function for the chaperonin CCT, which specifically associates with nascent TAF5 for subsequent handover to TAF6-TAF9 and ultimate holo-TFIID formation. Our findings illustrate at the molecular level how multisubunit complexes are generated within the cell via mechanisms that involve checkpoint decisions facilitated by a chaperone.


Subject(s)
Chaperonin Containing TCP-1/physiology , Models, Molecular , Transcription Factor TFIID/chemistry , Chaperonin Containing TCP-1/metabolism , Crystallography, X-Ray , HeLa Cells , Humans , Mass Spectrometry , Protein Domains , TATA-Binding Protein Associated Factors/chemistry , Transcription Factor TFIID/metabolism , Transcription, Genetic
10.
Elife ; 62017 11 07.
Article in English | MEDLINE | ID: mdl-29111974

ABSTRACT

General transcription factor TFIID is a key component of RNA polymerase II transcription initiation. Human TFIID is a megadalton-sized complex comprising TATA-binding protein (TBP) and 13 TBP-associated factors (TAFs). TBP binds to core promoter DNA, recognizing the TATA-box. We identified a ternary complex formed by TBP and the histone fold (HF) domain-containing TFIID subunits TAF11 and TAF13. We demonstrate that TAF11/TAF13 competes for TBP binding with TATA-box DNA, and also with the N-terminal domain of TAF1 previously implicated in TATA-box mimicry. In an integrative approach combining crystal coordinates, biochemical analyses and data from cross-linking mass-spectrometry (CLMS), we determine the architecture of the TAF11/TAF13/TBP complex, revealing TAF11/TAF13 interaction with the DNA binding surface of TBP. We identify a highly conserved C-terminal TBP-interaction domain (CTID) in TAF13, which is essential for supporting cell growth. Our results thus have implications for cellular TFIID assembly and suggest a novel regulatory state for TFIID function.


Subject(s)
TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism , Transcription Factor TFIID/metabolism , Crystallography, X-Ray , DNA/metabolism , Histone Acetyltransferases/metabolism , Humans , Mass Spectrometry , Promoter Regions, Genetic , Protein Binding , Protein Conformation , Protein Interaction Mapping , Transcription Factor TFIID/chemistry
11.
EMBO J ; 36(18): 2710-2725, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28724529

ABSTRACT

SAGA and ATAC are two distinct chromatin modifying co-activator complexes with distinct enzymatic activities involved in RNA polymerase II (Pol II) transcription regulation. To investigate the mobility of co-activator complexes and general transcription factors in live-cell nuclei, we performed imaging experiments based on photobleaching. SAGA and ATAC, but also two general transcription factors (TFIID and TFIIB), were highly dynamic, exhibiting mainly transient associations with chromatin, contrary to Pol II, which formed more stable chromatin interactions. Fluorescence correlation spectroscopy analyses revealed that the mobile pool of the two co-activators, as well as that of TFIID and TFIIB, can be subdivided into "fast" (free) and "slow" (chromatin-interacting) populations. Inhibiting transcription elongation decreased H3K4 trimethylation and reduced the "slow" population of SAGA, ATAC, TFIIB and TFIID In addition, inhibiting histone H3K4 trimethylation also reduced the "slow" populations of SAGA and ATAC Thus, our results demonstrate that in the nuclei of live cells the equilibrium between fast and slow population of SAGA or ATAC complexes is regulated by active transcription via changes in the abundance of H3K4me3 on chromatin.


Subject(s)
Cell Nucleus/enzymology , Transcription Factors/metabolism , Transcription, Genetic , Cell Line , Chromatin/metabolism , Humans , Optical Imaging
12.
Nat Commun ; 7: 13227, 2016 10 31.
Article in English | MEDLINE | ID: mdl-27796307

ABSTRACT

Lysine acetylation is a widespread post-translational modification regulating various biological processes. To characterize cellular functions of the human lysine acetyltransferases KAT2A (GCN5) and KAT2B (PCAF), we determined their acetylome by shotgun proteomics. One of the newly identified KAT2A/2B substrate is polo-like kinase 4 (PLK4), a key regulator of centrosome duplication. We demonstrate that KAT2A/2B acetylate the PLK4 kinase domain on residues K45 and K46. Molecular dynamics modelling suggests that K45/K46 acetylation impairs kinase activity by shifting the kinase to an inactive conformation. Accordingly, PLK4 activity is reduced upon in vitro acetylation of its kinase domain. Moreover, the overexpression of the PLK4 K45R/K46R mutant in cells does not lead to centrosome overamplification, as observed with wild-type PLK4. We also find that impairing KAT2A/2B-acetyltransferase activity results in diminished phosphorylation of PLK4 and in excess centrosome numbers in cells. Overall, our study identifies the global human KAT2A/2B acetylome and uncovers that KAT2A/2B acetylation of PLK4 prevents centrosome amplification.


Subject(s)
Acetylation , Centrosome/metabolism , Histone Acetyltransferases/metabolism , Protein Serine-Threonine Kinases/metabolism , p300-CBP Transcription Factors/metabolism , Amino Acid Motifs , Animals , Cell Cycle/physiology , Centrioles/metabolism , Centrosome/ultrastructure , Drosophila melanogaster , HEK293 Cells , HeLa Cells , Histones/chemistry , Humans , Lysine/chemistry , Mice , Models, Molecular , Molecular Dynamics Simulation , Phosphorylation , Plasmids/metabolism , Point Mutation , Protein Domains , Protein Processing, Post-Translational , RNA, Small Interfering/metabolism , Spindle Apparatus/metabolism
13.
J Biol Chem ; 290(48): 28997-9009, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26468280

ABSTRACT

Histone acetyl transferases (HATs) play a crucial role in eukaryotes by regulating chromatin architecture and locus specific transcription. GCN5 (KAT2A) is a member of the GNAT (Gcn5-related N-acetyltransferase) family of HATs. In metazoans this enzyme is found in two functionally distinct coactivator complexes, SAGA (Spt Ada Gcn5 acetyltransferase) and ATAC (Ada Two A-containing). These two multiprotein complexes comprise complex-specific and shared subunits, which are organized in functional modules. The HAT module of ATAC is composed of GCN5, ADA2a, ADA3, and SGF29, whereas in the SAGA HAT module ADA2b is present instead of ADA2a. To better understand how the activity of human (h) hGCN5 is regulated in the two related, but different, HAT complexes we carried out in vitro HAT assays. We compared the activity of hGCN5 alone with its activity when it was part of purified recombinant hATAC or hSAGA HAT modules or endogenous hATAC or hSAGA complexes using histone tail peptides and full-length histones as substrates. We demonstrated that the subunit environment of the HAT complexes into which GCN5 incorporates determines the enhancement of GCN5 activity. On histone peptides we show that all the tested GCN5-containing complexes acetylate mainly histone H3K14. Our results suggest a stronger influence of ADA2b as compared with ADA2a on the activity of GCN5. However, the lysine acetylation specificity of GCN5 on histone tails or full-length histones was not changed when incorporated in the HAT modules of ATAC or SAGA complexes. Our results thus demonstrate that the catalytic activity of GCN5 is stimulated by subunits of the ADA2a- or ADA2b-containing HAT modules and is further increased by incorporation of the distinct HAT modules in the ATAC or SAGA holo-complexes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Histone Acetyltransferases/metabolism , Multiprotein Complexes/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , DNA-Binding Proteins , Histone Acetyltransferases/genetics , Humans , Multiprotein Complexes/genetics , Sf9 Cells , Spodoptera , Transcription Factors/genetics
14.
Mol Cell Biol ; 35(12): 2103-18, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25870109

ABSTRACT

The ordered assembly of a functional preinitiation complex (PIC), composed of general transcription factors (GTFs), is a prerequisite for the transcription of protein-coding genes by RNA polymerase II. TFIID, comprised of the TATA binding protein (TBP) and 13 TBP-associated factors (TAFs), is the GTF that is thought to recognize the promoter sequences allowing site-specific PIC assembly. Transcriptional cofactors, such as SAGA, are also necessary for tightly regulated transcription initiation. The contribution of the two TAF10-containing complexes (TFIID, SAGA) to erythropoiesis remains elusive. By ablating TAF10 specifically in erythroid cells in vivo, we observed a differentiation block accompanied by deregulated GATA1 target genes, including Gata1 itself, suggesting functional cross talk between GATA1 and TAF10. Additionally, we analyzed by mass spectrometry the composition of TFIID and SAGA complexes in mouse and human cells and found that their global integrity is maintained, with minor changes, during erythroid cell differentiation and development. In agreement with our functional data, we show that TAF10 interacts directly with GATA1 and that TAF10 is enriched on the GATA1 locus in human fetal erythroid cells. Thus, our findings demonstrate a cross talk between canonical TFIID and SAGA complexes and cell-specific transcription activators during development and differentiation.


Subject(s)
Erythroid Cells/cytology , Erythropoiesis , GATA1 Transcription Factor/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Animals , Erythroid Cells/metabolism , GATA1 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Genetic Loci , Humans , Mice , Mice, Knockout , Protein Interaction Mapping , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics
15.
Nat Commun ; 6: 6011, 2015 Jan 14.
Article in English | MEDLINE | ID: mdl-25586196

ABSTRACT

General transcription factor TFIID is a cornerstone of RNA polymerase II transcription initiation in eukaryotic cells. How human TFIID-a megadalton-sized multiprotein complex composed of the TATA-binding protein (TBP) and 13 TBP-associated factors (TAFs)-assembles into a functional transcription factor is poorly understood. Here we describe a heterotrimeric TFIID subcomplex consisting of the TAF2, TAF8 and TAF10 proteins, which assembles in the cytoplasm. Using native mass spectrometry, we define the interactions between the TAFs and uncover a central role for TAF8 in nucleating the complex. X-ray crystallography reveals a non-canonical arrangement of the TAF8-TAF10 histone fold domains. TAF2 binds to multiple motifs within the TAF8 C-terminal region, and these interactions dictate TAF2 incorporation into a core-TFIID complex that exists in the nucleus. Our results provide evidence for a stepwise assembly pathway of nuclear holo-TFIID, regulated by nuclear import of preformed cytoplasmic submodules.


Subject(s)
Cytoplasm/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/chemistry , Amino Acid Motifs , Calorimetry , Cell Nucleus/metabolism , Crystallography, X-Ray , HeLa Cells , Histones/chemistry , Humans , Mass Spectrometry/methods , Models, Molecular , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Surface Plasmon Resonance , Transcription Factor TFIID/metabolism , Transcription Factors/metabolism
16.
Nature ; 493(7434): 699-702, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23292512

ABSTRACT

The initiation of gene transcription by RNA polymerase II is regulated by a plethora of proteins in human cells. The first general transcription factor to bind gene promoters is transcription factor IID (TFIID). TFIID triggers pre-initiation complex formation, functions as a coactivator by interacting with transcriptional activators and reads epigenetic marks. TFIID is a megadalton-sized multiprotein complex composed of TATA-box-binding protein (TBP) and 13 TBP-associated factors (TAFs). Despite its crucial role, the detailed architecture and assembly mechanism of TFIID remain elusive. Histone fold domains are prevalent in TAFs, and histone-like tetramer and octamer structures have been proposed in TFIID. A functional core-TFIID subcomplex was revealed in Drosophila nuclei, consisting of a subset of TAFs (TAF4, TAF5, TAF6, TAF9 and TAF12). These core subunits are thought to be present in two copies in holo-TFIID, in contrast to TBP and other TAFs that are present in a single copy, conveying a transition from symmetry to asymmetry in the TFIID assembly pathway. Here we present the structure of human core-TFIID determined by cryo-electron microscopy at 11.6 Å resolution. Our structure reveals a two-fold symmetric, interlaced architecture, with pronounced protrusions, that accommodates all conserved structural features of the TAFs including the histone folds. We further demonstrate that binding of one TAF8-TAF10 complex breaks the original symmetry of core-TFIID. We propose that the resulting asymmetric structure serves as a functional scaffold to nucleate holo-TFIID assembly, by accreting one copy each of the remaining TAFs and TBP.


Subject(s)
Models, Molecular , Transcription Factor TFIID/chemistry , Cells, Cultured , Cryoelectron Microscopy , HeLa Cells , Humans , Protein Binding , Protein Structure, Tertiary , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism
17.
J Biol Chem ; 287(33): 27580-92, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22696218

ABSTRACT

The general transcription factor TFIID recognizes specifically the core promoter of genes transcribed by eukaryotic RNA polymerase II, nucleating the assembly of the preinitiation complex at the transcription start site. However, the understanding in molecular terms of TFIID assembly and function remains poorly understood. Histone fold motifs have been shown to be extremely important for the heterodimerization of many TFIID subunits. However, these subunits display several evolutionary conserved noncanonical features when compared with histones, including additional regions whose role is unknown. Here we show that the conserved additional C-terminal region of TFIID subunit TAF6 can be divided into two domains: a small middle domain (TAF6M) and a large C-terminal domain (TAF6C). Our crystal structure of the TAF6C domain from Antonospora locustae at 1.9 Å resolution reveals the presence of five conserved HEAT repeats. Based on these data, we designed several mutants that were introduced into full-length human TAF6. Surprisingly, the mutants affect the interaction between TAF6 and TAF9, suggesting that the formation of the complex between these two TFIID subunits do not only depend on their histone fold motifs. In addition, the same mutants affect even more strongly the interaction between TAF6 and TAF9 in the context of a TAF5-TAF6-TAF9 complex. Expression of these mutants in HeLa cells reveals that most of them are unstable, suggesting their poor incorporation within endogenous TFIID. Taken together, our results suggest that the conserved additional domains in histone fold-containing subunits of TFIID and of co-activator SAGA are important for the assembly of these complexes.


Subject(s)
Multiprotein Complexes/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , HeLa Cells , Humans , Multiprotein Complexes/genetics , Mutation , Protein Structure, Tertiary , Repetitive Sequences, Amino Acid , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics
18.
Genes Dev ; 26(8): 797-802, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22465951

ABSTRACT

The linker histone H1 is a key player in chromatin organization, yet our understanding of the regulation of H1 functions by post-translational modifications is very limited. We provide here the first functional characterization of H1 acetylation. We show that H1.4K34 acetylation (H1.4K34ac) is mediated by GCN5 and is preferentially enriched at promoters of active genes, where it stimulates transcription by increasing H1 mobility and recruiting a general transcription factor. H1.4K34ac is dynamic during spermatogenesis and marks undifferentiated cells such as induced pluripotent stem (iPS) cells and testicular germ cell tumors. We propose a model for H1.4K34ac as a novel regulator of chromatin function with a dual role in transcriptional activation.


Subject(s)
Histones/metabolism , Lysine/metabolism , Transcriptional Activation , p300-CBP Transcription Factors/metabolism , Acetylation , Amino Acid Sequence , Cell Cycle/genetics , Gene Expression Regulation, Neoplastic , Histone Acetyltransferases , Histones/genetics , Humans , Lysine/genetics , Male , Molecular Sequence Data , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic , Seminoma/genetics , Seminoma/metabolism , Spermatogenesis/genetics , TATA-Binding Protein Associated Factors/metabolism , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Transcription Factor TFIID/metabolism , Transcription Initiation Site , Up-Regulation
19.
J Biol Chem ; 286(41): 35843-35851, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21852232

ABSTRACT

Global histone H1 phosphorylation correlates with cell cycle progression. However, the function of site-specific H1 variant phosphorylation remains unclear. Our mass spectrometry analysis revealed a novel N-terminal phosphorylation of the major H1 variant H1.4 at serine 35 (H1.4S35ph), which accumulates at mitosis immediately after H3 phosphorylation at serine 10. Protein kinase A (PKA) was found to be a kinase for H1.4S35. Importantly, Ser-35-phosphorylated H1.4 dissociates from mitotic chromatin. Moreover, H1.4S35A substitution mutant cannot efficiently rescue the mitotic defect following H1.4 depletion, and inhibition of PKA activity increases the mitotic chromatin compaction depending on H1.4. Our results not only indicate that PKA-mediated H1.4S35 phosphorylation dissociates H1.4 from mitotic chromatin but also suggest that this phosphorylation is necessary for specific mitotic functions.


Subject(s)
Chromatin/metabolism , Chromosomes, Human/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Histones/metabolism , Mitosis/physiology , Amino Acid Substitution , Chromatin/genetics , Chromosomes, Human/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , HeLa Cells , Histones/genetics , Humans , Mutation, Missense , Phosphorylation/physiology , Serine/genetics , Serine/metabolism
20.
J Cell Sci ; 123(Pt 15): 2663-71, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20627952

ABSTRACT

Gene transcription in mammalian cells is a dynamic process involving regulated assembly of transcription complexes on chromatin in which the TATA-binding protein (TBP) plays a central role. Here, we investigate the dynamic behaviour of TBP by a combination of fluorescence recovery after photobleaching (FRAP) and biochemical assays using human cell lines of different origin. The majority of nucleoplasmic TBP and other TFIID subunits associate with chromatin in a highly dynamic manner. TBP dynamics are regulated by the joint action of the SNF2-related BTAF1 protein and the NC2 complex. Strikingly, both BTAF1 and NC2 predominantly affect TBP dissociation rates, leaving the association rate unchanged. Chromatin immunoprecipitation shows that BTAF1 negatively regulates TBP and NC2 binding to active promoters. Our results support a model for a BTAF1-mediated release of TBP-NC2 complexes from chromatin.


Subject(s)
Chromatin/metabolism , TATA-Box Binding Protein/metabolism , Cell Line , Cell Line, Tumor , Chromatin/genetics , Chromatin Immunoprecipitation , Chromatography, Gel , Fluorescence Recovery After Photobleaching , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , TATA-Box Binding Protein/genetics , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...