Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Mol Cell ; 84(9): 1699-1710.e6, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38604172

ABSTRACT

The transition from transcription initiation to elongation is highly regulated in human cells but remains incompletely understood at the structural level. In particular, it is unclear how interactions between RNA polymerase II (RNA Pol II) and initiation factors are broken to enable promoter escape. Here, we reconstitute RNA Pol II promoter escape in vitro and determine high-resolution structures of initially transcribing complexes containing 8-, 10-, and 12-nt ordered RNAs and two elongation complexes containing 14-nt RNAs. We suggest that promoter escape occurs in three major steps. First, the growing RNA displaces the B-reader element of the initiation factor TFIIB without evicting TFIIB. Second, the rewinding of the transcription bubble coincides with the eviction of TFIIA, TFIIB, and TBP. Third, the binding of DSIF and NELF facilitates TFIIE and TFIIH dissociation, establishing the paused elongation complex. This three-step model for promoter escape fills a gap in our understanding of the initiation-elongation transition of RNA Pol II transcription.


Subject(s)
Phosphoproteins , Promoter Regions, Genetic , RNA Polymerase II , TATA-Box Binding Protein , Transcription Factor TFIIB , Transcription Factors , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Humans , Transcription Factor TFIIB/metabolism , Transcription Factor TFIIB/genetics , TATA-Box Binding Protein/metabolism , TATA-Box Binding Protein/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Initiation, Genetic , Transcription Factor TFIIH/metabolism , Transcription Factor TFIIH/genetics , Transcription Factor TFIIH/chemistry , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Protein Binding , Transcription Factor TFIIA/metabolism , Transcription Factor TFIIA/genetics , Transcription, Genetic , Transcription Elongation, Genetic , RNA/metabolism , RNA/genetics , Transcription Factors, TFII/metabolism , Transcription Factors, TFII/genetics
2.
EMBO J ; 42(10): e113519, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37013908

ABSTRACT

Recruitment of RNA polymerase II (Pol II) to promoters is essential for transcription. Despite conflicting evidence, the Pol II preinitiation complex (PIC) is often thought to have a uniform composition and to assemble at all promoters via an identical mechanism. Here, using Drosophila melanogaster S2 cells as a model, we demonstrate that different promoter classes function via distinct PICs. Promoter DNA of developmentally regulated genes readily associates with the canonical Pol II PIC, whereas housekeeping promoters do not, and instead recruit other factors such as DREF. Consistently, TBP and DREF are differentially required by distinct promoter types. TBP and its paralog TRF2 also function at different promoter types in a partially redundant manner. In contrast, TFIIA is required at all promoters, and we identify factors that can recruit and/or stabilize TFIIA at housekeeping promoters and activate transcription. Promoter activation by tethering these factors is sufficient to induce the dispersed transcription initiation patterns characteristic of housekeeping promoters. Thus, different promoter classes utilize distinct mechanisms of transcription initiation, which translate into different focused versus dispersed initiation patterns.


Subject(s)
Drosophila Proteins , Transcription Factors , Animals , Transcription Factors/genetics , Transcription Factor TFIIA/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Transcription, Genetic , Drosophila Proteins/genetics
3.
Hum Mol Genet ; 31(18): 3083-3094, 2022 09 10.
Article in English | MEDLINE | ID: mdl-35512351

ABSTRACT

BACKGROUND: TASP1 encodes an endopeptidase activating histone methyltransferases of the KMT2 family. Homozygous loss-of-function variants in TASP1 have recently been associated with Suleiman-El-Hattab syndrome. We report six individuals with Suleiman-El-Hattab syndrome and provide functional characterization of this novel histone modification disorder in a multi-omics approach. METHODS: Chromosomal microarray/exome sequencing in all individuals. Western blotting from fibroblasts in two individuals. RNA sequencing and proteomics from fibroblasts in one individual. Methylome analysis from blood in two individuals. Knock-out of tasp1 orthologue in zebrafish and phenotyping. RESULTS: All individuals had biallelic TASP1 loss-of-function variants and a phenotype including developmental delay, multiple congenital anomalies (including cardiovascular and posterior fossa malformations), a distinct facial appearance and happy demeanor. Western blot revealed absence of TASP1. RNA sequencing/proteomics showed HOX gene downregulation (HOXA4, HOXA7, HOXA1 and HOXB2) and dysregulation of transcription factor TFIIA. A distinct methylation profile intermediate between control and Kabuki syndrome (KMT2D) profiles could be produced. Zebrafish tasp1 knock-out revealed smaller head size and abnormal cranial cartilage formation in tasp1 crispants. CONCLUSION: This work further delineates Suleiman-El-Hattab syndrome, a recognizable neurodevelopmental syndrome. Possible downstream mechanisms of TASP1 deficiency include perturbed HOX gene expression and dysregulated TFIIA complex. Methylation pattern suggests that Suleiman-El-Hattab syndrome can be categorized into the group of histone modification disorders including Wiedemann-Steiner and Kabuki syndrome.


Subject(s)
Histone Code , Zebrafish , Abnormalities, Multiple , Animals , Endopeptidases/genetics , Face/abnormalities , Hematologic Diseases , Histone Methyltransferases/genetics , Phenotype , Transcription Factor TFIIA/genetics , Vestibular Diseases , Zebrafish/genetics
4.
PLoS Pathog ; 18(9): e1010850, 2022 09.
Article in English | MEDLINE | ID: mdl-36121876

ABSTRACT

Viroids, a fascinating group of plant pathogens, are subviral agents composed of single-stranded circular noncoding RNAs. It is well-known that nuclear-replicating viroids exploit host DNA-dependent RNA polymerase II (Pol II) activity for transcription from circular RNA genome to minus-strand intermediates, a classic example illustrating the intrinsic RNA-dependent RNA polymerase activity of Pol II. The mechanism for Pol II to accept single-stranded RNAs as templates remains poorly understood. Here, we reconstituted a robust in vitro transcription system and demonstrated that Pol II also accepts minus-strand viroid RNA template to generate plus-strand RNAs. Further, we purified the Pol II complex on RNA templates for nano-liquid chromatography-tandem mass spectrometry analysis and identified a remodeled Pol II missing Rpb4, Rpb5, Rpb6, Rpb7, and Rpb9, contrasting to the canonical 12-subunit Pol II or the 10-subunit Pol II core on DNA templates. Interestingly, the absence of Rpb9, which is responsible for Pol II fidelity, explains the higher mutation rate of viroids in comparison to cellular transcripts. This remodeled Pol II is active for transcription with the aid of TFIIIA-7ZF and appears not to require other canonical general transcription factors (such as TFIIA, TFIIB, TFIID, TFIIE, TFIIF, TFIIH, and TFIIS), suggesting a distinct mechanism/machinery for viroid RNA-templated transcription. Transcription elongation factors, such as FACT complex, PAF1 complex, and SPT6, were also absent in the reconstituted transcription complex. Further analyses of the critical zinc finger domains in TFIIIA-7ZF revealed the first three zinc finger domains pivotal for RNA template binding. Collectively, our data illustrated a distinct organization of Pol II complex on viroid RNA templates, providing new insights into viroid replication, the evolution of transcription machinery, as well as the mechanism of RNA-templated transcription.


Subject(s)
Transcription Factors, General , Viroids , DNA/metabolism , RNA/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Circular/genetics , RNA-Dependent RNA Polymerase/genetics , Transcription Factor TFIIA/genetics , Transcription Factor TFIIA/metabolism , Transcription Factor TFIIB/genetics , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Transcription Factor TFIIIA/metabolism , Transcription Factors, General/genetics , Transcription Factors, General/metabolism , Transcription, Genetic , Viroids/genetics , Viroids/metabolism
5.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article in English | MEDLINE | ID: mdl-34301908

ABSTRACT

The TATA box-binding protein (TBP) is highly conserved throughout eukaryotes and plays a central role in the assembly of the transcription preinitiation complex (PIC) at gene promoters. TBP binds and bends DNA, and directs adjacent binding of the transcription factors TFIIA and TFIIB for PIC assembly. Here, we show that yeast TBP can bind to a nucleosome containing the Widom-601 sequence and that TBP-nucleosome binding is stabilized by TFIIA. We determine three cryo-electron microscopy (cryo-EM) structures of TBP-nucleosome complexes, two of them containing also TFIIA. TBP can bind to superhelical location (SHL) -6, which contains a TATA-like sequence, but also to SHL +2, which is GC-rich. Whereas binding to SHL -6 can occur in the absence of TFIIA, binding to SHL +2 is only observed in the presence of TFIIA and goes along with detachment of upstream terminal DNA from the histone octamer. TBP-nucleosome complexes are sterically incompatible with PIC assembly, explaining why a promoter nucleosome generally impairs transcription and must be moved before initiation can occur.


Subject(s)
DNA/metabolism , Nucleosomes/metabolism , TATA-Box Binding Protein/metabolism , Transcription Factor TFIIA/metabolism , Transcription Factor TFIIB/metabolism , DNA/chemistry , Models, Molecular , Nucleosomes/chemistry , Protein Binding , Protein Conformation , Saccharomyces cerevisiae/metabolism , Transcription Factor TFIIA/genetics , Transcription Factor TFIIB/genetics
6.
PLoS Genet ; 17(1): e1009316, 2021 01.
Article in English | MEDLINE | ID: mdl-33493197

ABSTRACT

Citrus canker caused by Xanthomonas citri subsp. citri (Xcc) is one of the most devastating diseases in citrus industry worldwide. Most citrus cultivars such as sweet orange are susceptible to canker disease. Here, we utilized wild citrus to identify canker-resistant germplasms, and found that Atalantia buxifolia, a primitive (distant-wild) citrus, exhibited remarkable resistance to canker disease. Although the susceptibility gene LATERAL ORGAN BOUNDARIES 1 (LOB1) could also be induced in Atalantia after canker infection, the induction extent was far lower than that in sweet orange. In addition, three of amino acids encoded by transcription factor TFIIAγ in Atalantia (AbTFIIAγ) exhibited difference from those in sweet orange (CsTFIIAγ) which could stabilize the interaction between effector PthA4 and effector binding element (EBE) of LOB1 promoter. The mutation of AbTFIIAγ did not change its interaction with transcription factor binding motifs (TFBs). However, the AbTFIIAγ could hardly support the LOB1 expression induced by the PthA4. In addition, the activity of AbLOB1 promoter was significantly lower than that of CsLOB1 under the induction by PthA4. Our results demonstrate that natural variations of AbTFIIAγ and effector binding element (EBE) in the AbLOB1 promoter are crucial for the canker disease resistance of Atalantia. The natural mutations of AbTFIIAγ gene and AbLOB1 promoter in Atalantia provide candidate targets for improving the resistance to citrus canker disease.


Subject(s)
Disease Resistance/genetics , Plant Diseases/genetics , Rutaceae/genetics , Transcription Factor TFIIA/genetics , Citrus/genetics , Citrus/growth & development , Citrus/microbiology , Gene Expression Regulation, Plant , Mutation/genetics , Plant Diseases/microbiology , Plant Proteins/genetics , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Rutaceae/growth & development , Rutaceae/microbiology , Xanthomonas/genetics , Xanthomonas/pathogenicity
7.
Curr Genet ; 69(4-6): 289-300, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37947853

ABSTRACT

Binding of general transcription factors TFIID and TFIIA to basal promoters is rate-limiting for transcriptional initiation of eukaryotic protein-coding genes. Consequently, activator proteins interacting with subunits of TFIID and/or TFIIA can drastically increase the rate of initiation events. Yeast transcriptional activator Ino2 interacts with several Taf subunits of TFIID, among them the multifunctional Taf1 protein. In contrast to mammalian Taf1, yeast Taf1 lacks bromodomains which are instead encoded by separate proteins Bdf1 and Bdf2. In this work, we show that Bdf1 not only binds to acetylated histone H4 but can also be recruited by Ino2 and unrelated activators such as Gal4, Rap1, Leu3 and Flo8. An activator-binding domain was mapped in the N-terminus of Bdf1. Subunits Toa1 and Toa2 of yeast TFIIA directly contact sequences of basal promoters and TFIID subunit TBP but may also mediate the influence of activators. Indeed, Ino2 efficiently binds to two separate structural domains of Toa1, specifically with its N-terminal four-helix bundle structure required for dimerization with Toa2 and its C-terminal ß-barrel domain contacting TBP and sequences of the TATA element. These findings complete the functional analysis of yeast general transcription factors Bdf1 and Toa1 and identify them as targets of activator proteins.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Bromodomain Containing Proteins , Phospholipids , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription Factor TFIIA , Transcription Factors , Phospholipids/biosynthesis , Phospholipids/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Transcription Factor TFIIA/genetics , Transcription Factor TFIIA/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bromodomain Containing Proteins/genetics , Bromodomain Containing Proteins/metabolism
8.
Mol Cell ; 51(4): 480-92, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23973376

ABSTRACT

To bridge the gap between in vivo and in vitro molecular mechanisms, we dissected the transcriptional control of the endogenous histone gene cluster (His-C) by single-cell imaging. A combination of quantitative immunofluorescence, RNA FISH, and FRAP measurements revealed atypical promoter recognition complexes and differential transcription kinetics directing histone H1 versus core histone gene expression. While H1 is transcribed throughout S phase, core histones are only transcribed in a short pulse during early S phase. Surprisingly, no TFIIB or TFIID was detectable or functionally required at the initiation complexes of these promoters. Instead, a highly stable, preloaded TBP/TFIIA "pioneer" complex primes the rapid initiation of His-C transcription during early S phase. These results provide mechanistic insights for the role of gene-specific core promoter factors and implications for cell cycle-regulated gene expression.


Subject(s)
Drosophila melanogaster/genetics , Gene Expression Regulation , Histones/genetics , Multigene Family , S Phase/physiology , TATA-Box Binding Protein/metabolism , Transcription, Genetic , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Fluorescent Antibody Technique , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histones/metabolism , Image Processing, Computer-Assisted , Kinetics , Promoter Regions, Genetic/genetics , TATA Box/genetics , TATA-Box Binding Protein/genetics , Telomeric Repeat Binding Protein 2/metabolism , Transcription Factor TFIIA/genetics , Transcription Factor TFIIA/metabolism , Transcription Factor TFIIB/genetics , Transcription Factor TFIIB/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Transcription Initiation, Genetic
9.
J Biol Chem ; 292(28): 11873-11885, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28539359

ABSTRACT

RNA polymerase II (pol II) is required for the transcription of all protein-coding genes and as such represents a major enzyme whose activity is tightly regulated. Transcriptional initiation therefore requires numerous general transcriptional factors and cofactors that associate with pol II at the core promoter to form a pre-initiation complex. Transcription factor IIA (TFIIA) is a general cofactor that binds TFIID and stabilizes the TFIID-DNA complex during transcription initiation. Previous studies showed that TFIIA can make contact with the DNA sequence upstream or downstream of the TATA box, and that the region bound by TFIIA could overlap with the elements recognized by another factor, TFIIB, at adenovirus major late core promoter. Whether core promoters contain a DNA motif recognized by TFIIA remains unknown. Here we have identified a core promoter element upstream of the TATA box that is recognized by TFIIA. A search of the human promoter database revealed that many natural promoters contain a TFIIA recognition element (IIARE). We show that the IIARE enhances TFIIA-promoter binding and enhances the activity of TATA-containing promoters, but represses or activates promoters that lack a TATA box. Chromatin immunoprecipitation assays revealed that the IIARE activates transcription by increasing the recruitment of pol II, TFIIA, TAF4, and P300 at TATA-dependent promoters. These findings extend our understanding of the role of TFIIA in transcription, and provide new insights into the regulatory mechanism of core promoter elements in gene transcription by pol II.


Subject(s)
Gene Expression Regulation , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Response Elements , TATA Box , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIIA/metabolism , Transcription Factor TFIID/metabolism , Binding Sites , Chromatin Immunoprecipitation , DNA, Recombinant , E1A-Associated p300 Protein/chemistry , E1A-Associated p300 Protein/metabolism , Genes, Reporter , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Mutation , Nucleotide Motifs , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Polymerase II/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , TATA-Binding Protein Associated Factors/chemistry , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Transcription Factor TFIIA/chemistry , Transcription Factor TFIIA/genetics , Transcription Factor TFIID/chemistry , Upstream Stimulatory Factors/chemistry , Upstream Stimulatory Factors/genetics , Upstream Stimulatory Factors/metabolism
10.
Protein Expr Purif ; 133: 50-56, 2017 05.
Article in English | MEDLINE | ID: mdl-28259734

ABSTRACT

In vitro transcription systems have been utilized to elucidate detailed mechanisms of transcription. Purified RNA polymerase II (pol II) and general transcription factors (GTFs) are required for the in vitro reconstitution of eukaryotic transcription systems. Among GTFs, TFIID and TFIIA play critical roles in the early stage of transcription initiation; TFIID first binds to the DNA in transcription initiation and TFIIA regulates TFIID's DNA binding activity. Despite the important roles of TFIIA, the time-consuming steps required to purify it, such as denaturing and refolding, have hampered the preparation of in vitro transcription systems. Here, we report an improved method for soluble expression and rapid purification of yeast TFIIA. The subunits of TFIIA, TOA1 and TOA2, were bacterially expressed as fusion proteins in soluble form, then processed by the PreScission protease and co-purified. TFIIA's heterodimer formation was confirmed by size exclusion chromatography-multiangle light scattering (SEC-MALS). The hydrodynamic radius (Rh) and radius of gyration (Rg) were measured by dynamic light scattering (DLS) and small-angle X-ray scattering (SAXS), respectively. The Rg/Rh value implied that the intrinsically disordered region of TOA1 might not have an extended structure in solution. Our improved method provides highly purified TFIIA of sufficient quality for biochemical, biophysical, and structural analyses of eukaryotic transcription systems.


Subject(s)
Escherichia coli/metabolism , Protein Multimerization , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Transcription Factor TFIIA , Escherichia coli/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Solubility , Transcription Factor TFIIA/biosynthesis , Transcription Factor TFIIA/chemistry , Transcription Factor TFIIA/genetics
11.
J Biol Chem ; 288(32): 23273-94, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23814059

ABSTRACT

We have previously shown that yeast TFIID provides coactivator function on the promoters of ribosomal protein-encoding genes (RPGs) by making direct contact with the transactivator repressor activator protein 1 (Rap1). Further, our structural studies of assemblies generated with purified Rap1, TFIID, and TFIIA on RPG enhancer-promoter DNA indicate that Rap1-TFIID interaction induces dramatic conformational rearrangements of enhancer-promoter DNA and TFIID-bound TFIIA. These data indicate a previously unknown yet critical role for yeast TFIIA in the integration of activator-TFIID contacts with promoter conformation and downstream preinitiation complex formation and/or function. Here we describe the use of systematic mutagenesis to define how specific TFIIA contacts contribute to these processes. We have verified that TFIIA is required for RPG transcription in vivo and in vitro, consistent with the existence of a critical Rap1-TFIIA-TFIID interaction network. We also identified essential points of contact for TFIIA and Rap1 within the Rap1 binding domain of the Taf4 subunit of TFIID. These data suggest a mechanism for how interactions between TFIID, TFIIA, and Rap1 contribute to the high rate of transcription initiation seen on RPGs in vivo.


Subject(s)
Enhancer Elements, Genetic/physiology , Ribosomal Proteins/biosynthesis , Saccharomyces cerevisiae/metabolism , Transcription Factor TFIIA/metabolism , Transcription Factor TFIID/metabolism , Transcription Initiation, Genetic/physiology , Mutagenesis , Protein Binding/physiology , Ribosomal Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Shelterin Complex , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Transcription Factor TFIIA/genetics , Transcription Factor TFIID/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Lab Invest ; 94(8): 906-16, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24933421

ABSTRACT

Although many studies have suggested that estrogen prevents postmenopausal bone loss partially due to its anti-apoptosis effects in osteoblasts, the underlying mechanism has not been fully elucidated. In the present study, we found that 17ß-estradiol (17ß-E2), one of the primary estrogens, inhibited endoplasmic reticulum (ER) stress-induced apoptosis in MC3T3-E1 cells and primary osteoblasts. Interestingly, 17ß-E2-promoted Grp78 induction, but not CHOP induction in response to ER stress. We further confirmed that Grp78-specific siRNA reversed the inhibition of 17ß-E2 on ER stress-induced apoptosis by activating caspase-12 and caspase-3. Moreover, we found that 17ß-E2 markedly increased the phosphorylated TFII-I levels and nuclear localization of TFII-I in ER stress conditions. 17ß-E2 stimulated Grp78 promoter activity in a dose-dependent manner in the presence of TFII-I and enhanced the binding of TFII-I to the Grp78 promoter. In addition, 17ß-E2 notably increased phosphorylated ERK1/2 levels and Ras kinase activity in MC3T3-E1 cells. The ERK1/2 activity-specific inhibitor U0126 remarkably blocked 17ß-E2-induced TFII-I phosphorylation and Grp78 expression in response to ER stress. Together, 17ß-E2 protected MC3T3-E1 cells against ER stress-induced apoptosis by promoting Ras-ERK1/2-TFII-I signaling pathway-dependent Grp78 induction.


Subject(s)
Apoptosis/drug effects , Bone Density Conservation Agents/pharmacology , Endoplasmic Reticulum Stress/drug effects , Estradiol/pharmacology , Heat-Shock Proteins/agonists , Osteoblasts/drug effects , Transcription Factor TFIIA/agonists , Animals , Animals, Newborn , Bone Density Conservation Agents/chemistry , Bone Density Conservation Agents/therapeutic use , Cell Line , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Estradiol/chemistry , Estradiol/therapeutic use , Estrogens/chemistry , Estrogens/pharmacology , Estrogens/therapeutic use , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoporosis/chemically induced , Osteoporosis/metabolism , Osteoporosis/prevention & control , Phosphorylation/drug effects , Promoter Regions, Genetic/drug effects , Protein Processing, Post-Translational/drug effects , RNA Interference , Random Allocation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transcription Factor TFIIA/genetics , Transcription Factor TFIIA/metabolism
13.
Gene ; 889: 147800, 2023 Dec 30.
Article in English | MEDLINE | ID: mdl-37716588

ABSTRACT

In eukaryotes, TATA-binding protein (TBP) occupancy of the core promoter globally correlates with transcriptional activity of class II genes. Elucidating how TBP is delivered to the TATA box or TATA-like element is crucial to understand the mechanisms of transcriptional regulation. A previous study demonstrated that the inhibitory DNA binding (IDB) surface of human TBP plays an indispensable role during the two-step formation of the TBP-TATA complex, first assuming an unstable and unbent intermediate conformation, and subsequently converting slowly to a stable and bent conformation. The DNA binding property of TBP is altered by physical contact of this surface with TBP regulators. In the present study, we examined whether the interaction between Taf1 N-terminal domain 2 (TAND2) and the IDB surface affected DNA binding property of yeast TBP by exploiting TAND2-fused TBP derivatives. TAND2 promoted formation of two distinct types of TBP-TATA complexes, which we arbitrarily designated as complex I and II. While complex I was stable and similar to the well-characterized original TBP-TATA complex, complex II was unstable and moved along DNA. Removal of TAND2 from TBP after complex formation revealed that continuous contact of TAND2 with the IDB surface was required for formation of complex II but not complex I. Further, TFIIA could be incorporated into the complex of TAND2-fused TBP and the TATA box, which was dependent on the amino-terminal non-conserved region of TBP, implying that this region could facilitate the exchange between TAND2 and TFIIA on the IDB surface. Collectively, these findings provide novel insights into the mechanism by which TBP is relieved from the interaction with TAND to bind the TATA box or TATA-like element within promoter-bound TFIID.


Subject(s)
Gene Expression Regulation , Transcription Factor TFIID , Humans , Transcription Factor TFIID/genetics , Transcription Factor TFIIA/genetics , Transcription Factor TFIIA/metabolism , TATA-Box Binding Protein/chemistry , DNA/metabolism , Saccharomyces cerevisiae/genetics , TATA Box/genetics
14.
Gene ; 833: 146581, 2022 Jul 30.
Article in English | MEDLINE | ID: mdl-35597524

ABSTRACT

The assembly of transcription complexes on eukaryotic promoters involves a series of steps, including chromatin remodeling, recruitment of TATA-binding protein (TBP)-containing complexes, the RNA polymerase II holoenzyme, and additional basal transcription factors. This review describes the transcriptional regulation by TBP and its corresponding homologs that constitute the TBP family and their interactions with promoter DNA. The C-terminal core domain of TBP is highly conserved and contains two structural repeats that fold into a saddle-like structure, essential for the interaction with the TATA-box on DNA. Based on the TBP C-terminal core domain similarity, three TBP-related factors (TRFs) or TBP-like factors (TBPLs) have been discovered in metazoans, TRF1, TBPL1, and TBPL2. TBP is autoregulated, and once bound to DNA, repressors such as Mot1 induce TBP to dissociate, while other factors such as NC2 and the NOT complex convert the active TBP/DNA complex into inactive, negatively regulating TBP. TFIIA antagonizes the TBP repressors but may be effective only in conjunction with the RNA polymerase II holoenzyme recruitment to the promoter by promoter-bound activators. TRF1 has been discovered inDrosophila melanogasterandAnophelesbut found absent in vertebrates and yeast. TBPL1 cannot bind to the TATA-box; instead, TBPL1 prefers binding to TATA-less promoters. However, TBPL1 shows a stronger association with TFIIA than TBP. The TCT core promoter element is present in most ribosomal protein genes inDrosophilaand humans, and TBPL1 is required for the transcription of these genes. TBP directly participates in the DNA repair mechanism, and TBPL1 mediates cell cycle arrest and apoptosis. TBPL2 is closely related to its TBP paralog, showing 95% sequence similarity with the TBP core domain. Like TBP, TBPL2 also binds to the TATA-box and shows interactions with TFIIA, TFIIB, and other basal transcription factors. Despite these advances, much remains to be explored in this family of transcription factors.


Subject(s)
RNA Polymerase II , TATA-Box Binding Protein , Transcription Factors , Transcription, Genetic , Adenosine Triphosphatases/genetics , Animals , DNA/genetics , Drosophila , Holoenzymes/genetics , Holoenzymes/metabolism , Humans , Nuclear Proteins/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins , TATA Box/genetics , TATA Box Binding Protein-Like Proteins/chemistry , TATA Box Binding Protein-Like Proteins/genetics , TATA Box Binding Protein-Like Proteins/metabolism , TATA-Binding Protein Associated Factors , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Transcription Factor TFIIA/genetics , Transcription Factor TFIIA/metabolism , Transcription Factors/genetics
15.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34156981

ABSTRACT

Taspase1, a highly conserved threonine protease encoded by TASP1, cleaves nuclear histone-modifying factors and basal transcription regulators to orchestrate diverse transcription programs. Hereditary loss-of-function mutation of TASP1 has recently been reported in humans as resulting in an anomaly complex syndrome, which manifests with hematological, facial, and skeletal abnormalities. Here, we demonstrate that Taspase1-mediated cleavage of TFIIAα-ß, rather than of MLL1 or MLL2, in mouse embryos was required for proper fetal liver hematopoiesis and correct segmental identities of the axial skeleton. Homozygous genetic deletion of Taspase1 disrupted embryonic hematopoietic stem cell self-renewal and quiescence states and axial skeleton fates. Strikingly, mice carrying knockin noncleavable mutations of TFIIAα-ß, a well-characterized basal transcription factor, displayed more pronounced fetal liver and axial skeleton defects than those with noncleavable MLL1 and MLL2, 2 trithorax group histone H3 trimethyl transferases. Our study offers molecular insights into a syndrome in humans that results from loss of TASP1 and describes an unexpected role of TFIIAα-ß cleavage in embryonic cell fate decisions.


Subject(s)
Abnormalities, Multiple/genetics , Endopeptidases , Fetal Development/physiology , Transcription Factor TFIIA/genetics , Animals , Embryo, Mammalian , Endopeptidases/genetics , Endopeptidases/metabolism , Hematopoietic Stem Cells , Histone Code/genetics , Histone-Lysine N-Methyltransferase/metabolism , Mice , Mice, Knockout , Mutation , Myeloid-Lymphoid Leukemia Protein/metabolism , RNA Cleavage , Stem Cell Transplantation
16.
Biochim Biophys Acta ; 1789(3): 161-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19437618

ABSTRACT

Through the reductive divisions of meiosis, sexually reproducing organisms have gained the ability to produce specialized haploid cells called germ cells that fuse to establish the diploid genome of the resulting progeny. The totipotent nature of these germ cells is highlighted by their ability to provide a single fertilized egg cell with all the genetic information necessary to develop the complete repertoire of cell types of the future organism. Thus, the production of these germ cells must be tightly regulated to ensure the continued success of the germ line in future generations. One surprising germ cell development mechanism utilizes variation of the global transcriptional machinery, such as TFIID and TFIIA. Like histone variation, general transcription factor variation serves to produce gonadal-restricted or -enriched expression of selective transcriptional regulatory factors required for establishing and/or maintaining the germ line of diverse organisms. This strategy is observed among invertebrates and vertebrates, and perhaps plants, suggesting that a common theme in germ cell evolution is the diversification of selective promoter initiation factors to regulate critical gonadal-specific programs of gene expression required for sexual reproduction. This review discusses the identification and characterization of a subset of these specialized general transcription factors in diverse organisms that share a common goal of germ line regulation through transcriptional control at its most fundamental level.


Subject(s)
Gene Expression Regulation, Developmental , Germ Cells/metabolism , Transcription Factors/metabolism , Animals , Transcription Factor TFIIA/genetics , Transcription Factor TFIIA/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Transcription Factors/genetics
17.
BMC Evol Biol ; 10: 128, 2010 May 04.
Article in English | MEDLINE | ID: mdl-20438643

ABSTRACT

BACKGROUND: Gene duplication provides raw genetic materials for evolutionary novelty and adaptation. The evolutionary fate of duplicated transcription factor genes is less studied although transcription factor gene plays important roles in many biological processes. TFIIAgamma is a small subunit of TFIIA that is one of general transcription factors required by RNA polymerase II. Previous studies identified two TFIIAgamma-like genes in rice genome and found that these genes either conferred resistance to rice bacterial blight or could be induced by pathogen invasion, raising the question as to their functional divergence and evolutionary fates after gene duplication. RESULTS: We reconstructed the evolutionary history of the TFIIAgamma genes from main lineages of angiosperms and demonstrated that two TFIIAgamma genes (TFIIAgamma1 and TFIIAgamma5) arose from a whole genome duplication that happened in the common ancestor of grasses. Likelihood-based analyses with branch, codon, and branch-site models showed no evidence of positive selection but a signature of relaxed selective constraint after the TFIIAgamma duplication. In particular, we found that the nonsynonymous/synonymous rate ratio (omega = dN/dS) of the TFIIAgamma1 sequences was two times higher than that of TFIIAgamma5 sequences, indicating highly asymmetric rates of protein evolution in rice tribe and its relatives, with an accelerated rate of TFIIAgamma1 gene. Our expression data and EST database search further indicated that after whole genome duplication, the expression of TFIIAgamma1 gene was significantly reduced while TFIIAgamma5 remained constitutively expressed and maintained the ancestral role as a subunit of the TFIIA complex. CONCLUSION: The evolutionary fate of TFIIAgamma duplicates is not consistent with the neofunctionalization model that predicts that one of the duplicated genes acquires a new function because of positive Darwinian selection. Instead, we suggest that subfunctionalization might be involved in TFIIAgamma evolution in grasses. The fact that both TFIIAgamma1 and TFIIAgamma5 genes were effectively involved in response to biotic or abiotic factors might be explained by either Dykhuizen-Hartl effect or buffering hypothesis.


Subject(s)
Oryza/genetics , Transcription Factor TFIIA/genetics , Cloning, Molecular , Evolution, Molecular , Gene Duplication , Gene Expression Regulation, Plant , Genes, Plant , Phylogeny , Poaceae/genetics , Sequence Analysis, DNA
18.
Plant Cell Rep ; 29(8): 857-64, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20480367

ABSTRACT

Transcription mediated by RNA polymerase II depends on a set of different transcription factors to form the pre-initiation complex. TFIIA is involved in the construction of this complex and increases the affinity of TBP for the DNA union region in vitro. In this study, we characterized the ScTFIIAgamma gene, which encodes a homolog of the smaller subunit (gamma) of transcription factor TFIIA in sugarcane. RNA blot analysis showed that ScTFIIAgamma transcripts accumulate in all tissues evaluated, with higher levels in leaf roll and flowers. In situ hybridization showed that ScTFIIAgamma was expressed in different cells of the reproductive meristem. In sugarcane plantlets, methyl jasmonate and absicic acid treatments as well as phosphate starvation had no influence on ScTFIIAgamma transcript accumulation. The subcelullar localization assay demonstrates that ScTFIIAgamma protein is directed to the cell nucleus. The phylogenetic analysis, the expression in several tissues and under different treatments and the nuclear localization are in line with the putative role of ScTFIIAgamma as a subunit of basal transcription factor.


Subject(s)
Plant Proteins/metabolism , Saccharum/genetics , Transcription Factor TFIIA/metabolism , Amino Acid Sequence , Cell Nucleus/genetics , Cloning, Molecular , Molecular Sequence Data , Phylogeny , Plant Proteins/genetics , RNA, Plant/genetics , Saccharum/metabolism , Sequence Alignment , Sequence Analysis, DNA , Transcription Factor TFIIA/genetics
19.
Nucleic Acids Res ; 36(4): 1343-57, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18187511

ABSTRACT

Saccharomyces cerevisiae HMO1, a high mobility group B (HMGB) protein, associates with the rRNA locus and with the promoters of many ribosomal protein genes (RPGs). Here, the Sos recruitment system was used to show that HMO1 interacts with TBP and the N-terminal domain (TAND) of TAF1, which are integral components of TFIID. Biochemical studies revealed that HMO1 copurifies with TFIID and directly interacts with TBP but not with TAND. Deletion of HMO1 (Deltahmo1) causes a severe cold-sensitive growth defect and decreases transcription of some TAND-dependent genes. Deltahmo1 also affects TFIID occupancy at some RPG promoters in a promoter-specific manner. Interestingly, over-expression of HMO1 delays colony formation of taf1 mutants lacking TAND (taf1DeltaTAND), but not of the wild-type strain, indicating a functional link between HMO1 and TAND. Furthermore, Deltahmo1 exhibits synthetic growth defects in some spt15 (TBP) and toa1 (TFIIA) mutants while it rescues growth defects of some sua7 (TFIIB) mutants. Importantly, Deltahmo1 causes an upstream shift in transcriptional start sites of RPS5, RPS16A, RPL23B, RPL27B and RPL32, but not of RPS31, RPL10, TEF2 and ADH1, indicating that HMO1 may participate in start site selection of a subset of class II genes presumably via its interaction with TFIID.


Subject(s)
High Mobility Group Proteins/metabolism , RNA Polymerase II/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , TATA-Box Binding Protein/metabolism , Transcription Factor TFIID/metabolism , Transcription Initiation Site , High Mobility Group Proteins/genetics , Mutation , Promoter Regions, Genetic , Protein Structure, Tertiary , Ribosomal Proteins/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Deletion , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , TATA-Box Binding Protein/genetics , Transcription Factor TFIIA/genetics , Transcription Factor TFIIB/genetics , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/genetics , Transcription, Genetic
20.
Biochim Biophys Acta Gene Regul Mech ; 1863(10): 194611, 2020 10.
Article in English | MEDLINE | ID: mdl-32745626

ABSTRACT

General transcription factors and core promoter elements play a pivotal role in RNA polymerase II (Pol II)-mediated transcription initiation. In the previous work, we have defined a TFIIA recognition element (IIARE) that modulates Pol II-directed gene transcription in a promoter context-dependent manner. However, how TFIIA interacts with the IIARE and whether the interaction between TFIIA and the IIARE is involved in the regulation of gene transcription by Pol II are not fully understood. In the present study, we confirm that both K348 and K350 residues in TFIIAαß are required for the interaction between TFIIAαß and the IIARE. Disruption of the interaction between them by gene mutations dampens TFIIAαß binding to the AdML-IIARE promoter and the transcriptional activation of the promoter containing a IIARE in vitro and in vivo. Stable expression of the TFIIAαß mutant containing both K348A and K350A in the cell line with endogenous TFIIAαß silence represses endogenous gene expression by reducing the occupancies of TFIIAαß, TBP, p300, and Pol II at the promoters containing a IIARE. The findings from this study provide a novel insight into the regulatory mechanism of gene transcription mediated by TFIIA and the IIARE.


Subject(s)
Binding Sites , Gene Expression Regulation , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Response Elements , Transcription Factor TFIIA/metabolism , Transcription Factor TFIIB/metabolism , Transcriptional Activation , Amino Acids , Base Sequence , Cell Line , Humans , Models, Molecular , Mutation , Nucleotide Motifs , Protein Binding , Protein Conformation , Structure-Activity Relationship , Transcription Factor TFIIA/chemistry , Transcription Factor TFIIA/genetics , Transcription Factor TFIIB/chemistry , Transcription Factor TFIIB/genetics
SELECTION OF CITATIONS
SEARCH DETAIL