Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters











Publication year range
1.
J Virol ; 94(7)2020 03 17.
Article in English | MEDLINE | ID: mdl-31915284

ABSTRACT

Infectious bursal disease virus (IBDV) of the Birnaviridae family leads to immunosuppression of young chickens by destroying B cells in the bursa of Fabricius (BFs). Given the increasing number of variant IBDV strains, we urgently require a method to produce attenuated virus for vaccine development. To accomplish this goal, the dual-promoter plasmids in which the RNA polymerase II and RNA polymerase I (Pol I) promoters were placed upstream of the IBDV genomic sequence, which was followed by mouse Pol I terminator and a synthetic polyadenylation signal, were developed for rapid generation of IBDV. This approach did not require trans-supplementation of plasmids for the expression of VP1 and VP3, the main components of IBDV ribonucleoprotein (RNP). Based on the finding in this study that the IBDV RNP activity was partially retained by VP1-FLAG, we successfully rescued the replication-competent IBDV/1FLAG expressing VP1-FLAG. Compared with its parental counterpart, IBDV/1FLAG formed smaller size plaques in cultured cells and induced the same 100% immune protection in vivo However, neither retarded development nor severe BFs lesion was observed in the IBDV/1FLAG-inoculated chickens. Collectively, this is the first report that viral RNP activity was affected by the addition of an epitope tag on the componential viral proteins. Furthermore, this work demonstrates the rapid generation of attenuated IBDV from dual-promoter plasmids via reducing viral RNP activity by a fused FLAG tag on the C terminus of VP1. This would be a convenient strategy to attenuate epidemic variant IBDV strains for rapid and efficient vaccine development.IMPORTANCE Immunosuppression in chickens as a result of infectious bursal disease virus (IBDV) infection leads to significant economic losses in the poultry industry worldwide every year. Currently, vaccination is still the best way to prevent the prevalence of IBDV. However, with the occurrence of increasing numbers of variant IBDV strains, it is challenging to develop antigen-matched live attenuated vaccine. Here, we first developed a dual-promoter reverse-genetic system for the rapid generation of IBDV. Using this system, the attenuated IBDV/1FLAG expressing VP1-FLAG, which displays the decreased viral RNP activity, was rescued. Moreover, IBDV/1FLAG inoculation induced a similar level of neutralizing antibodies to that of its parental counterpart, protecting chickens against lethal challenge. Our study, for the first time, describes a dual-promoter reverse-genetic approach for the rapid generation of attenuated IBDV while maintaining entire parental antigenicity, suggesting a potential new method to attenuate epidemic variant IBDV strains for vaccine development.


Subject(s)
Infectious bursal disease virus/genetics , Ribonucleoproteins/metabolism , Vaccines, Attenuated , Viral Vaccines , Animals , Birnaviridae Infections/prevention & control , Birnaviridae Infections/veterinary , Cell Line , Chickens/virology , Epitopes/chemistry , HEK293 Cells , Humans , Mice , Plasmids/genetics , Pol1 Transcription Initiation Complex Proteins/chemistry , Polyadenylation , Poultry Diseases/prevention & control , Poultry Diseases/virology , Promoter Regions, Genetic , Protein Domains , Recombinant Proteins/chemistry , Reverse Genetics , Viral Structural Proteins/genetics
2.
PLoS Genet ; 15(2): e1008006, 2019 02.
Article in English | MEDLINE | ID: mdl-30802237

ABSTRACT

RNA polymerase I (Pol I) synthesizes ribosomal RNA (rRNA) in all eukaryotes, accounting for the major part of transcriptional activity in proliferating cells. Although basal Pol I transcription factors have been characterized in diverse organisms, the molecular basis of the robust rRNA production in vivo remains largely unknown. In S. cerevisiae, the multifunctional Net1 protein was reported to stimulate Pol I transcription. We found that the Pol I-stimulating function can be attributed to the very C-terminal region (CTR) of Net1. The CTR was required for normal cell growth and Pol I recruitment to rRNA genes in vivo and sufficient to promote Pol I transcription in vitro. Similarity with the acidic tail region of mammalian Pol I transcription factor UBF, which could partly functionally substitute for the CTR, suggests conserved roles for CTR-like domains in Pol I transcription from yeast to human.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA Polymerase I/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/growth & development , Amino Acid Sequence , Cell Cycle Proteins/genetics , Conserved Sequence , Humans , Nuclear Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/chemistry , Pol1 Transcription Initiation Complex Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/metabolism , RNA, Ribosomal/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sequence Deletion , Transcription, Genetic
3.
Nucleic Acids Res ; 46(12): 6188-6205, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29873780

ABSTRACT

p53 as an effector of nucleolar stress is well defined, but p53 independent mechanisms are largely unknown. Like p53, the NF-κB transcription factor plays a critical role in maintaining cellular homeostasis under stress. Many stresses that stimulate NF-κB also disrupt nucleoli. However, the link between nucleolar function and activation of the NF-κB pathway is as yet unknown. Here we demonstrate that artificial disruption of the PolI complex stimulates NF-κB signalling. Unlike p53 nucleolar stress response, this effect does not appear to be linked to inhibition of rDNA transcription. We show that specific stress stimuli of NF-κB induce degradation of a critical component of the PolI complex, TIF-IA. This degradation precedes activation of NF-κB and is associated with increased nucleolar size. It is mimicked by CDK4 inhibition and is dependent upon a novel pathway involving UBF/p14ARF and S44 of the protein. We show that blocking TIF-IA degradation blocks stress effects on nucleolar size and NF-κB signalling. Finally, using ex vivo culture, we show a strong correlation between degradation of TIF-IA and activation of NF-κB in freshly resected, human colorectal tumours exposed to the chemopreventative agent, aspirin. Together, our study provides compelling evidence for a new, TIF-IA-NF-κB nucleolar stress response pathway that has in vivo relevance and therapeutic implications.


Subject(s)
Cell Nucleolus/metabolism , NF-kappa B/metabolism , Pol1 Transcription Initiation Complex Proteins/metabolism , Stress, Physiological , Active Transport, Cell Nucleus , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Humans , Pol1 Transcription Initiation Complex Proteins/chemistry , RNA Polymerase I/metabolism , Serine/metabolism , Signal Transduction , Transcription Factor RelA/metabolism , Tumor Suppressor Protein p14ARF/physiology
4.
Mol Cell Biol ; 37(22)2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28874518

ABSTRACT

Upstream binding factor (UBF) is a member of the high-mobility group (HMG) box protein family, characterized by multiple HMG boxes and a C-terminal acidic region (AR). UBF is an essential transcription factor for rRNA genes and mediates the formation of transcriptionally active chromatin in the nucleolus. However, it remains unknown how UBF is specifically localized to the nucleolus. Here, we examined the molecular mechanisms that localize UBF to the nucleolus. We found that the first HMG box (HMG box 1), the linker region (LR), and the AR cooperatively regulate the nucleolar localization of UBF1. We demonstrated that the AR intramolecularly associates with and attenuates the DNA binding activity of HMG boxes and confers the structured DNA preference to HMG box 1. In contrast, the LR was found to serve as a nuclear localization signal and compete with HMG boxes to bind the AR, permitting nucleolar localization of UBF1. The LR sequence binds DNA and assists the stable chromatin binding of UBF. We also showed that the phosphorylation status of the AR does not clearly affect the localization of UBF1. Our results strongly suggest that associations of the AR with HMG boxes and the LR regulate UBF nucleolar localization.


Subject(s)
Cell Nucleolus/metabolism , Mutation , Pol1 Transcription Initiation Complex Proteins/chemistry , Pol1 Transcription Initiation Complex Proteins/metabolism , Binding Sites , Cell Line , Cell Nucleolus/genetics , HEK293 Cells , HMG-Box Domains , HeLa Cells , Humans , Phosphorylation , Pol1 Transcription Initiation Complex Proteins/genetics , Protein Binding
5.
Biochim Biophys Acta ; 1866(2): 189-196, 2016 12.
Article in English | MEDLINE | ID: mdl-27641688

ABSTRACT

Cancer cells devote the majority of their energy consumption to ribosome biogenesis, and pre-ribosomal RNA transcription accounts for 30-50% of all transcriptional activity. This aberrantly elevated biological activity is an attractive target for cancer therapeutic intervention if approaches can be developed to circumvent the development of side effects in normal cells. TIF-IA is a transcription factor that connects RNA polymerase I with the UBF/SL-1 complex to initiate the transcription of pre-ribosomal RNA. Its function is conserved in eukaryotes from yeast to mammals, and its activity is promoted by the phosphorylation of various oncogenic kinases in cancer cells. The depletion of TIF-IA induces cell death in lung cancer cells and mouse embryonic fibroblasts but not in several other normal tissue types evaluated in knock-out studies. Furthermore, the nuclear accumulation of TIF-IA under UTP down-regulated conditions requires the activity of LKB1 kinase, and LKB1-inactivated cancer cells are susceptible to cell death under such stress conditions. Therefore, TIF-IA may be a unique target to suppress ribosome biogenesis without significantly impacting the survival of normal tissues.


Subject(s)
Neoplasms/metabolism , Pol1 Transcription Initiation Complex Proteins/physiology , RNA Precursors/biosynthesis , Animals , Humans , Mice , Neoplasms/therapy , Phosphorylation , Pol1 Transcription Initiation Complex Proteins/chemistry , Pol1 Transcription Initiation Complex Proteins/genetics
6.
Nat Commun ; 7: 12129, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27418309

ABSTRACT

Transcription of ribosomal DNA by RNA polymerase I (Pol I) requires the initiation factor Rrn3. Here we report the cryo-EM structure of the Pol I-Rrn3 complex at 4.8 Å resolution. The structure reveals how Rrn3 binding converts an inactive Pol I dimer into an initiation-competent monomeric complex and provides insights into the mechanisms of Pol I-specific initiation and regulation.


Subject(s)
Pol1 Transcription Initiation Complex Proteins/chemistry , RNA Polymerase I/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Cryoelectron Microscopy , Pol1 Transcription Initiation Complex Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/metabolism , Protein Multimerization , RNA Polymerase I/genetics , RNA Polymerase I/metabolism , Saccharomyces cerevisiae Proteins/metabolism
7.
J Phys Chem B ; 119(34): 10998-1009, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-25830509

ABSTRACT

Single-stranded DNA binding (SSB) proteins bind with single-stranded DNA (ss-DNA) segments that are generated as intermediates during DNA metabolic processes. The primary function of an SSB protein is to protect the ss-DNA from being degraded so that other enzymes can effectively act on it. We have performed atomistic molecular dynamics simulations of the two DNA binding K homology (KH) domains (KH3 and KH4) of the far upstream element (FUSE) binding protein (FBP) complexed with two ss-DNA oligomers in aqueous solutions. Attempts have been made to study the effects of complexation on the internal motions of the protein domains and the correlated dynamics of the amino acid residue side chains. In agreement with experiments, KH3 domain has been found to be relatively more flexible in the complexed state. The calculations reveal increased long-range anticorrelated motions among several amino acid residues in the complexed forms. Compared to the KH4 domain, noticeable increase in N-H dipole ordering on complexation has been observed for the KH3 domain. Importantly, it is demonstrated that the effects of the DNA strands on the side chain orientations of the arginine and lysine residues and their ordering and dynamics play critical roles in forming the complexes and their structural stability.


Subject(s)
Carrier Proteins/chemistry , DNA, Single-Stranded/chemistry , Pol1 Transcription Initiation Complex Proteins/chemistry , Amino Acid Sequence , Base Sequence , Molecular Sequence Data , Protein Structure, Tertiary
8.
Nat Struct Mol Biol ; 21(9): 810-6, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25132180

ABSTRACT

Core Factor (CF) is a conserved RNA polymerase (Pol) I general transcription factor comprising Rrn6, Rrn11 and the TFIIB-related subunit Rrn7. CF binds TATA-binding protein (TBP), Pol I and the regulatory factors Rrn3 and upstream activation factor. We used chemical cross-linking-MS to determine the molecular architecture of CF and its interactions with TBP. The CF subunits assemble through an interconnected network of interactions between five structural domains that are conserved in orthologous subunits of the human Pol I factor SL1. We validated the cross-linking-derived model through a series of genetic and biochemical assays. Our combined results show the architecture of CF and the functions of the CF subunits in assembly of the complex. We extend these findings to model how CF assembles into the Pol I preinitiation complex, providing new insight into the roles of CF, TBP and Rrn3.


Subject(s)
Pol1 Transcription Initiation Complex Proteins/metabolism , RNA Polymerase I/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Models, Molecular , Pol1 Transcription Initiation Complex Proteins/chemistry , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA Polymerase I/chemistry , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/chemistry
9.
Nat Commun ; 5: 4310, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25007739

ABSTRACT

During transcription initiation at promoters of protein-coding genes, RNA polymerase (Pol) II assembles with TBP, TFIIB and TFIIF into a conserved core initiation complex that recruits additional factors. The core complex stabilizes open DNA and initiates RNA synthesis, and it is conserved in the Pol I and Pol III transcription systems. Here, we derive the domain architecture of the yeast core pol II initiation complex during transcription initiation. The yeast complex resembles the human initiation complex and reveals that the TFIIF Tfg2 winged helix domain swings over promoter DNA. An 'arm' and a 'charged helix' in TFIIF function in transcription start site selection and initial RNA synthesis, respectively, and apparently extend into the active centre cleft. Our model provides the basis for further structure-function analysis of the entire transcription initiation complex.


Subject(s)
Pol1 Transcription Initiation Complex Proteins/chemistry , Pol1 Transcription Initiation Complex Proteins/genetics , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Base Sequence , Eukaryotic Initiation Factors/chemistry , Eukaryotic Initiation Factors/genetics , Mass Spectrometry , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Structure, Tertiary/genetics , Transcription Factors, TFII/chemistry , Transcription Factors, TFII/genetics , Transcription Initiation, Genetic/physiology
10.
Nucleic Acids Res ; 42(3): 1628-43, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24234436

ABSTRACT

The remodeling of chromatin in the nucleolus is important for the control of ribosomal DNA (rDNA) transcription and ribosome biogenesis. Herein, we found that upstream binding factor (UBF) interacts with ESET, a histone H3K9 methyltransferase and is trimethylated at Lys (K) 232/254 by ESET. UBF trimethylation leads to nucleolar chromatin condensation and decreased rDNA transcriptional activity. UBF mutations at K232/254A and K232/254R restored rDNA transcriptional activity in response to ESET. Both ESET-ΔSET mutant and knockdown of ESET by short hairpin RNA reduced trimethylation of UBF and resulted in the restoration of rDNA transcription. Atomic force microscopy confirmed that UBF trimethylated by ESET modulates the plasticity of nucleolar chromatin. We further demonstrated that UBF trimethylation at K232/254 by ESET deregulates rDNA transcription in a cell model of Huntington's disease. Together, our findings show that a novel epigenetic modification of UBF is linked to impaired rDNA transcription and nucleolar chromatin remodeling, which may play key roles in the pathogenesis of neurodegeneration.


Subject(s)
Cell Nucleolus/enzymology , Cell Nucleolus/genetics , DNA, Ribosomal/metabolism , Heterochromatin/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Pol1 Transcription Initiation Complex Proteins/metabolism , Transcription, Genetic , Animals , Cell Line , Humans , Huntington Disease/enzymology , Methylation , Mice , Mutation , Pol1 Transcription Initiation Complex Proteins/chemistry , Pol1 Transcription Initiation Complex Proteins/genetics
11.
Nucleic Acids Res ; 41(22): 10135-49, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24021628

ABSTRACT

Ribosome biogenesis is a major metabolic effort for growing cells. In Saccharomyces cerevisiae, Hmo1, an abundant high-mobility group box protein (HMGB) binds to the coding region of the RNA polymerase I transcribed ribosomal RNAs genes and the promoters of ∼70% of ribosomal protein genes. In this study, we have demonstrated the functional conservation of eukaryotic HMGB proteins involved in ribosomal DNA (rDNA) transcription. We have shown that when expressed in budding yeast, human UBF1 and a newly identified Sp-Hmo1 (Schizosaccharomyces pombe) localize to the nucleolus and suppress growth defect of the RNA polymerase I mutant rpa49-Δ. Owing to the multiple functions of both proteins, Hmo1 and UBF1 are not fully interchangeable. By deletion and domains swapping in Hmo1, we identified essential domains that stimulate rDNA transcription but are not fully required for stimulation of ribosomal protein genes expression. Hmo1 is organized in four functional domains: a dimerization module, a canonical HMGB motif followed by a conserved domain and a C-terminal nucleolar localization signal. We propose that Hmo1 has acquired species-specific functions and shares with UBF1 and Sp-Hmo1 an ancestral function to stimulate rDNA transcription.


Subject(s)
DNA, Ribosomal/metabolism , HMGB Proteins/chemistry , HMGB Proteins/metabolism , High Mobility Group Proteins/chemistry , High Mobility Group Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic , Amino Acid Motifs , Amino Acid Sequence , Cell Nucleolus/metabolism , Conserved Sequence , HMGB Proteins/genetics , High Mobility Group Proteins/genetics , Humans , Pol1 Transcription Initiation Complex Proteins/chemistry , Pol1 Transcription Initiation Complex Proteins/metabolism , Protein Structure, Tertiary , RNA Polymerase I/metabolism , Ribosomal Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Structure-Activity Relationship
12.
J Biol Chem ; 288(13): 9135-44, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23393135

ABSTRACT

The human homologue of yeast Rrn3 is an RNA polymerase I-associated transcription factor that is essential for ribosomal DNA (rDNA) transcription. The generally accepted model is that Rrn3 functions as a bridge between RNA polymerase I and the transcription factors bound to the committed template. In this model Rrn3 would mediate an interaction between the mammalian Rrn3-polymerase I complex and SL1, the rDNA transcription factor that binds to the core promoter element of the rDNA. In the course of studying the role of Rrn3 in recruitment, we found that Rrn3 was in fact a DNA-binding protein. Analysis of the sequence of Rrn3 identified a domain with sequence similarity to the DNA binding domain of heat shock transcription factor 2. Randomization, or deletion, of the amino acids in this region in Rrn3, amino acids 382-400, abrogated its ability to bind DNA, indicating that this domain was an important contributor to DNA binding by Rrn3. Control experiments demonstrated that these mutant Rrn3 constructs were capable of interacting with both rpa43 and SL1, two other activities demonstrated to be essential for Rrn3 function. However, neither of these Rrn3 mutants was capable of functioning in transcription in vitro. Moreover, although wild-type human Rrn3 complemented a yeast rrn3-ts mutant, the DNA-binding site mutant did not. These results demonstrate that DNA binding by Rrn3 is essential for transcription by RNA polymerase I.


Subject(s)
DNA/chemistry , DNA/metabolism , Pol1 Transcription Initiation Complex Proteins/chemistry , Ribosomes/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Cell Line , Cell Nucleolus/metabolism , DNA, Ribosomal/metabolism , DNA-Binding Proteins/metabolism , Gene Deletion , Humans , Insecta , Mice , Molecular Sequence Data , Mutagenesis , Pol1 Transcription Initiation Complex Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , RNA/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Amino Acid , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism
13.
Int J Mol Sci ; 13(8): 10553-10567, 2012.
Article in English | MEDLINE | ID: mdl-22949879

ABSTRACT

X-ray crystallography requires high quality crystals above a given size. This requirement not only limits the proteins to be analyzed, but also reduces the speed of the structure determination. Indeed, the tertiary structures of many physiologically important proteins remain elusive because of the so-called "crystallization bottleneck". Once microcrystals have been obtained, crystallization conditions can be optimized to produce bigger and better crystals. However, the identification of microcrystals can be difficult due to the resolution limit of optical microscopy. Electron microscopy has sometimes been utilized instead, with the disadvantage that the microcrystals usually must be observed in vacuum, which precludes the usage for crystal screening. The atmospheric scanning electron microscope (ASEM) allows samples to be observed in solution. Here, we report the use of this instrument in combination with a special thin-membrane dish with a crystallization well. It was possible to observe protein crystals of lysozyme, lipase B and a histone chaperone TAF-Iß in crystallization buffers, without the use of staining procedures. The smallest crystals observed with ASEM were a few µm in width, and ASEM can be used with non-transparent solutions. Furthermore, the growth of salt crystals could be monitored in the ASEM, and the difference in contrast between salt and protein crystals made it easy to distinguish between these two types of microcrystals. These results indicate that the ASEM could be an important new tool for the screening of protein microcrystals.


Subject(s)
Lipase/chemistry , Microscopy, Electron, Scanning/methods , Muramidase/chemistry , Pol1 Transcription Initiation Complex Proteins/chemistry , Animals , Chickens , Crystallization , Crystallography, X-Ray , Humans , Lipase/metabolism , Muramidase/metabolism , Pol1 Transcription Initiation Complex Proteins/metabolism
14.
Genes Dev ; 25(19): 2093-105, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21940764

ABSTRACT

Cell growth is regulated during RNA polymerase (Pol) I transcription initiation by the conserved factor Rrn3/TIF-IA in yeast/humans. Here we provide a structure-function analysis of Rrn3 based on a combination of structural biology with in vivo and in vitro functional assays. The Rrn3 crystal structure reveals a unique HEAT repeat fold and a surface serine patch. Phosphorylation of this patch represses human Pol I transcription, and a phospho-mimetic patch mutation prevents Rrn3 binding to Pol I in vitro and reduces cell growth and Pol I gene occupancy in vivo. Cross-linking indicates that Rrn3 binds Pol I between its subcomplexes, AC40/19 and A14/43, which faces the serine patch. The corresponding region of Pol II binds the Mediator head that cooperates with transcription factor (TF) IIB. Consistent with this, the Rrn3-binding factor Rrn7 is predicted to be a TFIIB homolog. This reveals the molecular basis of Rrn3-regulated Pol I initiation and cell growth, and indicates a general architecture of eukaryotic transcription initiation complexes.


Subject(s)
DNA Polymerase I/metabolism , Models, Molecular , Pol1 Transcription Initiation Complex Proteins/chemistry , Pol1 Transcription Initiation Complex Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Cell Proliferation , Humans , Molecular Sequence Data , Mutation , Pol1 Transcription Initiation Complex Proteins/genetics , Promoter Regions, Genetic , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment , Serine/metabolism
15.
Science ; 333(6049): 1637-40, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21921198

ABSTRACT

Eukaryotic and archaeal multisubunit RNA polymerases (Pols) are structurally related and require several similar components for transcription initiation. However, none of the Pol I factors were known to share homology with transcription factor IIB (TFIIB) or TFIIB-related proteins, key factors in the initiation mechanisms of the other Pols. Here we show that Rrn7, a subunit of the yeast Pol I core factor, and its human ortholog TAF1B are TFIIB-like factors. Although distantly related, Rrn7 shares many activities associated with TFIIB-like factors. Domain swaps between TFIIB-related factors show that Rrn7 is most closely related to the Pol III general factor Brf1. Our results point to the conservation of initiation mechanisms among multisubunit Pols and reveal a key function of yeast core factor/human SL1 in Pol I transcription.


Subject(s)
Pol1 Transcription Initiation Complex Proteins/chemistry , Pol1 Transcription Initiation Complex Proteins/metabolism , RNA Polymerase I/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Humans , Molecular Sequence Data , Pol1 Transcription Initiation Complex Proteins/genetics , Protein Folding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment , TATA-Box Binding Protein/metabolism , Transcription Factor TFIIB/chemistry , Transcription Factor TFIIB/metabolism , Transcription Factor TFIIIB/chemistry , Transcription Factor TFIIIB/genetics , Transcription Factor TFIIIB/metabolism , Transcription, Genetic
16.
Science ; 333(6049): 1640-2, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21921199

ABSTRACT

Transcription by eukaryotic RNA polymerases (Pols) II and III and archaeal Pol requires structurally related general transcription factors TFIIB, Brf1, and TFB, respectively, which are essential for polymerase recruitment and initiation events. A TFIIB-like protein was not evident in the Pol I basal transcription machinery. We report that TAF1B, a subunit of human Pol I basal transcription factor SL1, is structurally related to TFIIB/TFIIB-like proteins, through predicted amino-terminal zinc ribbon and cyclin-like fold domains. SL1, essential for Pol I recruitment to the ribosomal RNA gene promoter, also has an essential postpolymerase recruitment role, operating through TAF1B. Therefore, a TFIIB-related protein is implicated in preinitiation complex assembly and postpolymerase recruitment events in Pol I transcription, underscoring the parallels between eukaryotic Pol I, II, and III and archaeal transcription machineries.


Subject(s)
Pol1 Transcription Initiation Complex Proteins/chemistry , Pol1 Transcription Initiation Complex Proteins/metabolism , RNA Polymerase I/metabolism , Transcription Factor TFIIB/chemistry , Transcription, Genetic , Amino Acid Sequence , DNA, Ribosomal , Humans , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation , Pol1 Transcription Initiation Complex Proteins/genetics , Promoter Regions, Genetic , Protein Binding , Protein Folding , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , Transcription Factor TFIIB/metabolism
17.
Proc Natl Acad Sci U S A ; 106(42): 17781-6, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19815529

ABSTRACT

AMP-activated protein kinase (AMPK) senses changes in the intracellular AMP/ATP ratio, switching off energy-consuming processes and switching on catabolic pathways in response to energy depletion. Here, we show that AMPK down-regulates rRNA synthesis under glucose restriction by phosphorylating the RNA polymerase I (Pol I)-associated transcription factor TIF-IA at a single serine residue (Ser-635). Phosphorylation by AMPK impairs the interaction of TIF-IA with the TBP-containing promoter selectivity factor SL1, thereby precluding the assembly of functional transcription initiation complexes. Mutation of Ser-635 compromises down-regulation of Pol I transcription in response to low energy supply, supporting that activation of AMPK adapts rRNA synthesis to nutrient availability and the cellular energy status.


Subject(s)
AMP-Activated Protein Kinases/metabolism , RNA, Ribosomal/biosynthesis , Adenosine Triphosphate/metabolism , Animals , Cell Line , Energy Metabolism , Glucose/metabolism , Humans , Mice , Models, Biological , NIH 3T3 Cells , Phosphorylation , Pol1 Transcription Initiation Complex Proteins/antagonists & inhibitors , Pol1 Transcription Initiation Complex Proteins/chemistry , Pol1 Transcription Initiation Complex Proteins/genetics , Pol1 Transcription Initiation Complex Proteins/metabolism , RNA Polymerase I/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Transcription, Genetic
18.
Proteins ; 77(2): 432-47, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19452555

ABSTRACT

Human upstream binding factor (hUBF) HMG Box-5 is a highly conserved protein domain, containing 84 amino acids and belonging to the family of the nonspecific DNA-binding HMG boxes. Its native structure adopts a twisted L shape, which consists of three alpha-helices and two hydrophobic cores: the major wing and the minor wing. In this article, we report a reversible three-state thermal unfolding equilibrium of hUBF HMG Box-5, which is investigated by differential scanning calorimetry (DSC), circular dichroism spectroscopy, fluorescence spectroscopy, and NMR spectroscopy. DSC data show that Box-5 unfolds reversibly in two separate stages. Spectroscopic analyses suggest that different structural elements exhibit noncooperative transitions during the unfolding process and that the major form of the Box-5 thermal intermediate ensemble at 55 degrees C shows partially unfolded characteristics. Compared with previous thermal stability studies of other boxes, it appears that Box-5 possesses a more stable major wing and two well separated subdomains. NMR chemical shift index and sequential (1)H(N) (i)-(1)H(N) (i+1) NOE analyses indicate that helices 1 and 2 are native-like in the thermal intermediate ensemble, while helix 3 is partially unfolded. Detailed NMR relaxation dynamics are compared between the native state and the intermediate ensemble. Our results implicate a fluid helix-turn-helix folding model of Box-5, where helices 1 and 2 potentially form the helix 1-turn-helix 2 motif in the intermediate, while helix 3 is consolidated only as two hydrophobic cores form to stabilize the native structure.


Subject(s)
HMGB Proteins/chemistry , Helix-Turn-Helix Motifs , Pol1 Transcription Initiation Complex Proteins/chemistry , HMGB Proteins/metabolism , Humans , Models, Molecular , Pol1 Transcription Initiation Complex Proteins/metabolism , Protein Conformation , Protein Folding , Thermodynamics
19.
Nucleic Acids Res ; 36(15): 5093-101, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18676449

ABSTRACT

The mammalian architectural HMGB-Box transcription factor UBF is ubiquitously expressed in two variant forms as the result of a differential splicing event, that in the UBF2 deletes 37 amino acid from the second of six HMGB-boxes. Several attempts to define a function for this shorter UBF2 protein have been less than satisfactory. However, since all mammals appear to display similar levels of the longer and shorter UBF variants, it is unlikely that UBF2 is simply nonfunctional. Previously we showed that phosphorylation of UBF by the MAP-kinase ERK regulates chromatin folding and transcription elongation, explaining the rapid response of the ribosomal RNA genes to growth factors. Here we have investigated the roles the UBF variants play in the response of these genes to ERK activity. We demonstrate that the variant HMGB-box 2 of UBF2 has lost the ability to bind bent DNA and hence to induce chromatin folding. As a result it is significantly less effective than UBF1 at arresting RNAPI elongation but at the same time is more responsive to ERK phosphorylation. Thus, UBF2 functionally simulates a hemi-phosphorylated UBF whose expression may provide a means by which to tune the response of the ribosomal RNA genes to growth factor stimulation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Pol1 Transcription Initiation Complex Proteins/physiology , RNA Polymerase I/metabolism , Transcription, Genetic , Animals , DNA/chemistry , DNA/metabolism , Genetic Variation , HMG-Box Domains , HMGB Proteins/chemistry , Mice , Phosphorylation , Pol1 Transcription Initiation Complex Proteins/chemistry , Pol1 Transcription Initiation Complex Proteins/genetics , RNA Polymerase I/antagonists & inhibitors , RNA Splicing , Rats
20.
Acta Crystallogr D Biol Crystallogr ; 63(Pt 6): 730-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17505112

ABSTRACT

The fifth HMG-box domain in human upstream binding factor (hUBF) contributes to the synthesis of rRNA by RNA polymerase I (Pol I). The 2.0 A resolution crystal structure of this protein has been solved using the single-wavelength anomalous dispersion method (SAD). The crystal structure and the reported NMR structure have r.m.s. deviations of 2.18-3.03 A for the C(alpha) atoms. However, there are significant differences between the two structures, with displacements of up to 9.0 A. Compared with other HMG-box structures, the r.m.s. deviations for C(alpha) atoms between hUBF HMG box 5 and HMG domains from Drosophila melanogaster protein D and Rattus norvegicus HMG1 are 1.5 and 1.6 A, respectively. This indicates that the differences between the crystal and NMR structures of hUBF HMG box 5 are larger than those with its homologous structures. The differences between the two structures potentially reflect two states with different structures. The specific interactions between the hUBF HMG box 5 and the first bromodomain of TBP-associated factor 1 (TAF1) were studied by ultrasensitive differential scanning calorimetry and chemical shift perturbation. Based on these experimental data, possible sites in hUBF HMG box 5 that may interact with the first bromodomain of TAF1 were proposed.


Subject(s)
HMGB Proteins/chemistry , HMGB Proteins/metabolism , Pol1 Transcription Initiation Complex Proteins/chemistry , Pol1 Transcription Initiation Complex Proteins/metabolism , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/metabolism , Animals , Base Sequence , Binding Sites , Crystallography, X-Ray , DNA Primers/genetics , Drosophila Proteins/chemistry , HMGB Proteins/genetics , Histone Acetyltransferases , Humans , In Vitro Techniques , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Pol1 Transcription Initiation Complex Proteins/genetics , Protein Structure, Tertiary , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , TATA-Binding Protein Associated Factors/genetics , Thermodynamics , Transcription Factor TFIID/genetics
SELECTION OF CITATIONS
SEARCH DETAIL