Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
1.
Biophys Chem ; 312: 107281, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38889653

ABSTRACT

RNA polymerase I (Pol I) is responsible for synthesizing ribosomal RNA, which is the rate limiting step in ribosome biogenesis. We have reported wide variability in the magnitude of the rate constants defining the rate limiting step in sequential nucleotide additions catalyzed by Pol I. in this study we sought to determine if base identity impacts the rate limiting step of nucleotide addition catalyzed by Pol I. To this end, we report a transient state kinetic interrogation of AMP, CMP, GMP, and UMP incorporations catalyzed by Pol I. We found that Pol I uses one kinetic mechanism to incorporate all nucleotides. However, we found that UMP incorporation is faster than AMP, CMP, and GMP additions. Further, we found that endonucleolytic removal of a dimer from the 3' end was fastest when the 3' terminal base is a UMP. It has been previously shown that both downstream and upstream template sequence identity impacts the kinetics of nucleotide addition. The results reported here show that the incoming base identity also impacts the magnitude of the observed rate limiting step.


Subject(s)
RNA Polymerase I , Kinetics , RNA Polymerase I/metabolism , RNA Polymerase I/chemistry , Nucleotides/metabolism , Nucleotides/chemistry
2.
Biol Chem ; 404(11-12): 979-1002, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37823775

ABSTRACT

Ribosomal RNAs (rRNAs) are structural components of ribosomes and represent the most abundant cellular RNA fraction. In the yeast Saccharomyces cerevisiae, they account for more than 60 % of the RNA content in a growing cell. The major amount of rRNA is synthesized by RNA polymerase I (Pol I). This enzyme transcribes exclusively the rRNA gene which is tandemly repeated in about 150 copies on chromosome XII. The high number of transcribed rRNA genes, the efficient recruitment of the transcription machinery and the dense packaging of elongating Pol I molecules on the gene ensure that enough rRNA is generated. Specific features of Pol I and of associated factors confer promoter selectivity and both elongation and termination competence. Many excellent reviews exist about the state of research about function and regulation of Pol I and how Pol I initiation complexes are assembled. In this report we focus on the Pol I specific lobe binding subunits which support efficient, error-free, and correctly terminated rRNA synthesis.


Subject(s)
RNA Polymerase I , Saccharomyces cerevisiae , RNA Polymerase I/chemistry , RNA Polymerase I/genetics , RNA Polymerase I/metabolism , Saccharomyces cerevisiae/metabolism , Transcription, Genetic , Ribosomes/genetics , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism
3.
Biol Chem ; 404(11-12): 1003-1023, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37454246

ABSTRACT

The ribosomal RNA precursor (pre-rRNA) comprises three of the four ribosomal RNAs and is synthesized by RNA polymerase (Pol) I. Here, we describe the mechanisms of Pol I transcription in human cells with a focus on recent insights gained from structure-function analyses. The comparison of Pol I-specific structural and functional features with those of other Pols and with the excessively studied yeast system distinguishes organism-specific from general traits. We explain the organization of the genomic rDNA loci in human cells, describe the Pol I transcription cycle regarding structural changes in the enzyme and the roles of human Pol I subunits, and depict human rDNA transcription factors and their function on a mechanistic level. We disentangle information gained by direct investigation from what had apparently been deduced from studies of the yeast enzymes. Finally, we provide information about how Pol I mutations may contribute to developmental diseases, and why Pol I is a target for new cancer treatment strategies, since increased rRNA synthesis was correlated with rapidly expanding cell populations.


Subject(s)
RNA Precursors , RNA, Ribosomal , Humans , RNA, Ribosomal/genetics , RNA Precursors/genetics , Saccharomyces cerevisiae/metabolism , Transcription, Genetic , RNA Polymerase I/genetics , RNA Polymerase I/chemistry , RNA Polymerase I/metabolism , DNA, Ribosomal/genetics
4.
J Mol Biol ; 435(15): 168186, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37355033

ABSTRACT

RNA polymerase I (Pol I) synthesizes ribosomal RNA (rRNA), which is the first and rate-limiting step in ribosome biosynthesis. A12.2 (A12) is a critical subunit of Pol I that is responsible for activating Pol I's exonuclease activity. We previously reported a kinetic mechanism for single-nucleotide incorporation catalyzed by Pol I lacking the A12 subunit (ΔA12 Pol I) purified from S. cerevisae and revealed that ΔA12 Pol I exhibited much slower incorporation compared to Pol I. However, it is unknown if A12 influences each nucleotide incorporation in the context of transcription elongation. Here, we show that A12 contributes to every repeating cycle of nucleotide addition and that deletion of A12 results in an entirely different kinetic mechanism compared to WT Pol I. We found that instead of one irreversible step between each nucleotide addition cycle, as reported for wild type (WT) Pol I, the ΔA12 variant requires one reversible step to describe each nucleotide addition. Reversibility fundamentally requires slow PPi release. Consistently, we show that Pol I is more pyrophosphate (PPi) concentration dependent than ΔA12 Pol I. This observation supports the model that PPi is retained in the active site of ΔA12 Pol I longer than WT Pol I. These results suggest that A12 promotes PPi release, revealing a larger role for the A12.2 subunit in the nucleotide addition cycle beyond merely activating exonuclease activity.


Subject(s)
Diphosphates , RNA Polymerase I , Diphosphates/metabolism , Exonucleases , Nucleotides/metabolism , RNA Polymerase I/chemistry , RNA Polymerase I/genetics , RNA Polymerase I/metabolism
5.
Nat Commun ; 12(1): 758, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33536435

ABSTRACT

RNA polymerase (Pol) I transcribes the ribosomal RNA precursor in all eukaryotes. The mechanisms 'activation by cleft contraction' and 'hibernation by dimerization' are unique to the regulation of this enzyme, but structure-function analysis is limited to baker's yeast. To understand whether regulation by such strategies is specific to this model organism or conserved among species, we solve three cryo-EM structures of Pol I from Schizosaccharomyces pombe in different functional states. Comparative analysis of structural models derived from high-resolution reconstructions shows that activation is accomplished by a conserved contraction of the active center cleft. In contrast to current beliefs, we find that dimerization of the S. pombe polymerase is also possible. This dimerization is achieved independent of the 'connector' domain but relies on two previously undescribed interfaces. Our analyses highlight the divergent nature of Pol I transcription systems from their counterparts and suggest conservation of regulatory mechanisms among organisms.


Subject(s)
RNA Polymerase I/chemistry , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces/enzymology , Transcription, Genetic , Amino Acid Sequence , Base Sequence , Cryoelectron Microscopy , Models, Molecular , Protein Conformation , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Polymerase I/genetics , RNA Polymerase I/metabolism , RNA, Ribosomal/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Sequence Homology, Amino Acid
6.
J Mol Biol ; 432(13): 3933-3949, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32325070

ABSTRACT

RNA polymerase transcribes certain genomic loci with higher errors rates. These transcription error-enriched genomic loci (TEELs) have implications in disease. Current deep-sequencing methods cannot distinguish TEELs from post-transcriptional modifications, stochastic transcription errors, and technical noise, impeding efforts to elucidate the mechanisms linking TEELs to disease. Here, we describe background error model-coupled precision nuclear run-on circular-sequencing (EmPC-seq) to discern genomic regions enriched for transcription misincorporations. EmPC-seq innovatively combines a nuclear run-on assay for capturing nascent RNA before post-transcriptional modifications, a circular-sequencing step that sequences the same nascent RNA molecules multiple times to improve accuracy, and a statistical model for distinguishing error-enriched regions among stochastic polymerase errors. Applying EmPC-seq to the ribosomal RNA transcriptome, we show that TEELs of RNA polymerase I are not randomly distributed but clustered together, with higher error frequencies at nascent transcript 3' ends. Our study establishes a reliable method of identifying TEELs with nucleotide precision, which can help elucidate their molecular origins.


Subject(s)
RNA Polymerase I/genetics , RNA/genetics , Transcription, Genetic , Transcriptome/genetics , High-Throughput Nucleotide Sequencing , Humans , RNA Polymerase I/chemistry , RNA Processing, Post-Transcriptional/genetics , RNA, Ribosomal/chemistry , RNA, Ribosomal/genetics
7.
Nat Commun ; 11(1): 1206, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32139698

ABSTRACT

Transcription of the ribosomal RNA precursor by RNA polymerase (Pol) I is a prerequisite for the biosynthesis of ribosomes in eukaryotes. Compared to Pols II and III, the mechanisms underlying promoter recognition, initiation complex formation and DNA melting by Pol I substantially diverge. Here, we report the high-resolution cryo-EM reconstruction of a Pol I early initiation intermediate assembled on a double-stranded promoter scaffold that prevents the establishment of downstream DNA contacts. Our analyses demonstrate how efficient promoter-backbone interaction is achieved by combined re-arrangements of flexible regions in the 'core factor' subunits Rrn7 and Rrn11. Furthermore, structure-function analysis illustrates how destabilization of the melted DNA region correlates with contraction of the polymerase cleft upon transcription activation, thereby combining promoter recruitment with DNA-melting. This suggests that molecular mechanisms and structural features of Pol I initiation have co-evolved to support the efficient melting, initial transcription and promoter clearance required for high-level rRNA synthesis.


Subject(s)
Promoter Regions, Genetic , RNA Polymerase I/metabolism , Saccharomyces cerevisiae/genetics , Transcription Initiation, Genetic , Amino Acid Sequence , DNA/chemistry , DNA/ultrastructure , Models, Molecular , Protein Structure, Secondary , Protein Subunits/metabolism , RNA Polymerase I/chemistry , RNA Polymerase I/ultrastructure , Transcription Factor TFIIB/metabolism
8.
J Biol Chem ; 295(15): 4782-4795, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32060094

ABSTRACT

RNA polymerase I (Pol I) is a highly efficient enzyme specialized in synthesizing most ribosomal RNAs. After nucleosome deposition at each round of rDNA replication, the Pol I transcription machinery has to deal with nucleosomal barriers. It has been suggested that Pol I-associated factors facilitate chromatin transcription, but it is unknown whether Pol I has an intrinsic capacity to transcribe through nucleosomes. Here, we used in vitro transcription assays to study purified WT and mutant Pol I variants from the yeast Saccharomyces cerevisiae and compare their abilities to pass a nucleosomal barrier with those of yeast Pol II and Pol III. Under identical conditions, purified Pol I and Pol III, but not Pol II, could transcribe nucleosomal templates. Pol I mutants lacking either the heterodimeric subunit Rpa34.5/Rpa49 or the C-terminal part of the specific subunit Rpa12.2 showed a lower processivity on naked DNA templates, which was even more reduced in the presence of a nucleosome. Our findings suggest that the lobe-binding subunits Rpa34.5/Rpa49 and Rpa12.2 facilitate passage through nucleosomes, suggesting possible cooperation among these subunits. We discuss the contribution of Pol I-specific subunit domains to efficient Pol I passage through nucleosomes in the context of transcription rate and processivity.


Subject(s)
Chromatin/metabolism , Nucleosomes/metabolism , RNA Polymerase III/metabolism , RNA Polymerase II/metabolism , RNA Polymerase I/metabolism , Saccharomyces cerevisiae/metabolism , Transcription, Genetic , Chromatin/genetics , DNA Replication , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Nucleosomes/genetics , Promoter Regions, Genetic , Protein Binding , Protein Subunits/metabolism , RNA Polymerase I/chemistry , RNA Polymerase I/genetics , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , RNA Polymerase III/chemistry , RNA Polymerase III/genetics , Ribosomes , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development
9.
Nat Commun ; 10(1): 5543, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31804486

ABSTRACT

RNA polymerase I (Pol I) assembles with core factor (CF) and Rrn3 on the rDNA core promoter for transcription initiation. Here, we report cryo-EM structures of closed, intermediate and open Pol I initiation complexes from 2.7 to 3.7 Å resolution to visualize Pol I promoter melting and to structurally and biochemically characterize the recognition mechanism of Pol I promoter DNA. In the closed complex, double-stranded DNA runs outside the DNA-binding cleft. Rotation of CF and upstream DNA with respect to Pol I and Rrn3 results in the spontaneous loading and opening of the promoter followed by cleft closure and positioning of the Pol I A49 tandem winged helix domain (tWH) onto DNA. Conformational rearrangement of A49 tWH leads to a clash with Rrn3 to initiate complex disassembly and promoter escape. Comprehensive insight into the Pol I transcription initiation cycle allows comparisons with promoter opening by Pol II and Pol III.


Subject(s)
DNA, Fungal/genetics , Promoter Regions, Genetic/genetics , RNA Polymerase I/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Transcription, Genetic , Binding Sites/genetics , Cryoelectron Microscopy , DNA, Fungal/chemistry , DNA, Fungal/metabolism , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Domains , RNA Polymerase I/chemistry , RNA Polymerase I/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Transcription Initiation, Genetic
10.
J Biol Chem ; 294(52): 19907-19922, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31727736

ABSTRACT

Our knowledge of the mechanism of rDNA transcription has benefited from the combined application of genetic and biochemical techniques in yeast. Nomura's laboratory (Nogi, Y., Vu, L., and Nomura, M. (1991) Proc. Natl. Acad. Sci. U.S.A. 88, 7026-7030 and Nogi, Y., Yano, R., and Nomura, M. (1991) Proc. Natl. Acad. Sci. U.S.A. 88, 3962-3966) developed a system in yeast to identify genes essential for ribosome biogenesis. Such systems have allowed investigators to determine whether a gene was essential and to determine its function in rDNA transcription. However, there are significant differences in both the structures and components of the transcription apparatus and the patterns of regulation between mammals and yeast. Thus, there are significant deficits in our understanding of mammalian rDNA transcription. We have developed a system combining CRISPR/Cas9 and an auxin-inducible degron that enables combining a "genetics-like"approach with biochemistry to study mammalian rDNA transcription. We now show that the mammalian orthologue of yeast RPA49, PAF53, is required for rDNA transcription and mitotic growth. We have studied the domains of the protein required for activity. We have found that the C-terminal, DNA-binding domain (tandem-winged helix), the heterodimerization, and the linker domain were essential. Analysis of the linker identified a putative helix-turn-helix (HTH) DNA-binding domain. This HTH constitutes a second DNA-binding domain within PAF53. The HTH of the yeast and mammalian orthologues is essential for function. In summary, we show that an auxin-dependent degron system can be used to rapidly deplete nucleolar proteins in mammalian cells, that PAF53 is necessary for rDNA transcription and cell growth, and that all three PAF53 domains are necessary for its function.


Subject(s)
Mitosis , RNA Polymerase I/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Animals , CRISPR-Cas Systems/genetics , DNA, Ribosomal/metabolism , Dimerization , Helix-Turn-Helix Motifs , Indoleacetic Acids/metabolism , Mice , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Polymerase I/chemistry , RNA Polymerase I/genetics , RNA, Guide, Kinetoplastida/metabolism , S Phase Cell Cycle Checkpoints , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic
11.
Biochim Biophys Acta Gene Regul Mech ; 1862(9): 194408, 2019 09.
Article in English | MEDLINE | ID: mdl-31382053

ABSTRACT

In Saccharomyces cerevisiae, Core Factor (CF) is a key evolutionarily conserved transcription initiation factor that helps recruit RNA polymerase I (Pol I) to the ribosomal DNA (rDNA) promoter. Upregulated Pol I transcription has been linked to many cancers, and targeting Pol I is an attractive and emerging anti-cancer strategy. Using yeast as a model system, we characterized how CF binds to the Pol I promoter by electrophoretic mobility shift assays (EMSA). Synthetic DNA competitors along with anti-tumor drugs and nucleic acid stains that act as DNA groove blockers were used to discover the binding preference of yeast CF. Our results show that CF employs a unique binding mechanism where it prefers the GC-rich minor groove within the rDNA promoter. In addition, we show that yeast CF is able to bind to the human rDNA promoter sequence that is divergent in DNA sequence and demonstrate CF sensitivity to the human specific Pol I inhibitor, CX-5461. Finally, we show that the human Core Promoter Element (CPE) can functionally replace the yeast Core Element (CE) in vivo when aligned by conserved DNA structural features rather than DNA sequence. Together, these findings suggest that the yeast CF and the human ortholog Selectivity Factor 1 (SL1) use an evolutionarily conserved, structure-based mechanism to target DNA. Their shared mechanism may offer a new avenue in using yeast to explore current and future Pol I anti-cancer compounds.


Subject(s)
DNA, Ribosomal/genetics , RNA Polymerase I/genetics , Transcription Factors/genetics , Transcription, Genetic , Benzothiazoles/pharmacology , Conserved Sequence/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , Humans , Naphthyridines/pharmacology , Nucleic Acid Conformation/drug effects , Promoter Regions, Genetic/genetics , RNA Polymerase I/chemistry , Saccharomyces cerevisiae/genetics , Transcription Factors/chemistry
12.
Nucleic Acids Res ; 47(1): 310-327, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30407541

ABSTRACT

RNA polymerase (RNAP) III synthesizes tRNAs and other transcripts, and mutations to its subunits cause human disorders. The RNAP III subunit-heterodimer C37/53 functions in initiation, elongation and in termination-associated reinitiation with subunit C11. C37/53 is related to heterodimers associated with RNAPs I and II, and C11 is related to TFIIS and Rpa12.2, the active site RNA 3' cleavage factors for RNAPs II and I. Critical to termination is stability of the RNA:DNA hybrid bound in the active center, which is loose for RNAP III relative to other RNAPs. Here, we examined RNAP III lacking C37/53/C11 and various reconstituted forms during termination. First, we established a minimal terminator as 5T and 3A on the non-template and template DNA strands, respectively. We demonstrate that C11 stimulates termination, and does so independently of its RNA cleavage activity. We found that C37/53 sensitizes RNAP III termination to RNA:DNA hybrid strength and promotes RNA 3' end pairing/annealing with the template. The latter counteracts C11-insensitive arrest in the proximal part of the oligo(T)-tract, promoting oligo(rU:dA) extension toward greater hybrid instability and RNA release. The data also indicate that RNA 3' end engagement with the active site is a determinant of termination. Broader implications are also discussed.


Subject(s)
DNA-Directed RNA Polymerases/genetics , RNA Polymerase III/genetics , Transcription Termination, Genetic , Transcription, Genetic , Amino Acid Sequence/genetics , Catalytic Domain/genetics , DNA-Directed RNA Polymerases/chemistry , Humans , Protein Multimerization/genetics , RNA Cleavage/genetics , RNA Polymerase I/chemistry , RNA Polymerase I/genetics , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , RNA Polymerase III/chemistry , Saccharomyces cerevisiae/genetics
13.
Proc Natl Acad Sci U S A ; 115(45): E10652-E10661, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30355770

ABSTRACT

The carboxy-terminal domain (CTD) code encrypted within the Y1S2P3T4S5P6S7 heptad repeats of RNA polymerase II (Pol2) is deeply rooted in eukaryal biology. Key steps to deciphering the code are identifying the events in gene expression that are governed by individual "letters" and then defining a vocabulary of multiletter "words" and their meaning. Thr4 and Ser7 exert opposite effects on the fission yeast pho1 gene, expression of which is repressed under phosphate-replete conditions by transcription of an upstream flanking long noncoding RNA (lncRNA). Here we attribute the derepression of pho1 by a CTD-S7A mutation to precocious termination of lncRNA synthesis, an effect that is erased by mutations of cleavage-polyadenylation factor (CPF) subunits Ctf1, Ssu72, Ppn1, Swd22, and Dis2 and termination factor Rhn1. By contrast, a CTD-T4A mutation hyperrepresses pho1, as do CPF subunit and Rhn1 mutations, implying that T4A reduces lncRNA termination. Moreover, CTD-T4A is synthetically lethal with ppn1∆ and swd22∆, signifying that Thr4 and the Ppn1•Swd22 module play important, functionally redundant roles in promoting Pol2 termination. We find that Ppn1 and Swd22 become essential for viability when the CTD array is curtailed and that S7A overcomes the need for Ppn1•Swd22 in the short CTD context. Mutational synergies highlight redundant essential functions of (i) Ppn1•Swd22 and Rhn1, (ii) Ppn1•Swd22 and Ctf1, and (iii) Ssu72 and Dis2 phosphatases. CTD alleles Y1F, S2A, and T4A have overlapping synthetic lethalities with ppn1∆ and swd22∆, suggesting that Tyr1-Ser2-Thr4 form a three-letter CTD word that abets termination, with Rhn1 being a likely "reader" of this word.


Subject(s)
Homeostasis , Phosphates/metabolism , RNA Polymerase I/metabolism , Schizosaccharomyces/metabolism , Mutation , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Protein Domains , RNA Polymerase I/chemistry , Signal Transduction
14.
Proc Natl Acad Sci U S A ; 115(36): 8972-8977, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30127008

ABSTRACT

RNA polymerase I (Pol I) transcribes ribosomal DNA (rDNA) to produce the ribosomal RNA (rRNA) precursor, which accounts for up to 60% of the total transcriptional activity in growing cells. Pol I monitors rDNA integrity and influences cell survival, but little is known about how this enzyme processes UV-induced lesions. We report the electron cryomicroscopy structure of Pol I in an elongation complex containing a cyclobutane pyrimidine dimer (CPD) at a resolution of 3.6 Å. The structure shows that the lesion induces an early translocation intermediate exhibiting unique features. The bridge helix residue Arg1015 plays a major role in CPD-induced Pol I stalling, as confirmed by mutational analysis. These results, together with biochemical data presented here, reveal the molecular mechanism of Pol I stalling by CPD lesions, which is distinct from Pol II arrest by CPD lesions. Our findings open the avenue to unravel the molecular mechanisms underlying cell endurance to lesions on rDNA.


Subject(s)
DNA Damage , DNA, Fungal/chemistry , DNA, Ribosomal/chemistry , RNA Polymerase I/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Ultraviolet Rays , DNA, Fungal/metabolism , DNA, Ribosomal/metabolism , RNA Polymerase I/metabolism , Saccharomyces cerevisiae Proteins/metabolism
15.
Biophys J ; 114(11): 2507-2515, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29874602

ABSTRACT

Despite sharing a highly conserved core architecture with their prokaryotic counterparts, eukaryotic multisubunit RNA polymerases (Pols) have undergone structural divergence and biological specialization. Interesting examples of structural divergence are the A12.2 and C11 subunits of Pols I and III, respectively. Whereas all known cellular Pols possess cognate protein factors that stimulate cleavage of the nascent RNA, Pols I and III have incorporated their cleavage factors as bona fide subunits. Although it is not yet clear why these polymerases have incorporated their cleavage factors as subunits, a picture is emerging that identifies roles for these subunits beyond providing nucleolytic activity. Specifically, it appears that both A12.2 and C11 are required for efficient termination of transcription by Pols I and III. Given that termination involves destabilization of the elongation complex (EC), we tested whether A12.2 influences stability of the Pol I EC. Using, to our knowledge, a novel assay to measure EC dissociation kinetics, we have determined that A12.2 is an intrinsic destabilizer of the Pol I EC. In addition, the salt concentration dependence of Pol I EC dissociation kinetics suggests that A12.2 alters electrostatic interactions within the EC. Importantly, these data present a mechanistic basis for the requirement of A12.2 in Pol I termination. Combined with recent work demonstrating the direct involvement of A12.2 in Pol I nucleotide incorporation, this study further supports the concept that A12.2 cannot be viewed solely as a cleavage factor.


Subject(s)
Protein Subunits/metabolism , RNA Polymerase I/chemistry , RNA Polymerase I/metabolism , Base Sequence , Enzyme Stability , RNA/genetics , RNA/metabolism , Saccharomyces cerevisiae/enzymology
16.
Annu Rev Biophys ; 47: 425-446, 2018 05 20.
Article in English | MEDLINE | ID: mdl-29792819

ABSTRACT

RNA polymerases I and II (Pol I and Pol II) are the eukaryotic enzymes that catalyze DNA-dependent synthesis of ribosomal RNA and messenger RNA, respectively. Recent work shows that the transcribing forms of both enzymes are similar and the fundamental mechanisms of RNA chain elongation are conserved. However, the mechanisms of transcription initiation and its regulation differ between Pol I and Pol II. Recent structural studies of Pol I complexes with transcription initiation factors provided insights into how the polymerase recognizes its specific promoter DNA, how it may open DNA, and how initiation may be regulated. Comparison with the well-studied Pol II initiation system reveals a distinct architecture of the initiation complex and visualizes promoter- and gene-class-specific aspects of transcription initiation. On the basis of new structural studies, we derive a model of the Pol I transcription cycle and provide a molecular movie of Pol I transcription that can be used for teaching.


Subject(s)
RNA Polymerase II/chemistry , RNA Polymerase I/chemistry , Transcription, Genetic/genetics , Humans
17.
Nature ; 553(7688): 301-306, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29345637

ABSTRACT

RNA polymerase (Pol) III transcribes essential non-coding RNAs, including the entire pool of transfer RNAs, the 5S ribosomal RNA and the U6 spliceosomal RNA, and is often deregulated in cancer cells. The initiation of gene transcription by Pol III requires the activity of the transcription factor TFIIIB to form a transcriptionally active Pol III preinitiation complex (PIC). Here we present electron microscopy reconstructions of Pol III PICs at 3.4-4.0 Å and a reconstruction of unbound apo-Pol III at 3.1 Å. TFIIIB fully encircles the DNA and restructures Pol III. In particular, binding of the TFIIIB subunit Bdp1 rearranges the Pol III-specific subunits C37 and C34, thereby promoting DNA opening. The unwound DNA directly contacts both sides of the Pol III cleft. Topologically, the Pol III PIC resembles the Pol II PIC, whereas the Pol I PIC is more divergent. The structures presented unravel the molecular mechanisms underlying the first steps of Pol III transcription and also the general conserved mechanisms of gene transcription initiation.


Subject(s)
RNA Polymerase III/metabolism , RNA Polymerase III/ultrastructure , Transcription Initiation, Genetic , Cryoelectron Microscopy , DNA/chemistry , DNA/metabolism , DNA/ultrastructure , Models, Molecular , Nucleic Acid Conformation , Promoter Regions, Genetic , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA Polymerase I/chemistry , RNA Polymerase II/chemistry , RNA Polymerase III/chemistry , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure , Templates, Genetic , Transcription Factor TFIIIB/chemistry , Transcription Factor TFIIIB/metabolism , Transcription Factor TFIIIB/ultrastructure , Transcription Factors, TFII/chemistry
18.
Biochemistry ; 56(42): 5654-5662, 2017 10 24.
Article in English | MEDLINE | ID: mdl-28846843

ABSTRACT

All cellular RNA polymerases are influenced by protein factors that stimulate RNA polymerase-catalyzed cleavage of the nascent RNA. Despite divergence in amino acid sequence, these so-called "cleavage factors" appear to share a common mechanism of action. Cleavage factors associate with the polymerase through a conserved structural element of the polymerase known as the secondary channel or pore. This mode of association enables the cleavage factor to reach through the secondary channel into the polymerase active site to reorient the active site divalent metal ions. This reorientation converts the polymerase active site into a nuclease active site. Interestingly, eukaryotic RNA polymerases I and III (Pols I and III, respectively) have incorporated their cleavage factors as bona fide subunits known as A12.2 and C11, respectively. Although it is clear that A12.2 and C11 dramatically stimulate the polymerase's cleavage activity, it is not known if or how these subunits affect the polymerization mechanism. In this work we have used transient-state kinetic techniques to characterize a Pol I isoform lacking A12.2. Our data clearly demonstrate that the A12.2 subunit profoundly affects the kinetics and energetics of the elementary steps of Pol I-catalyzed nucleotide incorporation. Given the high degree of conservation between polymerase-cleavage factor interactions, these data indicate that cleavage factor-modulated nucleotide incorporation mechanisms may be common to all cellular RNA polymerases.


Subject(s)
Nucleotides/chemistry , RNA Polymerase I/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , mRNA Cleavage and Polyadenylation Factors/chemistry , Kinetics , Nucleotides/metabolism , RNA Polymerase I/metabolism , RNA Polymerase III/chemistry , RNA Polymerase III/metabolism , Saccharomyces cerevisiae Proteins/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism
19.
Curr Opin Struct Biol ; 47: 88-94, 2017 12.
Article in English | MEDLINE | ID: mdl-28743025

ABSTRACT

The majority of non-protein-coding RNAs present in eukaryotic cells comprises rRNAs, tRNAs and U6 snRNA that are involved in protein biosynthesis and are synthesized by DNA-dependent-RNA polymerase I and III. The transcription cycle (initiation, elongation and termination) has similar principles in all three nuclear RNA polymerases with specific features that are reflected back in their structures. Recently, owing to the 'resolution revolution' in electron cryo-microscopy, there has been a significant advancement in the understanding of these molecular machines. Here, we highlight the structure-function adaptation in specificity and activity of these molecular machines and present parallels and distinctions between their transcription mechanisms.


Subject(s)
RNA Polymerase III/chemistry , RNA Polymerase III/metabolism , RNA Polymerase I/chemistry , RNA Polymerase I/metabolism , Cryoelectron Microscopy , Models, Molecular , Protein Binding , Protein Conformation , Protein Subunits , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Structure-Activity Relationship , Transcription Initiation, Genetic
20.
EMBO J ; 36(18): 2698-2709, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28739580

ABSTRACT

In eukaryotic cells, RNA polymerase I (Pol I) synthesizes precursor ribosomal RNA (pre-rRNA) that is subsequently processed into mature rRNA. To initiate transcription, Pol I requires the assembly of a multi-subunit pre-initiation complex (PIC) at the ribosomal RNA promoter. In yeast, the minimal PIC includes Pol I, the transcription factor Rrn3, and Core Factor (CF) composed of subunits Rrn6, Rrn7, and Rrn11. Here, we present the cryo-EM structure of the 18-subunit yeast Pol I PIC bound to a transcription scaffold. The cryo-EM map reveals an unexpected arrangement of the DNA and CF subunits relative to Pol I. The upstream DNA is positioned differently than in any previous structures of the Pol II PIC. Furthermore, the TFIIB-related subunit Rrn7 also occupies a different location compared to the Pol II PIC although it uses similar interfaces as TFIIB to contact DNA. Our results show that although general features of eukaryotic transcription initiation are conserved, Pol I and Pol II use them differently in their respective transcription initiation complexes.


Subject(s)
RNA Polymerase I/chemistry , RNA Polymerase I/metabolism , Saccharomyces cerevisiae/enzymology , Transcription, Genetic , Cryoelectron Microscopy , DNA, Fungal/metabolism , Models, Molecular , Protein Conformation , Protein Multimerization , RNA, Ribosomal/biosynthesis , Saccharomyces cerevisiae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...