Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
PLoS Biol ; 20(3): e3001548, 2022 03.
Article in English | MEDLINE | ID: mdl-35239649

ABSTRACT

Commitment to cell division at the end of G1 phase, termed Start in the budding yeast Saccharomyces cerevisiae, is strongly influenced by nutrient availability. To identify new dominant activators of Start that might operate under different nutrient conditions, we screened a genome-wide ORF overexpression library for genes that bypass a Start arrest caused by absence of the G1 cyclin Cln3 and the transcriptional activator Bck2. We recovered a hypothetical gene YLR053c, renamed NRS1 for Nitrogen-Responsive Start regulator 1, which encodes a poorly characterized 108 amino acid microprotein. Endogenous Nrs1 was nuclear-localized, restricted to poor nitrogen conditions, induced upon TORC1 inhibition, and cell cycle-regulated with a peak at Start. NRS1 interacted genetically with SWI4 and SWI6, which encode subunits of the main G1/S transcription factor complex SBF. Correspondingly, Nrs1 physically interacted with Swi4 and Swi6 and was localized to G1/S promoter DNA. Nrs1 exhibited inherent transactivation activity, and fusion of Nrs1 to the SBF inhibitor Whi5 was sufficient to suppress other Start defects. Nrs1 appears to be a recently evolved microprotein that rewires the G1/S transcriptional machinery under poor nitrogen conditions.


Subject(s)
G1 Phase/genetics , Gene Expression Regulation, Fungal , Nitrogen/metabolism , S Phase/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Cell Division/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Immunoblotting , Protein Binding , RNA-Seq/methods , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Amino Acid , Transcription Factors/genetics , Transcription Factors/metabolism
3.
J Cell Biol ; 219(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32744610

ABSTRACT

In budding yeast, the transcription factors SBF and MBF activate a large program of gene expression in late G1 phase that underlies commitment to cell division, termed Start. SBF/MBF are limiting with respect to target promoters in small G1 phase cells and accumulate as cells grow, raising the questions of how SBF/MBF are dynamically distributed across the G1/S regulon and how this impacts the Start transition. Super-resolution Photo-Activatable Localization Microscopy (PALM) mapping of the static positions of SBF/MBF subunits in fixed cells revealed each transcription factor was organized into discrete clusters containing approximately eight copies regardless of cell size and that the total number of clusters increased as cells grew through G1 phase. Stochastic modeling using reasonable biophysical parameters recapitulated growth-dependent SBF/MBF clustering and predicted TF dynamics that were confirmed in live cell PALM experiments. This spatio-temporal organization of SBF/MBF may help coordinate activation of G1/S regulon and the Start transition.


Subject(s)
G1 Phase/genetics , S Phase/genetics , Transcription Factors/genetics , Cell Division/genetics , Gene Expression Regulation, Fungal/genetics , Promoter Regions, Genetic/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomycetales/genetics
4.
Methods Mol Biol ; 809: 593-607, 2012.
Article in English | MEDLINE | ID: mdl-22113302

ABSTRACT

In most eukaryotes, the generation of the 3' end and transcription termination are initiated by cleavage of the pre-mRNA upstream of the polyadenylation site. This cleavage initiates 5'-3' degradation of the 3' end cleavage product by the exoribonuclease Rat1p leading to the dissociation of the RNA polymerase II (RNAPII) complex. The Rat1p-dependent transcription termination was also shown to be initiated by a polyadenylation-independent cleavage performed by the double-stranded RNA-specific ribonuclease (RNase) III (Rnt1p) suggesting that the majority of transcription termination events are RNase dependent. Therefore, it became essential for future studies on transcription termination to carefully consider both the nature of the RNase-dependent RNA transcripts and the association pattern of the RNAPII with the transcriptional unit. Here, we present methods allowing the evaluation of the impact of yeast RNases on the 3' end formation and their contribution to transcription termination. Northern blot analysis of transcripts generated downstream of known genes in the absence of RNases identifies potential transcription termination sites while chromatin immunoprecipitation of RNAPII differentiates between termination- and transcription-independent processing events.


Subject(s)
Transcription, Genetic/genetics , Blotting, Northern , Chromatin Immunoprecipitation , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism
5.
Mol Cell ; 36(1): 99-109, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19818713

ABSTRACT

Transcription termination of messenger RNA (mRNA) is normally achieved by polyadenylation followed by Rat1p-dependent 5'-3' exoribonuleolytic degradation of the downstream transcript. Here we show that the yeast ortholog of the dsRNA-specific ribonuclease III (Rnt1p) may trigger Rat1p-dependent termination of RNA transcripts that fail to terminate near polyadenylation signals. Rnt1p cleavage sites were found downstream of several genes, and the deletion of RNT1 resulted in transcription readthrough. Inactivation of Rat1p impaired Rnt1p-dependent termination and resulted in the accumulation of 3' end cleavage products. These results support a model for transcription termination in which cotranscriptional cleavage by Rnt1p provides access for exoribonucleases in the absence of polyadenylation signals.


Subject(s)
Ribonuclease III/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/physiology , Terminator Regions, Genetic/physiology , Transcription, Genetic/physiology , Actins/genetics , Acyltransferases/genetics , Acyltransferases/metabolism , Alcohol Dehydrogenase/genetics , DNA/metabolism , Exoribonucleases/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Polyadenylation/physiology , Promoter Regions, Genetic/genetics , Protein Binding/genetics , RNA Polymerase II/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribosomal Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
6.
J Biol Chem ; 282(7): 4373-4381, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17158880

ABSTRACT

In bakers' yeast, in vivo telomerase activity requires a ribonucleoprotein (RNP) complex with at least four associated proteins (Est2p, Est1p, Est3p, and Cdc13p) and one RNA species (Tlc1). The function of telomerase in maintaining chromosome ends, called telomeres, is tightly regulated and linked to the cell cycle. However, the mechanisms that regulate the expression of individual components of telomerase are poorly understood. Here we report that yeast RNase III (Rnt1p), a double-stranded RNA-specific endoribonuclease, regulates the expression of telomerase subunits and is required for maintaining normal telomere length. Deletion or inactivation of RNT1 induced the expression of Est1, Est2, Est3, and Tlc1 RNAs and increased telomerase activity, leading to elongation of telomeric repeat tracts. In silico analysis of the different RNAs coding for the telomerase subunits revealed a canonical Rnt1p cleavage site near the 3' end of Est1 mRNA. This predicted structure was cleaved by Rnt1p and its disruption abolished cleavage in vitro. Mutation of the Rnt1p cleavage signal in vivo impaired the cell cycle-dependent degradation of Est1 mRNA without affecting its steady-state level. These results reveal a new mechanism that influences telomeres length by controlling the expression of the telomerase subunits.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Fungal , Ribonuclease III/metabolism , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Telomerase/biosynthesis , 3' Untranslated Regions/genetics , 3' Untranslated Regions/metabolism , Cell Cycle/genetics , Chromosomes, Fungal/genetics , Chromosomes, Fungal/metabolism , Enzyme Activation/genetics , Gene Deletion , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Fungal/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Stability/genetics , RNA, Fungal/genetics , RNA, Fungal/metabolism , Ribonuclease III/genetics , Ribonucleoproteins/biosynthesis , Ribonucleoproteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Telomerase/genetics , Telomere/genetics , Telomere/metabolism
7.
J Mol Biol ; 363(2): 322-31, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16979185

ABSTRACT

RNase III enzymes are a highly conserved family of proteins that specifically cleave double-stranded RNA (dsRNA). These proteins are involved in a variety of cellular functions, including the processing of many non-coding RNAs, mRNA decay, and RNA interference. In yeast Rnt1p, a dsRNA-binding domain (dsRBD) recognizes its substrate by interacting with stems capped with conserved AGNN tetraloops. The enzyme uses the tetraloop to cut 14nt to 16nt away into the stem in a ruler-like mechanism. The solution structure of Rnt1p dsRBD complexed to one of its small nucleolar (sno) RNA substrate revealed non-sequence-specific contacts with the sugar-phosphate backbone in the minor groove of the AGNN fold and the two non-conserved tetraloop nucleotides. Recently, a new form of Rnt1p substrates lacking the conserved AGNN sequence but instead harboring an AAGU tetraloop was found at the 5' end of snoRNA 48 precursor. Here, we report the solution structure of this hairpin capped with an AAGU tetraloop. Some of the stacking interactions and the position of the turn in the sugar-phosphate backbone are similar to the one observed in the AGNN loop structure; however, the AAGU sequence adopts a different conformation. The most striking difference was found at the 3' end of the loop where Rnt1p interacts with AGNN substrates. The last nucleotide is extruded from the AAGU tetraloop structure in contrast to the compact AGNN fold. The AAGU hairpin structure suggests that Rnt1p recognizes substrates with different tetraloop structures, indicating that the structural repertoire specifically recognized by Rnt1p is larger than previously anticipated.


Subject(s)
Base Sequence , Nucleic Acid Conformation , Ribonuclease III , Saccharomyces cerevisiae Proteins , Animals , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , RNA, Small Nucleolar/chemistry , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , Ribonuclease III/chemistry , Ribonuclease III/genetics , Ribonuclease III/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity
8.
J Mol Biol ; 363(2): 332-44, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16962133

ABSTRACT

RNase III enzymes form a conserved family of proteins that specifically cleave double-stranded (dsRNA). These proteins are involved in a variety of cellular functions, including the processing of many non-coding RNAs, mRNA decay, and RNA interference. Yeast RNase III (Rnt1p) selects its substrate by recognizing the structure generated by a conserved NGNN tetraloop (G2-loop). Mutations of the invariant guanosine stringently inhibit binding and cleavage of all known Rnt1p substrates. Surprisingly, we have found that the 5' end of small nucleolar RNA 48 is processed by Rnt1p in the absence of a G2-loop. Instead, biochemical and structural analyses revealed that cleavage, in this case, is directed by a hairpin capped with an AAGU tetraloop, with a preferred adenosine in the first position (A1-loop). Chemical probing indicated that A1-loops adopt a distinct structure that varies at the 3' end where Rnt1p interacts with G2-loops. Consistently, chemical footprinting and chemical interference assays indicate that Rnt1p binds to G2 and A1-loops using different sets of nucleotides. Also, cleavage and binding assays showed that the N-terminal domain of Rnt1p aids selection of A1-capped hairpins. Together, the results suggest that Rnt1p recognizes at least two distinct classes of tetraloops using flexible protein RNA interactions. This underscores the capacity of double-stranded RNA binding proteins to use several recognition motifs for substrate identification.


Subject(s)
Base Sequence , Nucleic Acid Conformation , Ribonuclease III , Saccharomyces cerevisiae Proteins , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , RNA Caps/chemistry , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , Ribonuclease III/chemistry , Ribonuclease III/genetics , Ribonuclease III/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity
9.
Mol Cell Biol ; 25(8): 2981-94, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15798187

ABSTRACT

In Saccharomyces cerevisiae, the maturation of both pre-rRNA and pre-small nucleolar RNAs (pre-snoRNAs) involves common factors, thereby providing a potential mechanism for the coregulation of snoRNA and rRNA synthesis. In this study, we examined the global impact of the double-stranded-RNA-specific RNase Rnt1p, which is required for pre-rRNA processing, on the maturation of all known snoRNAs. In silico searches for Rnt1p cleavage signals, and genome-wide analysis of the Rnt1p-dependent expression profile, identified seven new Rnt1p substrates. Interestingly, two of the newly identified Rnt1p-dependent snoRNAs, snR39 and snR59, are located in the introns of the ribosomal protein genes RPL7A and RPL7B. In vitro and in vivo experiments indicated that snR39 is normally processed from the lariat of RPL7A, suggesting that the expressions of RPL7A and snR39 are linked. In contrast, snR59 is produced by a direct cleavage of the RPL7B pre-mRNA, indicating that a single pre-mRNA transcript cannot be spliced to produce a mature RPL7B mRNA and processed by Rnt1p to produce a mature snR59 simultaneously. The results presented here reveal a new role of yeast RNase III in the processing of intron-encoded snoRNAs that permits independent regulation of the host mRNA and its associated snoRNA.


Subject(s)
RNA Splicing/physiology , RNA, Small Nucleolar/metabolism , Ribonuclease III/physiology , Ribosomal Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Base Sequence , Computational Biology , Genomics , Introns/genetics , Molecular Sequence Data , Nucleic Acid Conformation , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing/genetics , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Small Nucleolar/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism , Substrate Specificity , Transcription, Genetic
10.
RNA ; 10(12): 1946-56, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15525710

ABSTRACT

Eukaryotes have two types of ribosomes containing either 5.8SL or 5.8SS rRNA that are produced by alternative pre-rRNA processing. The exact processing pathway for the minor 5.8SL rRNA species is poorly documented. We have previously shown that the trans-acting factor Rrp5p and the RNA exonuclease Rex4p genetically interact to influence the ratio between the two forms of 5.8S rRNA in the yeast Saccharomyces cerevisiae. Here we report a further analysis of ITS1 processing in various yeast mutants that reveals genetic interactions between, on the one hand, Rrp5p and RNase MRP, the endonuclease required for 5.8SS rRNA synthesis, and, on the other, Rex4p, the RNase III homolog Rnt1p, and the debranching enzyme Dbr1p. Yeast cells carrying a temperature-sensitive mutation in RNase MRP (rrp2-1) exhibit a pre-rRNA processing phenotype very similar to that of the previously studied rrp5-33 mutant: ITS2 processing precedes ITS1 processing, 5.8SL rRNA becomes the major species, and ITS1 is processed at the recently reported novel site A4 located midway between sites A2 and A3. As in the rrp5-Delta3 mutant, all of these phenotypical processing features disappear upon inactivation of the REX4 gene. Moreover, inactivation of the DBR1 gene in rrp2-1, or the RNT1 gene in rrp5-Delta3 mutant cells also negates the effects of the original mutation on pre-rRNA processing. These data link a total of three RNA catabolic enzymes, Rex4p, Rnt1p, and Dbr1p, to ITS1 processing and the relative production of 5.8SS and 5.8SL rRNA. A possible model for the indirect involvement of the three enzymes in yeast pre-rRNA processing is discussed.


Subject(s)
RNA Precursors/metabolism , RNA, Fungal/metabolism , Ribonucleases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Genes, Fungal , Models, Biological , Mutation , Phenotype , RNA Nucleotidyltransferases/genetics , RNA Nucleotidyltransferases/metabolism , RNA Precursors/genetics , RNA Processing, Post-Transcriptional , RNA Splicing , RNA, Fungal/genetics , RNA, Ribosomal, 5.8S/genetics , RNA, Ribosomal, 5.8S/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Ribonucleases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
11.
Mol Biol Cell ; 15(7): 3015-30, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15090619

ABSTRACT

Members of the double-stranded RNA-specific ribonuclease III (RNase III) family were shown to affect cell division and chromosome segregation, presumably through an RNA interference-dependent mechanism. Here, we show that in Saccharomyces cerevisiae, where the RNA interference machinery is not conserved, an orthologue of RNase III (Rnt1p) is required for progression of the cell cycle and nuclear division. The deletion of Rnt1p delayed cells in both G1 and G2/M phases of the cell cycle. Nuclear division and positioning at the bud neck were also impaired in Deltarnt1 cells. The cell cycle defects were restored by the expression of catalytically inactive Rnt1p, indicating that RNA cleavage is not essential for cell cycle progression. Rnt1p was found to exit from the nucleolus to the nucleoplasm in the G2/M phase, and perturbation of its localization pattern delayed the progression of cell division. A single mutation in the Rnt1p N-terminal domain prevented its accumulation in the nucleoplasm and slowed exit from mitosis without any detectable effects on RNA processing. Together, the data reveal a new role for a class II RNase III in the cell cycle and suggest that at least some members of the RNase III family possess catalysis-independent functions.


Subject(s)
Cell Nucleus Division , Cell Nucleus/enzymology , Ribonuclease III/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/enzymology , Catalytic Domain/genetics , Cell Cycle/genetics , Cell Nucleolus/metabolism , Cell Nucleolus/ultrastructure , Cell Nucleus Division/genetics , Microtubules/metabolism , Nuclear Localization Signals/analysis , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Point Mutation/genetics , RNA/metabolism , Ribonuclease III/analysis , Ribonuclease III/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/immunology , Saccharomyces cerevisiae Proteins/analysis , Saccharomyces cerevisiae Proteins/genetics , Sequence Deletion/genetics
12.
J Mol Biol ; 327(5): 985-1000, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12662924

ABSTRACT

Yeast Rnt1p is a member of the double-stranded RNA (dsRNA) specific RNase III family of endoribonucleases involved in RNA processing and RNA interference (RNAi). Unlike other RNase III enzymes, which recognize a variety of RNA duplexes, Rnt1p cleaves specifically RNA stems capped with the conserved AGNN tetraloop. This unusual substrate specificity challenges the established dogma for substrate selection by RNase III and questions the dsRNA contribution to recognition by Rnt1p. Here we show that the dsRNA sequence adjacent to the tetraloop regulates Rnt1p cleavage by interfering with RNA binding. In context, sequences surrounding the cleavage site directly influence the cleavage efficiency. Introduction of sequences that stabilize the RNA helix enhanced binding while reducing the turnover rate indicating that, unlike the tetraloop, Rnt1p binding to the dsRNA helix may become rate-limiting. These results suggest that Rnt1p activity is strictly regulated by a combination of primary and tertiary structural elements allowing a substrate-specific binding and cleavage efficiency.


Subject(s)
Endoribonucleases/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/enzymology , Base Sequence , Circular Dichroism , Hydrolysis , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Conformation , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism , RNA, Fungal/chemistry , RNA, Fungal/metabolism , Ribonuclease III , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...