Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
PLoS Genet ; 16(12): e1009133, 2020 12.
Article in English | MEDLINE | ID: mdl-33382702

ABSTRACT

Transcriptional regulation of the Saccharomyces cerevisiae HO gene is highly complex, requiring a balance of multiple activating and repressing factors to ensure that only a few transcripts are produced in mother cells within a narrow window of the cell cycle. Here, we show that the Ash1 repressor associates with two DNA sequences that are usually concealed within nucleosomes in the HO promoter and recruits the Tup1 corepressor and the Rpd3 histone deacetylase, both of which are required for full repression in daughters. Genome-wide ChIP identified greater than 200 additional sites of co-localization of these factors, primarily within large, intergenic regions from which they could regulate adjacent genes. Most Ash1 binding sites are in nucleosome depleted regions (NDRs), while a small number overlap nucleosomes, similar to HO. We demonstrate that Ash1 binding to the HO promoter does not occur in the absence of the Swi5 transcription factor, which recruits coactivators that evict nucleosomes, including the nucleosomes obscuring the Ash1 binding sites. In the absence of Swi5, artificial nucleosome depletion allowed Ash1 to bind, demonstrating that nucleosomes are inhibitory to Ash1 binding. The location of binding sites within nucleosomes may therefore be a mechanism for limiting repressive activity to periods of nucleosome eviction that are otherwise associated with activation of the promoter. Our results illustrate that activation and repression can be intricately connected, and events set in motion by an activator may also ensure the appropriate level of repression and reset the promoter for the next activation cycle.


Subject(s)
Deoxyribonucleases, Type II Site-Specific/genetics , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Deoxyribonucleases, Type II Site-Specific/metabolism , Gene Expression Regulation, Fungal , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Nuclear Proteins/genetics , Protein Binding , Repressor Proteins/genetics , Saccharomyces cerevisiae , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
2.
Nucleic Acids Res ; 48(19): 10877-10889, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33010153

ABSTRACT

The Saccharomyces cerevisiae HO gene is a model regulatory system with complex transcriptional regulation. Budding yeast divide asymmetrically and HO is expressed only in mother cells where a nucleosome eviction cascade along the promoter during the cell cycle enables activation. HO expression in daughter cells is inhibited by high concentration of Ash1 in daughters. To understand how Ash1 represses transcription, we used a myo4 mutation which boosts Ash1 accumulation in both mothers and daughters and show that Ash1 inhibits promoter recruitment of SWI/SNF and Gcn5. We show Ash1 is also required for the efficient nucleosome repopulation that occurs after eviction, and the strongest effects of Ash1 are seen when Ash1 has been degraded and at promoter locations distant from where Ash1 bound. Additionally, we defined a specific nucleosome/nucleosome-depleted region structure that restricts HO activation to one of two paralogous DNA-binding factors. We also show that nucleosome eviction occurs bidirectionally over a large distance. Significantly, eviction of the more distant nucleosomes is dependent upon the FACT histone chaperone, and FACT is recruited to these regions when eviction is beginning. These last observations, along with ChIP experiments involving the SBF factor, suggest a long-distance loop transiently forms at the HO promoter.


Subject(s)
DNA-Binding Proteins/metabolism , Deoxyribonucleases, Type II Site-Specific , Gene Expression Regulation, Fungal , High Mobility Group Proteins/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Transcriptional Elongation Factors/metabolism , Carrier Proteins/metabolism , Deoxyribonucleases, Type II Site-Specific/genetics , Deoxyribonucleases, Type II Site-Specific/metabolism , Histone Acetyltransferases/metabolism , Nucleosomes/metabolism , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins/genetics
3.
Proc Natl Acad Sci U S A ; 113(34): 9575-80, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27506791

ABSTRACT

The yeast HO endonuclease is expressed in late G1 in haploid mother cells to initiate mating-type interconversion. Cells can be arrested in G1 by nutrient deprivation or by pheromone exposure, but cells that resume cycling after nutrient deprivation or cyclin-dependent kinase (CDK) inactivation express HO in the first cell cycle, whereas HO is not expressed until the second cycle after release from pheromone arrest. Here, we show that transcription of a long noncoding RNA (lncRNA) mediates this differential response. The SBF and Mediator factors remain bound to the inactive promoter during arrest due to CDK inactivation, and these bound factors allow the cell to remember a transcriptional decision made before arrest. If the presence of mating pheromone indicates that this decision is no longer appropriate, a lncRNA originating at -2700 upstream of the HO gene is induced, and the transcription machinery displaces promoter-bound SBF, preventing HO transcription in the subsequent cell cycle. Further, we find that the displaced SBF is blocked from rebinding due to incorporation of its recognition sites within nucleosomes. Expressing the pHO-lncRNA in trans is ineffective, indicating that transcription in cis is required. Factor displacement during lncRNA transcription could be a general mechanism for regulating memory of previous events at promoters.


Subject(s)
Endonucleases/genetics , Gene Expression Regulation, Fungal , Promoter Regions, Genetic , RNA, Fungal/genetics , RNA, Long Noncoding/genetics , Saccharomyces cerevisiae/genetics , Base Sequence , Binding Sites , Endonucleases/metabolism , G1 Phase Cell Cycle Checkpoints , Mediator Complex/genetics , Mediator Complex/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Protein Binding , RNA, Fungal/biosynthesis , RNA, Long Noncoding/biosynthesis , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
4.
Genes Dev ; 25(23): 2429-35, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22156205

ABSTRACT

The Tup1-Cyc8 complex is responsible for repression of a large and diverse collection of genes in Saccharomyces cerevisiae. The predominant view has been that Tup1-Cyc8 functions as a corepressor, actively associating with regulatory proteins and organizing chromatin to block transcription. A new study by Wong and Struhl in this issue of Genes & Development (pp. 2525-2539) challenges nearly 20 years of models by demonstrating that Tup1-Cyc8 functions primarily as a shield to block DNA-binding proteins from recruiting transcriptional coactivators.


Subject(s)
Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription, Genetic
5.
Trends Biochem Sci ; 38(9): 467-75, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23870664

ABSTRACT

The chromatin structure at a promoter can define how a gene is regulated. Studies of two yeast genes expressed in the G1 phase of the cell cycle, HO and CLN2, have provided important paradigms for transcriptional regulation. Although the SBF (Swi4/Swi6 box factor) transcription factor activates both genes, the chromatin landscapes that regulate SBF binding are different. Specifically, the CLN2 promoter is constitutively available for SBF binding, whereas HO has a complex two-step promoter in which chromatin changes in one region allow SBF to bind at a downstream location. These studies reveal the role of chromatin in defining the regulatory properties of promoters.


Subject(s)
Fungal Proteins/metabolism , Yeasts/cytology , Yeasts/metabolism , Cell Cycle/genetics , Cell Cycle/physiology , Fungal Proteins/genetics , Gene Expression Regulation, Fungal , Promoter Regions, Genetic/genetics
6.
Mol Cell ; 34(4): 405-15, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19481521

ABSTRACT

Transcriptional activators and coactivators overcome repression by chromatin, but regulation of chromatin disassembly and coactivator binding to promoters is poorly understood. Activation of the yeast HO gene follows the sequential binding of both sequence-specific DNA-binding proteins and coactivators during the cell cycle. Here, we show that the nucleosome disassembly occurs in waves both along the length of the promoter and during the cell cycle. Different chromatin modifiers are required for chromatin disassembly at different regions of the promoter, with Swi/Snf, the FACT chromatin reorganizer, and the Asf1 histone chaperone each required for nucleosome eviction at distinct promoter regions. FACT and Asf1 both bind to upstream elements of the HO promoter well before the gene is transcribed. The Swi/Snf, SAGA, and Mediator coactivators bind first to the far upstream promoter region and subsequently to a promoter proximal region, and FACT and Asf1 are both required for this coactivator re-recruitment.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Deoxyribonucleases, Type II Site-Specific , High Mobility Group Proteins/metabolism , Molecular Chaperones/metabolism , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae , Transcriptional Elongation Factors/metabolism , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/genetics , Deoxyribonucleases, Type II Site-Specific/genetics , Deoxyribonucleases, Type II Site-Specific/metabolism , Gene Expression Regulation, Fungal , High Mobility Group Proteins/genetics , Molecular Chaperones/genetics , Nucleosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Elongation Factors/genetics
7.
Mol Cell ; 35(3): 365-76, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19683499

ABSTRACT

FACT has been proposed to function by displacing H2A-H2B dimers from nucleosomes to form hexasomes. Results described here with yeast FACT (yFACT) suggest instead that nucleosomes are reorganized to a form with the original composition but a looser, more dynamic structure. First, yFACT enhances hydroxyl radical accessibility and endonuclease digestion in vitro at sites throughout the nucleosome, not just in regions contacted by H2A-H2B. Accessibility increases dramatically, but the DNA remains partially protected. Second, increased nuclease sensitivity can occur without displacement of dimers from the nucleosome. Third, yFACT is required for eviction of nucleosomes from the GAL1-10 promoter during transcriptional activation in vivo, but the preferential reduction in dimer occupancy expected for hexasome formation is not observed. We propose that yFACT promotes a reversible transition between two nucleosomal forms, and that this activity contributes to the establishment and maintenance of the chromatin barrier as well as to overcoming it.


Subject(s)
Chromatin Assembly and Disassembly/physiology , DNA-Binding Proteins/physiology , High Mobility Group Proteins/physiology , Histones/metabolism , Nucleosomes/chemistry , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Transcriptional Elongation Factors/physiology , DNA, Fungal/chemistry , DNA, Fungal/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deoxyribonucleases, Type II Site-Specific/metabolism , Dimerization , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Models, Genetic , Models, Molecular , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Transcription Factors/physiology , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism
8.
Proc Natl Acad Sci U S A ; 110(34): 14012-7, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23836672

ABSTRACT

Eukaryotic gene regulation usually involves sequence-specific transcription factors and sequence-nonspecific cofactors. A large effort has been made to understand how these factors affect the average gene expression level among a population. However, little is known about how they regulate gene expression in individual cells. In this work, we address this question by mutating multiple factors in the regulatory pathway of the yeast HO promoter (HOpr) and probing the corresponding promoter activity in single cells using time-lapse fluorescence microscopy. We show that the HOpr fires in an "on/off" fashion in WT cells as well as in different genetic backgrounds. Many chromatin-related cofactors that affect the average level of HO expression do not actually affect the firing amplitude of the HOpr; instead, they affect the firing frequency among individual cell cycles. With certain mutations, the bimodal expression exhibits short-term epigenetic memory across the mitotic boundary. This memory is propagated in "cis" and reflects enhanced activator binding after a previous "on" cycle. We present evidence that the memory results from slow turnover of the histone acetylation marks.


Subject(s)
Deoxyribonucleases, Type II Site-Specific/metabolism , Epigenesis, Genetic/physiology , Gene Expression Regulation, Fungal/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Acetylation , Chromatin Immunoprecipitation , Deoxyribonucleases, Type II Site-Specific/genetics , Histones/metabolism , Microfluidic Analytical Techniques , Microscopy, Fluorescence , Mutagenesis , Promoter Regions, Genetic/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Stochastic Processes , Time-Lapse Imaging
9.
J Biol Chem ; 289(51): 35431-7, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25352596

ABSTRACT

The RTS1 gene encodes a subunit of the PP2A phosphatase that regulates cell cycle progression. Ace2 and Swi5 are cell cycle-regulated transcription factors, and we recently showed that phosphorylation of Ace2 and Swi5 is altered in an rts1 mutant. Here we examine expression of Ace2 and Swi5 target genes and find that an rts1 mutation markedly reduces expression of the HO gene. The decreased HO expression in an rts1 mutant is significantly restored by an additional ace2 mutation, a surprising result because HO is normally activated by Swi5 but not by Ace2. Ace2 normally accumulates only in daughter cells, and only activates transcription in daughters. However, in an rts1 mutant, Ace2 is present in both mother and daughter cells. One of the genes activated by Ace2 is ASH1, a protein that normally accumulates mostly in daughter cells; Ash1 is a transcriptional repressor, and it blocks HO expression in daughters. We show that in the rts1 mutant, Ace2 accumulation in mother cells results in Ash1 expression in mothers, and the Ash1 can now repress HO expression in mothers.


Subject(s)
DNA-Binding Proteins/genetics , Deoxyribonucleases, Type II Site-Specific/genetics , Protein Phosphatase 2/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Deoxyribonucleases, Type II Site-Specific/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Fungal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Mutation , Protein Phosphatase 2/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Saccharomyces cerevisiae Proteins/metabolism , Time-Lapse Imaging , Transcription Factors/metabolism
10.
EMBO J ; 28(21): 3378-89, 2009 Nov 04.
Article in English | MEDLINE | ID: mdl-19745812

ABSTRACT

Regulation of the CLN1 and CLN2 G1 cyclin genes controls cell cycle progression. The SBF activator binds to these promoters but is kept inactive by the Whi5 and Stb1 inhibitors. The Cdc28 cyclin-dependent kinase phosphorylates Whi5, ending the inhibition. Our chromatin immunoprecipitation (ChIP) experiments show that SBF, Whi5 and Stb1 recruit both Cdc28 and the Rpd3(L) histone deacetylase to CLN promoters, extending the analogy with mammalian G1 cyclin promoters in which Rb recruits histone deacetylases. Finally, we show that the SBF subunit Swi6 recruits the FACT chromatin reorganizer to SBF- and MBF-regulated genes. Mutations affecting FACT reduce the transient nucleosome eviction seen at these promoters during a normal cell cycle and also reduce expression. Temperature-sensitive mutations affecting FACT and Cdc28 can be suppressed by disruption of STB1 and WHI5, suggesting that one critical function of FACT and Cdc28 is overcoming chromatin repression at G1 cyclin promoters. Thus, SBF recruits complexes to promoters that either enhance (FACT) or repress (Rpd3L) accessibility to chromatin, and also recruits the kinase that activates START.


Subject(s)
Cyclin G/genetics , Histone Deacetylases/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , CDC28 Protein Kinase, S cerevisiae/genetics , CDC28 Protein Kinase, S cerevisiae/metabolism , Cell Cycle , Chromatin/metabolism , Cyclins/metabolism , Gene Expression Regulation, Fungal , Mutation , Nucleosomes/metabolism , Promoter Regions, Genetic , Protein Binding , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics
11.
J Biol Chem ; 286(40): 34809-19, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21840992

ABSTRACT

The yeast HO gene is tightly regulated, with multiple activators and coactivators needed to overcome repressive chromatin structures that form over this promoter. Coactivator binding is strongly interdependent, as loss of one factor sharply reduces recruitment of other factors. The Rpd3(L) histone deacetylase is recruited to HO at two distinct times during the cell cycle, first by Ash1 to the URS1 region of the promoter and then by SBF/Whi5/Stb1 to URS2. SBF itself is localized to only a subset of its potential binding sites in URS2, and this localization takes longer and is less robust than at other SBF target genes, suggesting that binding to the HO promoter is limited by chromatin structures that dynamically change as the cell cycle progresses. Ash1 only binds at the URS1 region of the promoter, but an ash1 mutation results in markedly increased binding of SBF and Rpd3(L) at URS2, some 450 bp distant from the site of Ash1 binding, suggesting these two regions of the promoter interact. An ash1 mutation also results in increased coactivator recruitment, Swi/Snf and Mediator localization in the absence of the normally required Gcn5 histone acetyltransferase, and HO expression even in the presence of a taf1 mutation affecting TFIID activity that otherwise blocks HO transcription. Ash1 therefore appears to play a central role in generating the strongly repressive environment at the HO promoter, which limits the binding of several coactivators at URS2 and TATA region.


Subject(s)
Chromatin/chemistry , Deoxyribonucleases, Type II Site-Specific/genetics , Deoxyribonucleases, Type II Site-Specific/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , CDC28 Protein Kinase, S cerevisiae/genetics , Cell Cycle , Chromatin/metabolism , Endonucleases/metabolism , Gene Expression Regulation, Fungal , Histone Deacetylases/metabolism , Mutation , Nucleosomes/metabolism , Promoter Regions, Genetic , Protein Binding , Repressor Proteins/genetics , TATA Box , Transcription, Genetic
12.
Biochim Biophys Acta ; 1799(1-2): 175-80, 2010.
Article in English | MEDLINE | ID: mdl-20123079

ABSTRACT

The small Nhp6 protein from budding yeast is an abundant protein that binds DNA non-specifically and bends DNA sharply. It contains only a single HMGB domain that binds DNA in the minor groove and a basic N-terminal extension that wraps around DNA to contact the major groove. This review describes the genetic and biochemical experiments that indicate Nhp6 functions in promoting RNA pol III transcription, in formation of preinitiation complexes at promoters transcribed by RNA pol II, and in facilitating the activity of chromatin modifying complexes. The FACT complex may provide a paradigm for how Nhp6 functions with chromatin factors, as Nhp6 allows Spt16-Pob3 to bind to and reorganize nucleosomes in vitro.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , HMGN Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , DNA, Fungal/metabolism , DNA-Binding Proteins/chemistry , HMGN Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Transcription, Genetic
13.
EMBO J ; 26(20): 4324-34, 2007 Oct 17.
Article in English | MEDLINE | ID: mdl-17898805

ABSTRACT

Transcription factors with identical DNA-binding specificity often activate different genes in vivo. Yeast Ace2 and Swi5 are such activators, with targets we classify as Swi5-only, Ace2-only, or both. We define two unique regulatory modes. Ace2 and Swi5 both bind in vitro to Swi5-only genes such as HO, but only Swi5 binds and activates in vivo. In contrast, Ace2 and Swi5 both bind in vivo to Ace2-only genes, such as CTS1, but promoter-bound Swi5 fails to activate. We show that activation by Swi5 is prevented by the binding of the Forkhead factors Fkh1 and Fkh2, which recruit the Rpd3(Large) histone deacetylase complex to the CTS1 promoter. Global analysis shows that all Ace2-only genes are bound by both Ace2 and Swi5, and also by Fkh1/2. Genes normally activated by either Ace2 or Swi5 can be converted to Ace2-only genes by the insertion of Fkh-binding sites. Thus Fkh proteins, which function initially to activate SWI5 and ACE2, subsequently function as Swi5-specific antiactivators.


Subject(s)
DNA-Binding Proteins/chemistry , Forkhead Transcription Factors/metabolism , Fungal Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Transcription Factors/metabolism , Acetylation , Cell Cycle , Chromatin Immunoprecipitation , DNA/chemistry , Gene Expression Regulation, Fungal , Histones/chemistry , Open Reading Frames , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Saccharomyces cerevisiae/metabolism , Transcription Factors/chemistry
14.
Genetics ; 219(2)2021 10 02.
Article in English | MEDLINE | ID: mdl-34849878

ABSTRACT

The Tup1-Cyc8 corepressor complex of Saccharomyces cerevisiae is recruited to promoters by DNA-binding proteins to repress transcription of genes, including the a-specific mating-type genes. We report here a tup1(S649F) mutant that displays mating irregularities and an α-predominant growth defect. RNA-Seq and ChIP-Seq were used to analyze gene expression and Tup1 occupancy changes in mutant vs wild type in both a and α cells. Increased Tup1(S649F) occupancy tended to occur upstream of upregulated genes, whereas locations with decreased occupancy usually did not show changes in gene expression, suggesting this mutant not only loses corepressor function but also behaves as a coactivator. Based upon studies demonstrating a dual role of Tup1 in both repression and activation, we postulate that the coactivator function of Tup1(S649F) results from diminished interaction with repressor proteins, including α2. We also found that large changes in mating-type-specific gene expression between a and α or between mutant and wild type were not easily explained by the range of Tup1 occupancy levels within their promoters, as predicted by the classic model of a-specific gene repression by Tup1. Most surprisingly, we observed Tup1 occupancy upstream of the a-specific gene MFA2 and the α-specific gene MF(ALPHA)1 in cells in which each gene was expressed rather than repressed. These results, combined with the identification of additional mating-related genes upregulated in the tup1(S649F) α strain, illustrate that the role of Tup1 in distinguishing mating types in yeast appears to be both more comprehensive and more nuanced than previously appreciated.


Subject(s)
Gene Expression Regulation, Fungal , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Lipoproteins/genetics , Lipoproteins/metabolism , Nuclear Proteins/genetics , Pheromones/genetics , Pheromones/metabolism , Promoter Regions, Genetic , Repressor Proteins/genetics , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/genetics , Transcriptional Activation
15.
Mol Cell Biol ; 27(18): 6279-87, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17620414

ABSTRACT

CHD1 encodes an ATP-dependent chromatin remodeler with two chromodomains. Deletion of CHD1 suppresses the temperature-sensitive growth defect caused by mutations in either SPT16 or POB3, which encode subunits of the yFACT chromatin-reorganizing complex. chd1 also suppresses synthetic defects caused by combining an spt16 mutation with other transcription factor mutations, including the synthetic lethality caused by combining an spt16 mutation with TATA binding protein (TBP) or TFIIA defects. Binding of TBP and RNA polymerase II to the GAL1 promoter is reduced in a pob3 mutant, resulting in low levels of GAL1 expression, and all three defects are suppressed by removing Chd1. These results suggest that Chd1 and yFACT have opposing roles in regulating TBP binding at promoters. Additionally, overexpression of Chd1 is tolerated in wild-type cells but is toxic in spt16 mutants. Further, both the ATPase and chromodomain are required for Chd1 activity in opposing yFACT function. Similar to the suppression by chd1, mutations in the SET2 histone methyltransferase also suppress defects caused by yFACT mutations. chd1 and set2 are additive in suppressing pob3, suggesting that Chd1 and Set2 act in distinct pathways. Although human Chd1 has been shown to bind to H3-K4-Me, we discuss evidence arguing that yeast Chd1 binds to neither H3-K4-Me nor H3-K36-Me.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Fungal , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Deletion , Promoter Regions, Genetic , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , TATA-Box Binding Protein/metabolism , Transcription Factor TFIIA/metabolism , Transcription Factors/genetics , Transcriptional Elongation Factors
16.
Genetics ; 215(2): 407-420, 2020 06.
Article in English | MEDLINE | ID: mdl-32327563

ABSTRACT

Mediator is an essential, multisubunit complex that functions as a transcriptional coactivator in yeast and other eukaryotic organisms. Mediator has four conserved modules, Head, Middle, Tail, and Kinase, and has been implicated in nearly all aspects of gene regulation. The Tail module has been shown to recruit the Mediator complex to the enhancer or upstream activating sequence (UAS) regions of genes via interactions with transcription factors, and the Kinase module facilitates the transition of Mediator from the UAS/enhancer to the preinitiation complex via protein phosphorylation. Here, we analyze expression of the Saccharomyces cerevisiaeHO gene using a sin4 Mediator Tail mutation that separates the Tail module from the rest of the complex; the sin4 mutation permits independent recruitment of the Tail module to promoters without the rest of Mediator. Significant increases in recruitment of the SWI/SNF and SAGA coactivators to the HO promoter UAS were observed in a sin4 mutant, along with increased gene activation. These results are consistent with recent studies that have suggested that the Kinase module functions negatively to inhibit activation by the Tail. However, we found that Kinase module mutations did not mimic the effect of a sin4 mutation on HO expression. This suggests that at HO the core Mediator complex (Middle and Head modules) must play a role in limiting Tail binding to the promoter UAS and gene activation. We propose that the core Mediator complex helps modulate Mediator binding to the UAS regions of genes to limit coactivator recruitment and ensure proper regulation of gene transcription.


Subject(s)
Gene Expression Regulation, Fungal , Mediator Complex/metabolism , RNA Polymerase II/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription, Genetic , Transcriptional Activation , Mediator Complex/genetics , Promoter Regions, Genetic , RNA Polymerase II/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics
17.
Genetics ; 178(2): 649-59, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18245327

ABSTRACT

Chromatin-modifying factors regulate both transcription and DNA replication. The yFACT chromatin-reorganizing complex is involved in both processes, and the sensitivity of some yFACT mutants to the replication inhibitor hydroxyurea (HU) is one indication of a replication role. This HU sensitivity can be suppressed by disruptions of the SET2 or CHD1 genes, encoding a histone H3(K36) methyltransferase and a chromatin remodeling factor, respectively. The additive effect of set2 and chd1 mutations in suppressing the HU sensitivity of yFACT mutants suggests that these two factors function in separate pathways. The HU suppression is not an indirect effect of altered regulation of ribonucleotide reductase induced by HU. set2 and chd1 mutations also suppress the HU sensitivity of mutations in other genes involved in DNA replication, including CDC2, CTF4, ORC2, and MEC1. Additionally, a chd1 mutation can suppress the lethality normally caused by disruption of either MEC1 or RAD53 DNA damage checkpoint genes, as well as the lethality seen when a mec1 sml1 mutant is exposed to low levels of HU. The pob3 defect in S-phase progression is suppressed by set2 or chd1 mutations, suggesting that Set2 and Chd1 have specific roles in negatively regulating DNA replication.


Subject(s)
DNA Replication , DNA-Binding Proteins/genetics , Methyltransferases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Chromatin/genetics , DNA-Binding Proteins/metabolism , Gene Deletion , Hydroxyurea/pharmacology , Methyltransferases/metabolism , Mutagenesis , Mutation , Ribonucleoside Diphosphate Reductase/genetics , S Phase , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic
18.
Genetics ; 180(4): 1799-808, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18832360

ABSTRACT

We have created a resource to rapidly map genetic traits to specific chromosomes in yeast. This mapping is done using a set of 16 yeast strains each containing a different chromosome with a conditionally functional centromere. Conditional centromere function is achieved by integration of a GAL1 promoter in cis to centromere sequences. We show that the 16 yeast chromosomes can be individually lost in diploid strains, which become hemizygous for the destabilized chromosome. Interestingly, most 2n - 1 strains endoduplicate and become 2n. We also demonstrate how chromosome loss in this set of strains can be used to map both recessive and dominant markers to specific chromosomes. In addition, we show that this method can be used to rapidly validate gene assignments from screens of strain libraries such as the yeast gene disruption collection.


Subject(s)
Chromosomes, Fungal/genetics , Saccharomyces cerevisiae/genetics , Chromosome Mapping , Diploidy , Loss of Heterozygosity , Meiosis , Models, Genetic , Phenotype , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
19.
Mol Cell Biol ; 26(11): 4095-110, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16705163

ABSTRACT

We use chromatin immunoprecipitation assays to show that the Gcn5 histone acetyltransferase in SAGA is required for SWI/SNF association with the HO promoter and that binding of SWI/SNF and SAGA are interdependent. Previous results showed that SWI/SNF binding to HO was Gcn5 independent, but that work used a strain with a mutation in the Ash1 daughter-specific repressor of HO expression. Here, we show that Ash1 functions as a repressor that inhibits SWI/SNF binding and that Gcn5 is required to overcome Ash1 repression in mother cells to allow HO transcription. Thus, Gcn5 facilitates SWI/SNF binding by antagonizing Ash1. Similarly, a mutation in SIN3, like an ash1 mutation, allows both HO expression and SWI/SNF binding in the absence of Gcn5. Although Ash1 has recently been identified in a Sin3-Rpd3 complex, our genetic analysis shows that Ash1 and Sin3 have distinct functions in regulating HO. Analysis of mutant strains shows that SWI/SNF binding and HO expression are correlated and regulated by histone acetylation. The defect in HO expression caused by a mutant SWI/SNF with a Swi2(E834K) substitution can be partially suppressed by ash1 or spt3 mutation or by a gain-of-function V71E substitution in the TATA-binding protein (TBP). Spt3 inhibits TBP binding at HO, and genetic analysis suggests that Spt3 and TBP(V71E) act in the same pathway, distinct from that of Ash1. We have detected SWI/SNF binding at the HO TATA region, and our results suggest that SWI/SNF, either directly or indirectly, facilitates TBP binding at HO.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Deoxyribonucleases, Type II Site-Specific/genetics , Histones/metabolism , Promoter Regions, Genetic/genetics , TATA-Box Binding Protein/metabolism , Transcription Factors/metabolism , Acetylation , Adenosine Triphosphatases , Alleles , Catalysis , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Histone Acetyltransferases/antagonists & inhibitors , Histone Deacetylases , Models, Biological , Mutation/genetics , Protein Binding , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/metabolism , Suppression, Genetic , Transcription Factors/deficiency
20.
Genetics ; 212(4): 1181-1204, 2019 08.
Article in English | MEDLINE | ID: mdl-31167839

ABSTRACT

Activation of the Saccharomyces cerevisiae HO promoter is highly regulated, requiring the ordered recruitment of activators and coactivators and allowing production of only a few transcripts in mother cells within a short cell cycle window. We conducted genetic screens to identify the negative regulators of HO expression necessary to limit HO transcription. Known repressors of HO (Ash1 and Rpd3) were identified, as well as several additional chromatin-associated factors including the Hda1 histone deacetylase, the Isw2 chromatin remodeler, and the corepressor Tup1 We also identified clusters of HO promoter mutations that suggested roles for the Dot6/Tod6 (PAC site) and Ume6 repression pathways. We used ChIP assays with synchronized cells to validate the involvement of these factors and map the association of Ash1, Dot6, and Ume6 with the HO promoter to a brief window in the cell cycle between binding of the initial activating transcription factor and initiation of transcription. We found that Ash1 and Ume6 each recruit the Rpd3 histone deacetylase to HO, and their effects are additive. In contrast, Rpd3 was not recruited significantly to the PAC site, suggesting this site has a distinct mechanism for repression. Increases in HO expression and SWI/SNF recruitment were all additive upon loss of Ash1, Ume6, and PAC site factors, indicating the convergence of independent pathways for repression. Our results demonstrate that multiple protein complexes are important for limiting the spread of SWI/SNF-mediated nucleosome eviction across the HO promoter, suggesting that regulation requires a delicate balance of activities that promote and repress transcription.


Subject(s)
Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Deoxyribonucleases, Type II Site-Specific/genetics , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Deoxyribonucleases, Type II Site-Specific/metabolism , Gene Expression Regulation, Fungal , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL