Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 51(7): 3205-3222, 2023 04 24.
Article in English | MEDLINE | ID: mdl-36951111

ABSTRACT

Chromosomal instability (CIN) drives cell-to-cell heterogeneity, and the development of genetic diseases, including cancer. Impaired homologous recombination (HR) has been implicated as a major driver of CIN, however, the underlying mechanism remains unclear. Using a fission yeast model system, we establish a common role for HR genes in suppressing DNA double-strand break (DSB)-induced CIN. Further, we show that an unrepaired single-ended DSB arising from failed HR repair or telomere loss is a potent driver of widespread CIN. Inherited chromosomes carrying a single-ended DSB are subject to cycles of DNA replication and extensive end-processing across successive cell divisions. These cycles are enabled by Cullin 3-mediated Chk1 loss and checkpoint adaptation. Subsequent propagation of unstable chromosomes carrying a single-ended DSB continues until transgenerational end-resection leads to fold-back inversion of single-stranded centromeric repeats and to stable chromosomal rearrangements, typically isochromosomes, or to chromosomal loss. These findings reveal a mechanism by which HR genes suppress CIN and how DNA breaks that persist through mitotic divisions propagate cell-to-cell heterogeneity in the resultant progeny.


Subject(s)
Schizosaccharomyces , Humans , Chromosomal Instability , DNA Breaks, Double-Stranded , DNA Repair , Homologous Recombination , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism
2.
PLoS One ; 18(1): e0271016, 2023.
Article in English | MEDLINE | ID: mdl-36626373

ABSTRACT

We constructed a panel of S. pombe strains expressing DNA polymerase ε variants associated with cancer, specifically POLES297F, POLEV411L, POLEL424V, POLES459F, and used these to compare mutation rates determined by canavanine resistance with other selective methods. Canavanine-resistance mutation rates are broadly similar to those seen with reversion of the ade-485 mutation to adenine prototrophy, but lower than 5-fluoroorotic acid (FOA)-resistance rates (inactivation of ura4+ or ura5+ genes). Inactivation of several genes has been associated with canavanine resistance in S. pombe but surprisingly whole genome sequencing showed that 8/8 spontaneous canavanine-resistant mutants have an R175C mutation in the any1/arn1 gene. This gene encodes an α-arrestin-like protein involved in mediating Pub1 ubiquitylation of target proteins, and the phenotypic resistance to canavanine by this single mutation is similar to that shown by the original "can1-1" strain, which also has the any1R175C mutation. Some of the spontaneous mutants have additional mutations in arginine transporters, suggesting that this may marginally increase resistance to canavanine. The any1R175C strain showed internalisation of the Cat1 arginine transporter as previously reported, explaining the canavanine-resistance phenotype.


Subject(s)
Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Canavanine/pharmacology , Canavanine/metabolism , Mutation Rate , Schizosaccharomyces pombe Proteins/metabolism , Mutation , Arginine/metabolism , Arrestins/metabolism
3.
PLoS Genet ; 17(7): e1009526, 2021 07.
Article in English | MEDLINE | ID: mdl-34228709

ABSTRACT

Somatic and germline mutations in the proofreading domain of the replicative DNA polymerase ε (POLE-exonuclease domain mutations, POLE-EDMs) are frequently found in colorectal and endometrial cancers and, occasionally, in other tumours. POLE-associated cancers typically display hypermutation, and a unique mutational signature, with a predominance of C > A transversions in the context TCT and C > T transitions in the context TCG. To understand better the contribution of hypermutagenesis to tumour development, we have modelled the most recurrent POLE-EDM (POLE-P286R) in Schizosaccharomyces pombe. Whole-genome sequencing analysis revealed that the corresponding pol2-P287R allele also has a strong mutator effect in vivo, with a high frequency of base substitutions and relatively few indel mutations. The mutations are equally distributed across different genomic regions, but in the immediate vicinity there is an asymmetry in AT frequency. The most abundant base-pair changes are TCT > TAT transversions and, in contrast to human mutations, TCG > TTG transitions are not elevated, likely due to the absence of cytosine methylation in fission yeast. The pol2-P287R variant has an increased sensitivity to elevated dNTP levels and DNA damaging agents, and shows reduced viability on depletion of the Pfh1 helicase. In addition, S phase is aberrant and RPA foci are elevated, suggestive of ssDNA or DNA damage, and the pol2-P287R mutation is synthetically lethal with rad3 inactivation, indicative of checkpoint activation. Significantly, deletion of genes encoding some translesion synthesis polymerases, most notably Pol κ, partially suppresses pol2-P287R hypermutation, indicating that polymerase switching contributes to this phenotype.


Subject(s)
DNA Polymerase II/genetics , DNA Replication , Mutation , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/genetics , Checkpoint Kinase 2/genetics , DNA Helicases/genetics , DNA Polymerase II/metabolism , Genome, Fungal , Humans , Neoplasms/genetics , Poly-ADP-Ribose Binding Proteins/genetics , S Phase/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism
4.
Nucleic Acids Res ; 48(3): 1271-1284, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31828313

ABSTRACT

The healing of broken chromosomes by de novo telomere addition, while a normal developmental process in some organisms, has the potential to cause extensive loss of heterozygosity, genetic disease, or cell death. However, it is unclear how de novo telomere addition (dnTA) is regulated at DNA double-strand breaks (DSBs). Here, using a non-essential minichromosome in fission yeast, we identify roles for the HR factors Rqh1 helicase, in concert with Rad55, in suppressing dnTA at or near a DSB. We find the frequency of dnTA in rqh1Δ rad55Δ cells is reduced following loss of Exo1, Swi5 or Rad51. Strikingly, in the absence of the distal homologous chromosome arm dnTA is further increased, with nearly half of the breaks being healed in rqh1Δ rad55Δ or rqh1Δ exo1Δ cells. These findings provide new insights into the genetic context of highly efficient dnTA within HR intermediates, and how such events are normally suppressed to maintain genome stability.


Subject(s)
DNA Helicases/genetics , DNA-Binding Proteins/genetics , Recombinational DNA Repair/genetics , Schizosaccharomyces pombe Proteins/genetics , Telomere/genetics , Chromosomes, Fungal/genetics , DNA Breaks, Double-Stranded , Exodeoxyribonucleases/genetics , Gene Expression Regulation, Fungal/genetics , Genome, Fungal/genetics , Genomic Instability/genetics , Loss of Heterozygosity/genetics , Rad51 Recombinase/genetics , Schizosaccharomyces/genetics
5.
J Cell Sci ; 132(6)2019 03 25.
Article in English | MEDLINE | ID: mdl-30674555

ABSTRACT

Replication stress is a common feature of cancer cells, and thus a potentially important therapeutic target. Here, we show that cyclin-dependent kinase (CDK)-induced replication stress, resulting from Wee1 inactivation, is synthetic lethal with mutations disrupting dNTP homeostasis in fission yeast. Wee1 inactivation leads to increased dNTP demand and replication stress through CDK-induced firing of dormant replication origins. Subsequent dNTP depletion leads to inefficient DNA replication, DNA damage and to genome instability. Cells respond to this replication stress by increasing dNTP supply through histone methyltransferase Set2-dependent MBF-induced expression of Cdc22, the catalytic subunit of ribonucleotide reductase (RNR). Disrupting dNTP synthesis following Wee1 inactivation, through abrogating Set2-dependent H3K36 tri-methylation or DNA integrity checkpoint inactivation results in critically low dNTP levels, replication collapse and cell death, which can be rescued by increasing dNTP levels. These findings support a 'dNTP supply and demand' model in which maintaining dNTP homeostasis is essential to prevent replication catastrophe in response to CDK-induced replication stress.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinases/metabolism , Nucleotides/metabolism , Protein-Tyrosine Kinases/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Cell Cycle Checkpoints , DNA Damage , DNA Replication , Histone Code , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Homeostasis , Methylation , Schizosaccharomyces/metabolism , Synthetic Lethal Mutations , Transcription Factors/metabolism
6.
Cold Spring Harb Protoc ; 2018(4)2018 04 02.
Article in English | MEDLINE | ID: mdl-28733409

ABSTRACT

Pulsed field gel electrophoresis (PFGE) uses alternatively oriented pulsed electrical fields to separate large DNA molecules. Here, we describe PFGE protocols and conditions for separating and visualizing chromosomes between 0.5 and 6 Mb (optimal for analyzing the endogenous fission yeast chromosomes of 5.7, 4.6, and 3.5 Mb), and for shorter chromosomal elements of between 50 and 600 kb, such as the 530 kb Ch16 minichromosome. In addition to determining chromosome size, this technique has a wide range of applications, including determining whether DNA replication or repair is complete, defining the molecular karyotype of cells, analyzing chromosomal rearrangements, assigning genes or constructs to particular chromosomes, and isolating DNA from specific chromosomes.


Subject(s)
Chromosomes, Fungal/genetics , Electrophoresis, Gel, Pulsed-Field/methods , Schizosaccharomyces/genetics , Cells, Cultured , DNA, Fungal/genetics , Electrophoresis, Agar Gel , Imaging, Three-Dimensional , Staining and Labeling
7.
Cold Spring Harb Protoc ; 2018(4)2018 04 02.
Article in English | MEDLINE | ID: mdl-28733408

ABSTRACT

DNA double-strand breaks (DSBs), arising during normal DNA metabolism or following exposure to mutagenic agents such as ionizing radiation can lead to chromosomal rearrangements and genome instability, and are potentially lethal if unrepaired. Therefore, understanding the mechanisms of DSB repair and misrepair, and identifying the factors involved in these processes is of biological as well as medical interest. Here we describe a DSB assay in Schizosaccharomyces pombe that can be used to identify and quantify different repair, misrepair, and failed repair events resulting from a site-specific DSB within the context of a nonessential minichromosome, Ch16 This assay can be used to determine the contribution of most genes or genetic backgrounds to DSB repair and genome stability, and can also provide mechanistic insights into their function.


Subject(s)
Biological Assay/methods , DNA Breaks, Double-Stranded , DNA Repair , Cell Survival , Chromosomes, Fungal/genetics , Crosses, Genetic , Mutation/genetics , Plasmids/metabolism , Schizosaccharomyces/genetics , Transformation, Genetic
8.
Cell Rep ; 20(11): 2693-2705, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28903048

ABSTRACT

Chromatin modification through histone H3 lysine 36 methylation by the SETD2 tumor suppressor plays a key role in maintaining genome stability. Here, we describe a role for Set2-dependent H3K36 methylation in facilitating DNA replication and the transcriptional responses to both replication stress and DNA damage through promoting MluI cell-cycle box (MCB) binding factor (MBF)-complex-dependent transcription in fission yeast. Set2 loss leads to reduced MBF-dependent ribonucleotide reductase (RNR) expression, reduced deoxyribonucleoside triphosphate (dNTP) synthesis, altered replication origin firing, and a checkpoint-dependent S-phase delay. Accordingly, prolonged S phase in the absence of Set2 is suppressed by increasing dNTP synthesis. Furthermore, H3K36 is di- and tri-methylated at these MBF gene promoters, and Set2 loss leads to reduced MBF binding and transcription in response to genotoxic stress. Together, these findings provide new insights into how H3K36 methylation facilitates DNA replication and promotes genotoxic stress responses in fission yeast.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , DNA Replication , Histone-Lysine N-Methyltransferase/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces/genetics , Transcription Factors/metabolism , Transcription, Genetic , Cell Cycle Checkpoints/genetics , DNA Damage/genetics , DNA Replication/genetics , DNA, Fungal/metabolism , Down-Regulation/genetics , Gene Expression Regulation, Fungal , Genes, Fungal , Mutation/genetics , Nucleotides/metabolism , Replication Origin/genetics , S Phase/genetics
9.
Genes (Basel) ; 8(2)2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28146119

ABSTRACT

A crucial factor in maintaining genome stability is establishing deoxynucleoside triphosphate (dNTP) levels within a range that is optimal for chromosomal replication. Since DNA replication is relevant to a wide range of other chromosomal activities, these may all be directly or indirectly affected when dNTP concentrations deviate from a physiologically normal range. The importance of understanding these consequences is relevant to genetic disorders that disturb dNTP levels, and strategies that inhibit dNTP synthesis in cancer chemotherapy and for treatment of other disorders. We review here how abnormal dNTP levels affect DNA replication and discuss the consequences for genome stability.

10.
Nucleic Acids Res ; 44(4): 1703-17, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26682798

ABSTRACT

The formation of RNA-DNA hybrids, referred to as R-loops, can promote genome instability and cancer development. Yet the mechanisms by which R-loops compromise genome instability are poorly understood. Here, we establish roles for the evolutionarily conserved Nrl1 protein in pre-mRNA splicing regulation, R-loop suppression and in maintaining genome stability. nrl1Δ mutants exhibit endogenous DNA damage, are sensitive to exogenous DNA damage, and have defects in homologous recombination (HR) repair. Concomitantly, nrl1Δ cells display significant changes in gene expression, similar to those induced by DNA damage in wild-type cells. Further, we find that nrl1Δ cells accumulate high levels of R-loops, which co-localize with HR repair factors and require Rad51 and Rad52 for their formation. Together, our findings support a model in which R-loop accumulation and subsequent DNA damage sequesters HR factors, thereby compromising HR repair at endogenously or exogenously induced DNA damage sites, leading to genome instability.


Subject(s)
Alternative Splicing/genetics , Genomic Instability/genetics , Homologous Recombination/genetics , RNA Precursors/genetics , Schizosaccharomyces pombe Proteins/genetics , DNA/chemistry , DNA/genetics , DNA Repair/genetics , RNA/chemistry , RNA/genetics , Rad51 Recombinase/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Schizosaccharomyces/genetics , Spliceosomes/genetics , Spliceosomes/metabolism
11.
Cancer Cell ; 28(5): 557-568, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26602815

ABSTRACT

Histone H3K36 trimethylation (H3K36me3) is frequently lost in multiple cancer types, identifying it as an important therapeutic target. Here we identify a synthetic lethal interaction in which H3K36me3-deficient cancers are acutely sensitive to WEE1 inhibition. We show that RRM2, a ribonucleotide reductase subunit, is the target of this synthetic lethal interaction. RRM2 is regulated by two pathways here: first, H3K36me3 facilitates RRM2 expression through transcription initiation factor recruitment; second, WEE1 inhibition degrades RRM2 through untimely CDK activation. Therefore, WEE1 inhibition in H3K36me3-deficient cells results in RRM2 reduction, critical dNTP depletion, S-phase arrest, and apoptosis. Accordingly, this synthetic lethality is suppressed by increasing RRM2 expression or inhibiting RRM2 degradation. Finally, we demonstrate that WEE1 inhibitor AZD1775 regresses H3K36me3-deficient tumor xenografts.


Subject(s)
Cell Cycle Proteins/metabolism , Histones/metabolism , Neoplasms/metabolism , Nuclear Proteins/metabolism , Nucleotides/metabolism , Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Regulation, Neoplastic/drug effects , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Humans , Lysine/genetics , Lysine/metabolism , Methylation/drug effects , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Neoplasms/genetics , Neoplasms/prevention & control , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nucleotides/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Xenograft Model Antitumor Assays
12.
Nat Commun ; 5: 4091, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24909977

ABSTRACT

DNA double-strand break (DSB) repair is a highly regulated process performed predominantly by non-homologous end joining (NHEJ) or homologous recombination (HR) pathways. How these pathways are coordinated in the context of chromatin is unclear. Here we uncover a role for histone H3K36 modification in regulating DSB repair pathway choice in fission yeast. We find Set2-dependent H3K36 methylation reduces chromatin accessibility, reduces resection and promotes NHEJ, while antagonistic Gcn5-dependent H3K36 acetylation increases chromatin accessibility, increases resection and promotes HR. Accordingly, loss of Set2 increases H3K36Ac, chromatin accessibility and resection, while Gcn5 loss results in the opposite phenotypes following DSB induction. Further, H3K36 modification is cell cycle regulated with Set2-dependent H3K36 methylation peaking in G1 when NHEJ occurs, while Gcn5-dependent H3K36 acetylation peaks in S/G2 when HR prevails. These findings support an H3K36 chromatin switch in regulating DSB repair pathway choice.


Subject(s)
Acetyltransferases/metabolism , Chromatin/metabolism , DNA End-Joining Repair , DNA Repair , DNA, Fungal/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Recombinational DNA Repair , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/genetics , Acetylation , Methylation , Schizosaccharomyces/metabolism
13.
Nucleic Acids Res ; 42(9): 5644-56, 2014 May.
Article in English | MEDLINE | ID: mdl-24623809

ABSTRACT

DNA double-strand breaks (DSBs) can cause chromosomal rearrangements and extensive loss of heterozygosity (LOH), hallmarks of cancer cells. Yet, how such events are normally suppressed is unclear. Here we identify roles for the DNA damage checkpoint pathway in facilitating homologous recombination (HR) repair and suppressing extensive LOH and chromosomal rearrangements in response to a DSB. Accordingly, deletion of Rad3(ATR), Rad26ATRIP, Crb2(53BP1) or Cdc25 overexpression leads to reduced HR and increased break-induced chromosome loss and rearrangements. We find the DNA damage checkpoint pathway facilitates HR, in part, by promoting break-induced Cdt2-dependent nucleotide synthesis. We also identify additional roles for Rad17, the 9-1-1 complex and Chk1 activation in facilitating break-induced extensive resection and chromosome loss, thereby suppressing extensive LOH. Loss of Rad17 or the 9-1-1 complex results in a striking increase in break-induced isochromosome formation and very low levels of chromosome loss, suggesting the 9-1-1 complex acts as a nuclease processivity factor to facilitate extensive resection. Further, our data suggest redundant roles for Rad3ATR and Exo1 in facilitating extensive resection. We propose that the DNA damage checkpoint pathway coordinates resection and nucleotide synthesis, thereby promoting efficient HR repair and genome stability.


Subject(s)
DNA Breaks, Double-Stranded , DNA Cleavage , Genomic Instability , Recombinational DNA Repair , Schizosaccharomyces/genetics , Cell Cycle Checkpoints , Checkpoint Kinase 2/metabolism , Chromosomes, Fungal/genetics , Comparative Genomic Hybridization , Exodeoxyribonucleases/metabolism , Genome, Fungal , Loss of Heterozygosity , Nucleotides/biosynthesis , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism
14.
Methods ; 57(2): 227-33, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22504526

ABSTRACT

The fission yeast Schizosaccharomyces pombe is a useful model for analysing DNA replication as genetic methods to allow conditional inactivation of relevant proteins can provide important information about S-phase execution. A number of strategies are available to allow regulation of protein level or activity but there are disadvantages specific to each method and this may have limitations for particular proteins or experiments. We have investigated the utility of the inducible hormone-binding domain (HBD) system, which has been described in other organisms but little used in fission yeast, for the creation of conditional-lethal replication mutants. In this method, proteins are tagged with HBD and can be regulated with ß-estradiol. In this article, we describe the application of this method in fission yeast, specifically with regard to analysis of the function of GINS, an essential component of the eukaryotic replicative helicase, the CMG complex.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Down-Regulation , Recombinant Fusion Proteins/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/genetics , Cell Cycle Checkpoints/genetics , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone/biosynthesis , DNA Replication , Estradiol/pharmacology , Estrogens/pharmacology , Gene Expression/drug effects , Phenotype , Promoter Regions, Genetic , Protein Structure, Tertiary , Protein Transport , Recombinant Fusion Proteins/biosynthesis , S Phase/genetics , Schizosaccharomyces/growth & development , Schizosaccharomyces pombe Proteins/biosynthesis
15.
Mol Biol Cell ; 20(4): 1213-22, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19109429

ABSTRACT

The tetrameric GINS complex, consisting of Sld5-Psf1-Psf2-Psf3, plays an essential role in the initiation and elongation steps of eukaryotic DNA replication, although its biochemical function is unclear. Here we investigate the function of GINS in fission yeast, using fusion of Psf1 and Psf2 subunits to a steroid hormone-binding domain (HBD) to make GINS function conditional on the presence of beta-estradiol. We show that inactivation of Psf1-HBD causes a tight but rapidly reversible DNA replication arrest phenotype. Inactivation of Psf2-HBD similarly blocks premeiotic DNA replication and leads to loss of nuclear localization of another GINS subunit, Psf3. Inactivation of GINS has distinct effects on the replication origin association and chromatin binding of two of the replicative DNA polymerases. Inactivation of Psf1 leads to loss of chromatin binding of DNA polymerase epsilon, and Cdc45 is similarly affected. In contrast, chromatin association of the catalytic subunit of DNA polymerase alpha is not affected by defective GINS function. We suggest that GINS functions in a pathway that involves Cdc45 and is necessary for DNA polymerase epsilon chromatin binding, but that a separate pathway sets up the chromatin association of DNA polymerase alpha.


Subject(s)
Chromatin/metabolism , DNA Polymerase II/metabolism , DNA Polymerase I/metabolism , DNA Replication , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces/genetics , Catalytic Domain , DNA-Binding Proteins/metabolism , Estradiol/metabolism , Nuclear Proteins/metabolism , Phenotype , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Replication Origin , S Phase , Schizosaccharomyces/cytology
16.
BMC Cell Biol ; 9: 23, 2008 May 05.
Article in English | MEDLINE | ID: mdl-18457584

ABSTRACT

BACKGROUND: The fission yeast Schizosaccharomyces pombe is widely-used as a model organism for the study of a broad range of eukaryotic cellular processes such as cell cycle, genome stability and cell morphology. Despite the availability of extensive set of genetic, molecular biological, biochemical and cell biological tools for analysis of protein function in fission yeast, studies are often hampered by the lack of an effective method allowing for the rapid regulation of protein level or protein activity. RESULTS: In order to be able to regulate protein function, we have made use of a previous finding that the hormone binding domain of steroid receptors can be used as a regulatory cassette to subject the activity of heterologous proteins to hormonal regulation. The approach is based on fusing the protein of interest to the hormone binding domain (HBD) of the estrogen receptor (ER). The HBD tag will attract the Hsp90 complex, which can render the fusion protein inactive. Upon addition of estradiol the protein is quickly released from the Hsp90 complex and thereby activated. We have tagged and characterised the induction of activity of four different HBD-tagged proteins. Here we show that the tag provided the means to effectively regulate the activity of two of these proteins. CONCLUSION: The estradiol-regulatable hormone binding domain provides a means to regulate the function of some, though not all, fission yeast proteins. This system may result in very quick and reversible activation of the protein of interest. Therefore it will be a powerful tool and it will open experimental approaches in fission yeast that have previously not been possible. Since fission yeast is a widely-used model organism, this will be valuable in many areas of research.


Subject(s)
Gene Expression Regulation, Fungal , Receptors, Estrogen/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/metabolism , Animals , Binding Sites , Cell Cycle/drug effects , Cell Cycle/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cloning, Molecular , Cyclin B/genetics , Cyclin B/metabolism , DNA Replication/drug effects , DNA Replication/genetics , Estradiol/metabolism , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/pharmacology , Receptors, Estrogen/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Schizosaccharomyces pombe Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL