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










Publication year range
1.
Mol Cell Biol ; 38(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29263158

ABSTRACT

A tightly controlled cellular deoxyribonucleotide (deoxynucleoside triphosphate [dNTP]) pool is critical for maintenance of genome integrity. One mode of dNTP pool regulation is through subcellular localization of ribonucleotide reductase (RNR), the enzyme that catalyzes the rate-limiting step of dNTP biosynthesis. In Saccharomyces cerevisiae, the RNR small subunit, Rnr2-Rnr4, is localized to the nucleus, whereas the large subunit, Rnr1, is cytoplasmic. As cells enter S phase or encounter DNA damage, Rnr2-Rnr4 relocalizes to the cytoplasm to form an active holoenzyme complex with Rnr1. Although the DNA damage-induced relocalization requires the checkpoint kinases Mec1-Rad53-Dun1, the S-phase-specific redistribution does not. Here, we report that the S-phase cyclin-cyclin-dependent kinase (CDK) complex Clb6-Cdc28 controls Rnr2-Rnr4 relocalization in S phase. Rnr2 contains a consensus CDK site and exhibits Clb6-dependent phosphorylation in S phase. Deletion of CLB6 or removal of the CDK site results in an increased association of Rnr2 with its nuclear anchor Wtm1, nuclear retention of Rnr2-Rnr4, and an enhanced sensitivity to the RNR inhibitor hydroxyurea. Thus, we propose that Rnr2-Rnr4 redistribution in S phase is triggered by Clb6-Cdc28-mediated phosphorylation of Rnr2, which disrupts the Rnr2-Wtm1 interaction and promotes the release of Rnr2-Rnr4 from the nucleus.


Subject(s)
CDC28 Protein Kinase, S cerevisiae/metabolism , Cyclin B/metabolism , Ribonucleoside Diphosphate Reductase/metabolism , Ribonucleotide Reductases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , CDC28 Protein Kinase, S cerevisiae/analysis , Cyclin B/analysis , Phosphorylation , Protein Transport , Ribonucleoside Diphosphate Reductase/analysis , Ribonucleotide Reductases/analysis , S Phase , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/analysis
2.
J Biol Chem ; 292(27): 11445-11451, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28515324

ABSTRACT

How each metalloprotein assembles the correct metal at the proper binding site presents challenges to the cell. The di-iron enzyme ribonucleotide reductase (RNR) uses a diferric-tyrosyl radical (FeIII2-Y•) cofactor to initiate nucleotide reduction. Assembly of this cofactor requires O2, FeII, and a reducing equivalent. Recent studies show that RNR cofactor biosynthesis shares the same source of iron, in the form of [2Fe-2S]-GSH2 from the monothiol glutaredoxin Grx3/4, and the same electron source, in the form of the Dre2-Tah18 electron transfer chain, with the cytosolic iron-sulfur protein assembly (CIA) machinery required for maturation of [4Fe-4S] clusters in cytosolic and nuclear proteins. Here, we further investigated the interplay between the formation of the FeIII2-Y• cofactor in RNR and the cellular iron-sulfur (Fe-S) protein biogenesis pathways by examining both the iron loading into the RNR ß subunit and the RNR catalytic activity in yeast mutants depleted of individual components of the mitochondrial iron-sulfur cluster assembly (ISC) and the CIA machineries. We found that both iron loading and cofactor assembly in RNR are dependent on the ISC machinery. We also found that Dre2 is required for RNR cofactor formation but appears to be dispensable for iron loading. None of the CIA components downstream of Dre2 was required for RNR cofactor formation. Thus, the pathways for RNR and Fe-S cluster biogenesis bifurcate after the Dre2-Tah18 step. We conclude that RNR cofactor biogenesis requires the ISC machinery to mature the Grx3/4 and Dre2 Fe-S proteins, which then function in iron and electron delivery to RNR, respectively.


Subject(s)
Free Radicals/metabolism , Glutaredoxins/metabolism , Iron-Sulfur Proteins/metabolism , Oxidoreductases/metabolism , Ribonucleotide Reductases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Glutaredoxins/genetics , Iron-Sulfur Proteins/genetics , Oxidoreductases/genetics , Ribonucleotide Reductases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
3.
J Biol Chem ; 291(18): 9807-17, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-26970775

ABSTRACT

Ribonucleotide reductase (RNR) is an essential iron-dependent enzyme that catalyzes deoxyribonucleotide synthesis in eukaryotes. Living organisms have developed multiple strategies to tightly modulate RNR function to avoid inadequate or unbalanced deoxyribonucleotide pools that cause DNA damage and genome instability. Yeast cells activate RNR in response to genotoxic stress and iron deficiency by facilitating redistribution of its small heterodimeric subunit Rnr2-Rnr4 from the nucleus to the cytoplasm, where it forms an active holoenzyme with large Rnr1 subunit. Dif1 protein inhibits RNR by promoting nuclear import of Rnr2-Rnr4. Upon DNA damage, Dif1 phosphorylation by the Dun1 checkpoint kinase and its subsequent degradation enhances RNR function. In this report, we demonstrate that Dun1 kinase triggers Rnr2-Rnr4 redistribution to the cytoplasm in response to iron deficiency. We show that Rnr2-Rnr4 relocalization by low iron requires Dun1 kinase activity and phosphorylation site Thr-380 in the Dun1 activation loop, but not the Dun1 forkhead-associated domain. By using different Dif1 mutant proteins, we uncover that Dun1 phosphorylates Dif1 Ser-104 and Thr-105 residues upon iron scarcity. We observe that the Dif1 phosphorylation pattern differs depending on the stimuli, which suggests different Dun1 activating pathways. Importantly, the Dif1-S104A/T105A mutant exhibits defects in nucleus-to-cytoplasm redistribution of Rnr2-Rnr4 by iron limitation. Taken together, these results reveal that, in response to iron starvation, Dun1 kinase phosphorylates Dif1 to stimulate Rnr2-Rnr4 relocalization to the cytoplasm and promote RNR function.


Subject(s)
Cell Cycle Proteins/metabolism , Iron/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribonucleoside Diphosphate Reductase/metabolism , Ribonucleotide Reductases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cell Cycle Proteins/genetics , DNA Damage , Protein Serine-Threonine Kinases/genetics , Protein Transport/physiology , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleotide Reductases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
4.
Microbiologyopen ; 4(6): 941-51, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26450372

ABSTRACT

We have identified a new downstream target gene of the Aft1/2-regulated iron regulon in budding yeast Saccharomyces cerevisiae, the late-annotated small open reading frame LSO1. LSO1 transcript is among the most highly induced from a transcriptome analysis of a fet3-1 mutant grown in the presence of the iron chelator bathophenanthrolinedisulfonic acid. LSO1 has a paralog, LSO2, which is constitutively expressed and not affected by iron availability. In contrast, we find that the LSO1 promoter region contains three consensus binding sites for the Aft1/2 transcription factors and that an LSO1-lacZ reporter is highly induced under low-iron conditions in a Aft1-dependent manner. The expression patterns of the Lso1 and Lso2 proteins mirror those of their mRNAs. Both proteins are localized to the nucleus and cytoplasm, but become more cytoplasmic upon iron deprivation consistent with a role in iron transport. LSO1 and LSO2 appear to play overlapping roles in the cellular response to iron starvation since single lso1 and lso2 mutants are sensitive to iron deprivation and this sensitivity is exacerbated when both genes are deleted.


Subject(s)
Iron/metabolism , Regulon , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Binding Sites , Gene Expression Regulation, Fungal , Promoter Regions, Genetic , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription, Genetic
5.
Mol Cell Biol ; 34(17): 3259-71, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24958100

ABSTRACT

Iron is an essential micronutrient for all eukaryotic organisms because it participates as a redox-active cofactor in many biological processes, including DNA replication and repair. Eukaryotic ribonucleotide reductases (RNRs) are Fe-dependent enzymes that catalyze deoxyribonucleoside diphosphate (dNDP) synthesis. We show here that the levels of the Sml1 protein, a yeast RNR large-subunit inhibitor, specifically decrease in response to both nutritional and genetic Fe deficiencies in a Dun1-dependent but Mec1/Rad53- and Aft1-independent manner. The decline of Sml1 protein levels upon Fe starvation depends on Dun1 forkhead-associated and kinase domains, the 26S proteasome, and the vacuolar proteolytic pathway. Depletion of core components of the mitochondrial iron-sulfur cluster assembly leads to a Dun1-dependent diminution of Sml1 protein levels. The physiological relevance of Sml1 downregulation by Dun1 under low-Fe conditions is highlighted by the synthetic growth defect observed between dun1Δ and fet3Δ fet4Δ mutants, which is rescued by SML1 deletion. Consistent with an increase in RNR function, Rnr1 protein levels are upregulated upon Fe deficiency. Finally, dun1Δ mutants display defects in deoxyribonucleoside triphosphate (dNTP) biosynthesis under low-Fe conditions. Taken together, these results reveal that the Dun1 checkpoint kinase promotes RNR function in response to Fe starvation by stimulating Sml1 protein degradation.


Subject(s)
Cell Cycle Proteins/metabolism , Iron/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribonucleotide Reductases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Binding Sites/genetics , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deoxyribonucleotides/biosynthesis , Gene Deletion , Genes, Fungal , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Proteolysis , Ribonucleotide Reductases/antagonists & inhibitors , Ribonucleotide Reductases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tristetraprolin/genetics , Tristetraprolin/metabolism
6.
Proc Natl Acad Sci U S A ; 111(17): E1695-704, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24733891

ABSTRACT

Eukaryotic ribonucleotide reductases (RNRs) require a diferric-tyrosyl radical (Fe(III)2-Y•) cofactor to produce deoxynucleotides essential for DNA replication and repair. This metallocofactor is an important target of RNR-based therapeutics, although mechanisms of in vivo cofactor assembly, inactivation, and reactivation are poorly understood. Here, we demonstrate that the conserved Fe-S protein-diflavin reductase complex, Dre2-Tah18, plays a critical role in RNR cofactor biosynthesis. Depletion of Dre2 affects both RNR gene transcription and mRNA turnover through the activation of the DNA-damage checkpoint and the Aft1/Aft2-controlled iron regulon. Under conditions of comparable RNR protein levels, cells with diminishing Dre2 have significantly reduced ability to make deoxynucleotides. Furthermore, the kinetics and levels of in vivo reconstitution of the RNR cofactor are severely impaired in two conditional tah18 mutants. Together, these findings provide insight into RNR cofactor formation and reveal a shared mechanism underlying assembly of the Fe(III)2-Y• cofactor in RNR and the Fe-S clusters in cytosolic and nuclear proteins.


Subject(s)
DNA, Fungal/biosynthesis , Electrons , Iron-Sulfur Proteins/metabolism , Iron/metabolism , Oxidoreductases/metabolism , Ribonucleoside Diphosphate Reductase/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Coenzymes/metabolism , DNA Damage , Mutation/genetics , Repressor Proteins/metabolism , S Phase , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/growth & development , Temperature
7.
J Biol Chem ; 288(20): 13951-13959, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23532842

ABSTRACT

The small subunit (ß2) of class Ia ribonucleotide reductase (RNR) houses a diferric tyrosyl cofactor (Fe2(III)-Y(•)) that initiates nucleotide reduction in the large subunit (α2) via a long range radical transfer (RT) pathway in the holo-(α2)m(ß2)n complex. The C-terminal tails of ß2 are predominantly responsible for interaction with α2, with a conserved tyrosine residue in the tail (Tyr(356) in Escherichia coli NrdB) proposed to participate in cofactor assembly/maintenance and in RT. In the absence of structure of any holo-RNR, the role of the ß tail in cluster assembly/maintenance and its predisposition within the holo-complex have remained unknown. In this study, we have taken advantage of the unusual heterodimeric nature of the Saccharomyces cerevisiae RNR small subunit (ßß'), of which only ß contains a cofactor, to address both of these issues. We demonstrate that neither ß-Tyr(376) nor ß'-Tyr(323) (Tyr(356) equivalent in NrdB) is required for cofactor assembly in vivo, in contrast to the previously proposed mechanism for E. coli cofactor maintenance and assembly in vitro. Furthermore, studies with reconstituted-ßß' and an in vivo viability assay show that ß-Tyr(376) is essential for RT, whereas Tyr(323) in ß' is not. Although the C-terminal tail of ß' is dispensable for cofactor formation and RT, it is essential for interactions with ß and α to form the active holo-RNR. Together the results provide the first evidence of a directed orientation of the ß and ß' C-terminal tails relative to α within the holoenzyme consistent with a docking model of the two subunits and argue against RT across the ß ß' interface.


Subject(s)
Ribonucleotide Reductases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Amino Acid Sequence , Electron Spin Resonance Spectroscopy , Escherichia coli/metabolism , Free Radicals , Iron/chemistry , Models, Chemical , Molecular Sequence Data , Mutation , Plasmids/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Tyrosine/chemistry
8.
J Mol Biol ; 419(5): 315-29, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22465672

ABSTRACT

Ribonucleotide reductases (RRs) catalyze the rate-limiting step of de novo deoxynucleotide (dNTP) synthesis. Eukaryotic RRs consist of two proteins, RR1 (α) that contains the catalytic site and RR2 (ß) that houses a diferric-tyrosyl radical essential for ribonucleoside diphosphate reduction. Biochemical analysis has been combined with isothermal titration calorimetry (ITC), X-ray crystallography and yeast genetics to elucidate the roles of two loop 2 mutations R293A and Q288A in Saccharomyces cerevisiae RR1 (ScRR1). These mutations, R293A and Q288A, cause lethality and severe S phase defects, respectively, in cells that use ScRR1 as the sole source of RR1 activity. Compared to the wild-type enzyme activity, R293A and Q288A mutants show 4% and 15%, respectively, for ADP reduction, whereas they are 20% and 23%, respectively, for CDP reduction. ITC data showed that R293A ScRR1 is unable to bind ADP and binds CDP with 2-fold lower affinity compared to wild-type ScRR1. With the Q288A ScRR1 mutant, there is a 6-fold loss of affinity for ADP binding and a 2-fold loss of affinity for CDP compared to the wild type. X-ray structures of R293A ScRR1 complexed with dGTP and AMPPNP-CDP [AMPPNP, adenosine 5-(ß,γ-imido)triphosphate tetralithium salt] reveal that ADP is not bound at the catalytic site, and CDP binds farther from the catalytic site compared to wild type. Our in vivo functional analyses demonstrated that R293A cannot support mitotic growth, whereas Q288A can, albeit with a severe S phase defect. Taken together, our structure, activity, ITC and in vivo data reveal that the arginine 293 and glutamine 288 residues of ScRR1 are crucial in facilitating ADP and CDP substrate selection.


Subject(s)
Arginine/chemistry , Glutamine/chemistry , Ribonucleotide Reductases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Adenosine Diphosphate/chemistry , Allosteric Site , Arginine/genetics , Catalytic Domain , Crystallography, X-Ray , Cytidine Diphosphate/chemistry , Glutamine/genetics , Models, Molecular , Mutation , Protein Binding , Ribonucleotide Reductases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Substrate Specificity
9.
J Biol Chem ; 286(48): 41499-41509, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-21931161

ABSTRACT

The ß(2) subunit of class Ia ribonucleotide reductase (RNR) contains a diferric tyrosyl radical cofactor (Fe(2)(III)-Tyr(•)) that is essential for nucleotide reduction. The ß(2) subunit of Saccharomyces cerevisiae is a heterodimer of Rnr2 (ß) and Rnr4 (ß'). Although only ß is capable of iron binding and Tyr(•) formation, cells lacking ß' are either dead or exhibit extremely low Tyr(•) levels and RNR activity depending on genetic backgrounds. Here, we present evidence supporting the model that ß' is required for iron loading and Tyr(•) formation in ß in vivo via a pathway that is likely dependent on the cytosolic monothiol glutaredoxins Grx3/Grx4 and the Fe-S cluster protein Dre2. rnr4 mutants are defective in iron loading into nascent ß and are hypersensitive to iron depletion and the Tyr(•)-reducing agent hydroxyurea. Transient induction of ß' in a GalRNR4 strain leads to a concomitant increase in iron loading and Tyr(•) levels in ß. Tyr(•) can also be rapidly generated using endogenous iron when permeabilized Δrnr4 spheroplasts are supplemented with recombinant ß' and is inhibited by adding an iron chelator prior to, but not after, ß' supplementation. The growth defects of rnr4 mutants are enhanced by deficiencies in grx3/grx4 and dre2. Moreover, depletion of Dre2 in GalDRE2 cells leads to a decrease in both Tyr(•) levels and ßß' activity. This result, in combination with previous findings that a low level of Grx3/4 impairs RNR function, strongly suggests that Grx3/4 and Dre2 serve in the assembly of the deferric Tyr(•) cofactor in RNR.


Subject(s)
Glutaredoxins/metabolism , Iron-Sulfur Proteins/metabolism , Iron/metabolism , Oxidoreductases/metabolism , Ribonucleoside Diphosphate Reductase/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Tyrosine/analogs & derivatives , Glutaredoxins/genetics , Hydroxyurea , Iron-Sulfur Proteins/genetics , Mutation , Oxidoreductases/genetics , Ribonucleoside Diphosphate Reductase/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Spheroplasts/genetics , Spheroplasts/metabolism , Tyrosine/genetics , Tyrosine/metabolism
10.
Genetics ; 173(1): 63-73, 2006 May.
Article in English | MEDLINE | ID: mdl-16489218

ABSTRACT

Ribonucleotide reductase (RNR) catalyzes the rate-limiting step in de novo deoxyribonucleotide biosynthesis and is essential in DNA replication and repair. Cells have evolved complex mechanisms to modulate RNR activity during normal cell cycle progression and in response to genotoxic stress. A recently characterized mode of RNR regulation is DNA damage-induced RNR subunit redistribution. The RNR holoenzyme consists of a large subunit, R1, and a small subunit, R2. The Saccharomyces cerevisiae R2 is an Rnr2:Rnr4 heterodimer. Rnr2 generates a diferric-tyrosyl radical cofactor required for catalysis; Rnr4 facilitates cofactor assembly and stabilizes the resulting holo-heterodimer. Upon DNA damage, Rnr2 and Rnr4 undergo checkpoint-dependent, nucleus-to-cytoplasm redistribution, resulting in colocalization of R1 and R2. Here we present evidence that Rnr2 and Rnr4 are transported between the nucleus and the cytoplasm as one protein complex. Tagging either Rnr2 or Rnr4 with a nuclear export sequence causes cytoplasmic localization of both proteins. Moreover, mutations at the Rnr2:Rnr4 heterodimer interface can affect the localization of both proteins without disrupting the heterodimeric complex. Finally, the relocalization of Rnr4 appears to involve both active export and blockage of nuclear import. Our findings provide new insights into the mechanism of DNA damage-induced RNR subunit redistribution.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Biological Transport, Active , DNA Damage , Dimerization , Gene Expression , Mutation/genetics , Nuclear Export Signals/genetics , Protein Transport
11.
Proc Natl Acad Sci U S A ; 103(5): 1422-7, 2006 Jan 31.
Article in English | MEDLINE | ID: mdl-16432237

ABSTRACT

Ribonucleotide reductase (RNR) catalyzes the reduction of ribonucleotides to the corresponding deoxyribonucleotides and is an essential enzyme for DNA replication and repair. Cells have evolved intricate mechanisms to regulate RNR activity to ensure high fidelity of DNA replication during normal cell-cycle progression and of DNA repair upon genotoxic stress. The RNR holoenzyme is composed of a large subunit R1 (alpha, oligomeric state unknown) and a small subunit R2 (beta(2)). R1 binds substrates and allosteric effectors; R2 contains a diferric-tyrosyl radical [(Fe)(2)-Y.] cofactor that is required for catalysis. In Saccharomyces cerevisiae, R1 is predominantly localized in the cytoplasm, whereas R2, which is a heterodimer (betabeta'), is predominantly in the nucleus. When cells encounter DNA damage or stress during replication, betabeta' is redistributed from the nucleus to the cytoplasm in a checkpoint-dependent manner, resulting in the colocalization of R1 and R2. We have identified two proteins that have an important role in betabeta' nuclear localization: the importin beta homolog Kap122 and the WD40 repeat protein Wtm1. Deletion of either WTM1 or KAP122 leads to loss of betabeta' nuclear localization. Wtm1 and its paralog Wtm2 are both nuclear proteins that are in the same protein complex with betabeta'. Wtm1 also interacts with Kap122 in vivo and requires Kap122 for its nuclear localization. Our results suggest that Wtm1 acts either as an adaptor to facilitate nuclear import of betabeta' by Kap122 or as an anchor to retain betabeta' in the nucleus.


Subject(s)
Cell Nucleus/enzymology , Karyopherins/physiology , Repressor Proteins/physiology , Ribonucleotide Reductases/biosynthesis , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/enzymology , Transcription Factors/physiology , beta Karyopherins/physiology , Catalysis , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , Cytoplasm/metabolism , DNA Damage , DNA Replication , Dimerization , Electrophoresis, Polyacrylamide Gel , Fungal Proteins/chemistry , Gene Deletion , Gene Expression Regulation, Fungal , Genes, Fungal , Green Fluorescent Proteins/chemistry , Karyopherins/metabolism , Models, Biological , Mutation , Plasmids/metabolism , Protein Binding , Recombinant Fusion Proteins/chemistry , Repressor Proteins/chemistry , Ribonucleotide Reductases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/chemistry , beta Karyopherins/metabolism
12.
Proc Natl Acad Sci U S A ; 100(11): 6628-33, 2003 May 27.
Article in English | MEDLINE | ID: mdl-12732713

ABSTRACT

The fidelity of DNA replication and repair processes is critical for maintenance of genomic stability. Ribonucleotide reductase (RNR) catalyzes the rate-limiting step in dNTP production and thus plays an essential role in DNA synthesis. The level and activity of RNR are highly regulated by the cell cycle and DNA damage checkpoints, which maintain optimal dNTP pools required for genetic fidelity. RNRs are composed of a large subunit that binds the nucleoside diphosphate substrates and allosteric effectors and a small subunit that houses the di-iron tyrosyl radical cofactor essential for the reduction process. In Saccharomyces cerevisiae, there are two large subunits (Rnr1 and Rnr3) and two small subunits (Rnr2 and Rnr4). Here we report the subcellular localization of Rnr1-4 during normal cell growth and the redistribution of Rnr2 and Rnr4 in response to DNA damage and replicational stress. During the normal cell cycle, Rnr1 and Rnr3 are predominantly localized to the cytoplasm and Rnr2 and Rnr4 are predominantly present in the nucleus. Under genotoxic stress, Rnr2 and Rnr4 become redistributed to the cytoplasm in a checkpoint-dependent manner. Subcellular redistribution of Rnr2 and Rnr4 can occur in the absence of the transcriptional induction of the RNR genes after DNA damage and likely represents a posttranslational event. These results suggest a mechanism by which DNA damage checkpoint modulates RNR activity through the temporal and spatial regulation of its subunits.


Subject(s)
DNA Damage , DNA Replication , Ribonucleotide Reductases/metabolism , Saccharomyces cerevisiae/enzymology , Subcellular Fractions/enzymology , Fluorescent Antibody Technique, Indirect , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...