Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 78(3): 396-410.e4, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32169162

RESUMEN

The Mec1 and Rad53 kinases play a central role during acute replication stress in budding yeast. They are also essential for viability in normal growth conditions, but the signal that activates the Mec1-Rad53 pathway in the absence of exogenous insults is currently unknown. Here, we show that this pathway is active at the onset of normal S phase because deoxyribonucleotide triphosphate (dNTP) levels present in G1 phase may not be sufficient to support processive DNA synthesis and impede DNA replication. This activation can be suppressed experimentally by increasing dNTP levels in G1 phase. Moreover, we show that unchallenged cells entering S phase in the absence of Rad53 undergo irreversible fork collapse and mitotic catastrophe. Together, these data indicate that cells use suboptimal dNTP pools to detect the onset of DNA replication and activate the Mec1-Rad53 pathway, which in turn maintains functional forks and triggers dNTP synthesis, allowing the completion of DNA replication.


Asunto(s)
Replicación del ADN/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Fase S/fisiología , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinasa de Punto de Control 2/genética , Quinasa de Punto de Control 2/metabolismo , Desoxirribonucleótidos/genética , Desoxirribonucleótidos/metabolismo , Regulación Fúngica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Mitosis , Proteínas Serina-Treonina Quinasas/genética , Origen de Réplica , Saccharomyces cerevisiae/citología , Proteínas de Saccharomyces cerevisiae/genética
2.
Mol Cell ; 76(5): 784-796.e6, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31588022

RESUMEN

Oligoribonucleases are conserved enzymes that degrade short RNA molecules of up to 5 nt in length and are assumed to constitute the final stage of RNA turnover. Here we demonstrate that REXO2 is a specialized dinucleotide-degrading enzyme that shows no preference between RNA and DNA dinucleotide substrates. A heart- and skeletal-muscle-specific knockout mouse displays elevated dinucleotide levels and alterations in gene expression patterns indicative of aberrant dinucleotide-primed transcription initiation. We find that dinucleotides act as potent stimulators of mitochondrial transcription initiation in vitro. Our data demonstrate that increased levels of dinucleotides can be used to initiate transcription, leading to an increase in transcription levels from both mitochondrial promoters and other, nonspecific sequence elements in mitochondrial DNA. Efficient RNA turnover by REXO2 is thus required to maintain promoter specificity and proper regulation of transcription in mammalian mitochondria.


Asunto(s)
Proteínas 14-3-3/metabolismo , Biomarcadores de Tumor/metabolismo , Exorribonucleasas/metabolismo , Mitocondrias/enzimología , Oligonucleótidos/metabolismo , Regiones Promotoras Genéticas , Estabilidad del ARN , ARN Mitocondrial/metabolismo , Proteínas 14-3-3/deficiencia , Proteínas 14-3-3/genética , Animales , Biomarcadores de Tumor/genética , Exorribonucleasas/genética , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mitocondrial/genética , Células Sf9 , Spodoptera
3.
PLoS Genet ; 20(5): e1011148, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38776358

RESUMEN

The helicase MCM and the ribonucleotide reductase RNR are the complexes that provide the substrates (ssDNA templates and dNTPs, respectively) for DNA replication. Here, we demonstrate that MCM interacts physically with RNR and some of its regulators, including the kinase Dun1. These physical interactions encompass small subpopulations of MCM and RNR, are independent of the major subcellular locations of these two complexes, augment in response to DNA damage and, in the case of the Rnr2 and Rnr4 subunits of RNR, depend on Dun1. Partial disruption of the MCM/RNR interactions impairs the release of Rad52 -but not RPA-from the DNA repair centers despite the lesions are repaired, a phenotype that is associated with hypermutagenesis but not with alterations in the levels of dNTPs. These results suggest that a specifically regulated pool of MCM and RNR complexes plays non-canonical roles in genetic stability preventing persistent Rad52 centers and hypermutagenesis.


Asunto(s)
Proteínas de Ciclo Celular , Daño del ADN , Reparación del ADN , Replicación del ADN , Inestabilidad Genómica , Proteína Recombinante y Reparadora de ADN Rad52 , Ribonucleótido Reductasas , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Replicación del ADN/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Daño del ADN/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Proteína Recombinante y Reparadora de ADN Rad52/genética , Ribonucleótido Reductasas/genética , Ribonucleótido Reductasas/metabolismo , Reparación del ADN/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Proteínas de Mantenimiento de Minicromosoma/genética , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética , Ribonucleósido Difosfato Reductasa/genética , Ribonucleósido Difosfato Reductasa/metabolismo
4.
Nucleic Acids Res ; 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39360631

RESUMEN

Alterations in deoxyribonucleoside triphosphate (dNTP) pools have been linked to increased mutation rates and genome instability in unicellular organisms and cell cultures. However, the role of dNTP pool changes in tumor development in mammals remains unclear. In this study, we present a mouse model with a point mutation at the allosteric specificity site of ribonucleotide reductase, RRM1-Y285A. This mutation reduced ribonucleotide reductase activity, impairing the synthesis of deoxyadenosine triphosphate (dATP) and deoxyguanosine triphosphate (dGTP). Heterozygous Rrm1+/Y285A mice exhibited distinct alterations in dNTP pools across various organs, shorter lifespans and earlier tumor onset compared with wild-type controls. Mutational spectrum analysis of tumors revealed two distinct signatures, one resembling a signature extracted from a human cancer harboring a mutation of the same amino acid residue in ribonucleotide reductase, RRM1Y285C. Our findings suggest that mutations in enzymes involved in dNTP metabolism can serve as drivers of cancer development.

5.
Genes Dev ; 32(11-12): 822-835, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29899143

RESUMEN

The Mec1/ATR kinase coordinates multiple cellular responses to replication stress. In addition to its canonical role in activating the checkpoint kinase Rad53, Mec1 also plays checkpoint-independent roles in genome maintenance that are not well understood. Here we used a combined genetic-phosphoproteomic approach to manipulate Mec1 activation and globally monitor Mec1 signaling, allowing us to delineate distinct checkpoint-independent modes of Mec1 action. Using cells in which endogenous Mec1 activators were genetically ablated, we found that expression of "free" Mec1 activation domains (MADs) can robustly activate Mec1 and rescue the severe DNA replication and growth defects of these cells back to wild-type levels. However, unlike the activation mediated by endogenous activator proteins, "free" MADs are unable to stimulate Mec1-mediated suppression of gross chromosomal rearrangements (GCRs), revealing that Mec1's role in genome maintenance is separable from a previously unappreciated proreplicative function. Both Mec1's functions in promoting replication and suppressing GCRs are independent of the downstream checkpoint kinases. Additionally, Mec1-dependent GCR suppression seems to require localized Mec1 action at DNA lesions, which correlates with the phosphorylation of activator-proximal substrates involved in homologous recombination-mediated DNA repair. These findings establish that Mec1 initiates checkpoint signaling, promotes DNA replication, and maintains genetic stability through distinct modes of action.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Replicación del ADN/genética , Genoma Fúngico/genética , Saccharomyces cerevisiae/genética , Transducción de Señal/genética , Activación Enzimática/genética , Inestabilidad Genómica/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mutación , Fosforilación , Dominios Proteicos/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteómica , Proteínas Recombinantes de Fusión/metabolismo , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
6.
Mol Cell ; 68(2): 446-455.e3, 2017 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-29033319

RESUMEN

The checkpoint kinase Rad53 is activated during replication stress to prevent fork collapse, an essential but poorly understood process. Here we show that Rad53 couples leading- and lagging-strand synthesis under replication stress. In rad53-1 cells stressed by dNTP depletion, the replicative DNA helicase, MCM, and the leading-strand DNA polymerase, Pol ε, move beyond the site of DNA synthesis, likely unwinding template DNA. Remarkably, DNA synthesis progresses further along the lagging strand than the leading strand, resulting in the exposure of long stretches of single-stranded leading-strand template. The asymmetric DNA synthesis in rad53-1 cells is suppressed by elevated levels of dNTPs in vivo, and the activity of Pol ε is compromised more than lagging-strand polymerase Pol δ at low dNTP concentrations in vitro. Therefore, we propose that Rad53 prevents the generation of excessive ssDNA under replication stress by coordinating DNA unwinding with synthesis of both strands.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Quinasa de Punto de Control 2/metabolismo , ADN Polimerasa III/metabolismo , ADN Polimerasa II/metabolismo , Replicación del ADN/fisiología , ADN de Hongos/biosíntesis , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/genética , Quinasa de Punto de Control 2/genética , ADN Polimerasa II/genética , ADN Polimerasa III/genética , ADN de Hongos/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
7.
Nature ; 557(7703): 57-61, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29670289

RESUMEN

SAMHD1 was previously characterized as a dNTPase that protects cells from viral infections. Mutations in SAMHD1 are implicated in cancer development and in a severe congenital inflammatory disease known as Aicardi-Goutières syndrome. The mechanism by which SAMHD1 protects against cancer and chronic inflammation is unknown. Here we show that SAMHD1 promotes degradation of nascent DNA at stalled replication forks in human cell lines by stimulating the exonuclease activity of MRE11. This function activates the ATR-CHK1 checkpoint and allows the forks to restart replication. In SAMHD1-depleted cells, single-stranded DNA fragments are released from stalled forks and accumulate in the cytosol, where they activate the cGAS-STING pathway to induce expression of pro-inflammatory type I interferons. SAMHD1 is thus an important player in the replication stress response, which prevents chronic inflammation by limiting the release of single-stranded DNA from stalled replication forks.


Asunto(s)
Replicación del ADN , Interferón Tipo I/metabolismo , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Citosol/metabolismo , ADN de Cadena Simple/metabolismo , Células HEK293 , Células HeLa , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/prevención & control , Interferón Tipo I/inmunología , Proteína Homóloga de MRE11/metabolismo , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , RecQ Helicasas/metabolismo , Proteína 1 que Contiene Dominios SAM y HD/deficiencia
8.
Nucleic Acids Res ; 50(3): e18, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-34850106

RESUMEN

Information about the cellular concentrations of deoxyribonucleoside triphosphates (dNTPs) is instrumental for mechanistic studies of DNA replication and for understanding diseases caused by defects in dNTP metabolism. The dNTPs are measured by methods based on either HPLC or DNA polymerization. An advantage with the HPLC-based techniques is that the parallel analysis of ribonucleoside triphosphates (rNTPs) can serve as an internal quality control of nucleotide integrity and extraction efficiency. We have developed a Freon-free trichloroacetic acid-based method to extract cellular nucleotides and an isocratic reverse phase HPLC-based technique that is able to separate dNTPs, rNTPs and ADP in a single run. The ability to measure the ADP levels improves the control of nucleotide integrity, and the use of an isocratic elution overcomes the shifting baseline problems in previously developed gradient-based reversed phase protocols for simultaneously measuring dNTPs and rNTPs. An optional DNA-polymerase-dependent step is used for confirmation that the dNTP peaks do not overlap with other components of the extracts, further increasing the reliability of the analysis. The method is compatible with a wide range of biological samples and has a sensitivity better than other UV-based HPLC protocols, closely matching that of mass spectrometry-based detection.


Asunto(s)
Cromatografía Líquida de Alta Presión , Desoxirribonucleótidos , Ribonucleótidos/análisis , Adenosina Difosfato , Cromatografía Líquida de Alta Presión/métodos , ADN , Desoxirribonucleótidos/análisis , Reproducibilidad de los Resultados
9.
Proc Natl Acad Sci U S A ; 117(25): 14306-14313, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32513727

RESUMEN

Ribonucleotides (rNMPs) incorporated in the nuclear genome are a well-established threat to genome stability and can result in DNA strand breaks when not removed in a timely manner. However, the presence of a certain level of rNMPs is tolerated in mitochondrial DNA (mtDNA) although aberrant mtDNA rNMP content has been identified in disease models. We investigated the effect of incorporated rNMPs on mtDNA stability over the mouse life span and found that the mtDNA rNMP content increased during early life. The rNMP content of mtDNA varied greatly across different tissues and was defined by the rNTP/dNTP ratio of the tissue. Accordingly, mtDNA rNMPs were nearly absent in SAMHD1-/- mice that have increased dNTP pools. The near absence of rNMPs did not, however, appreciably affect mtDNA copy number or the levels of mtDNA molecules with deletions or strand breaks in aged animals near the end of their life span. The physiological rNMP load therefore does not contribute to the progressive loss of mtDNA quality that occurs as mice age.


Asunto(s)
ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Inestabilidad Genómica/fisiología , Ribonucleótidos/genética , Ribonucleótidos/metabolismo , Animales , Daño del ADN , Femenino , Dosificación de Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nucleótidos , Proteína 1 que Contiene Dominios SAM y HD/genética
10.
EMBO J ; 37(21)2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30158111

RESUMEN

The S-phase checkpoint maintains the integrity of the genome in response to DNA replication stress. In budding yeast, this pathway is initiated by Mec1 and is amplified through the activation of Rad53 by two checkpoint mediators: Mrc1 promotes Rad53 activation at stalled forks, and Rad9 is a general mediator of the DNA damage response. Here, we have investigated the interplay between Mrc1 and Rad9 in response to DNA damage and found that they control DNA replication through two distinct but complementary mechanisms. Mrc1 rapidly activates Rad53 at stalled forks and represses late-firing origins but is unable to maintain this repression over time. Rad9 takes over Mrc1 to maintain a continuous checkpoint signaling. Importantly, the Rad9-mediated activation of Rad53 slows down fork progression, supporting the view that the S-phase checkpoint controls both the initiation and the elongation of DNA replication in response to DNA damage. Together, these data indicate that Mrc1 and Rad9 play distinct functions that are important to ensure an optimal completion of S phase under replication stress conditions.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Replicación del ADN , ADN de Hongos/biosíntesis , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/genética , Quinasa de Punto de Control 2/genética , Quinasa de Punto de Control 2/metabolismo , ADN de Hongos/genética , Fase S/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
11.
EMBO J ; 37(17)2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-30065069

RESUMEN

Generation of single-stranded DNA (ssDNA) is required for the template strand formation during DNA replication. Replication Protein A (RPA) is an ssDNA-binding protein essential for protecting ssDNA at replication forks in eukaryotic cells. While significant progress has been made in characterizing the role of the RPA-ssDNA complex, how RPA is loaded at replication forks remains poorly explored. Here, we show that the Saccharomyces cerevisiae protein regulator of Ty1 transposition 105 (Rtt105) binds RPA and helps load it at replication forks. Cells lacking Rtt105 exhibit a dramatic reduction in RPA loading at replication forks, compromised DNA synthesis under replication stress, and increased genome instability. Mechanistically, we show that Rtt105 mediates the RPA-importin interaction and also promotes RPA binding to ssDNA directly in vitro, but is not present in the final RPA-ssDNA complex. Single-molecule studies reveal that Rtt105 affects the binding mode of RPA to ssDNA These results support a model in which Rtt105 functions as an RPA chaperone that escorts RPA to the nucleus and facilitates its loading onto ssDNA at replication forks.


Asunto(s)
Genoma Fúngico , Inestabilidad Genómica , Modelos Biológicos , Chaperonas Moleculares/metabolismo , Proteína de Replicación A/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , ADN de Hongos/genética , ADN de Hongos/metabolismo , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Carioferinas/genética , Carioferinas/metabolismo , Chaperonas Moleculares/genética , Proteína de Replicación A/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
12.
Nucleic Acids Res ; 48(8): 4274-4297, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32187369

RESUMEN

Cellular levels of ribonucleoside triphosphates (rNTPs) are much higher than those of deoxyribonucleoside triphosphates (dNTPs), thereby influencing the frequency of incorporation of ribonucleoside monophosphates (rNMPs) by DNA polymerases (Pol) into DNA. RNase H2-initiated ribonucleotide excision repair (RER) efficiently removes single rNMPs in genomic DNA. However, processing of rNMPs by Topoisomerase 1 (Top1) in absence of RER induces mutations and genome instability. Here, we greatly increased the abundance of genomic rNMPs in Saccharomyces cerevisiae by depleting Rnr1, the major subunit of ribonucleotide reductase, which converts ribonucleotides to deoxyribonucleotides. We found that in strains that are depleted of Rnr1, RER-deficient, and harbor an rNTP-permissive replicative Pol mutant, excessive accumulation of single genomic rNMPs severely compromised growth, but this was reversed in absence of Top1. Thus, under Rnr1 depletion, limited dNTP pools slow DNA synthesis by replicative Pols and provoke the incorporation of high levels of rNMPs in genomic DNA. If a threshold of single genomic rNMPs is exceeded in absence of RER and presence of limited dNTP pools, Top1-mediated genome instability leads to severe growth defects. Finally, we provide evidence showing that accumulation of RNA/DNA hybrids in absence of RNase H1 and RNase H2 leads to cell lethality under Rnr1 depletion.


Asunto(s)
ADN-Topoisomerasas de Tipo I/metabolismo , Ribonucleótido Reductasas/genética , Ribonucleótidos/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Daño del ADN , Desoxirribonucleótidos/metabolismo , Genoma Fúngico , Inestabilidad Genómica , Mutación , Ribonucleasa H/genética , Ribonucleasas/genética , Puntos de Control de la Fase S del Ciclo Celular , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/metabolismo , Eliminación de Secuencia
13.
Nucleic Acids Res ; 48(1): 264-277, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31647103

RESUMEN

The accumulation of mutations is frequently associated with alterations in gene function leading to the onset of diseases, including cancer. Aiming to find novel genes that contribute to the stability of the genome, we screened the Saccharomyces cerevisiae deletion collection for increased mutator phenotypes. Among the identified genes, we discovered MET7, which encodes folylpolyglutamate synthetase (FPGS), an enzyme that facilitates several folate-dependent reactions including the synthesis of purines, thymidylate (dTMP) and DNA methylation. Here, we found that Met7-deficient strains show elevated mutation rates, but also increased levels of endogenous DNA damage resulting in gross chromosomal rearrangements (GCRs). Quantification of deoxyribonucleotide (dNTP) pools in cell extracts from met7Δ mutant revealed reductions in dTTP and dGTP that cause a constitutively active DNA damage checkpoint. In addition, we found that the absence of Met7 leads to dUTP accumulation, at levels that allowed its detection in yeast extracts for the first time. Consequently, a high dUTP/dTTP ratio promotes uracil incorporation into DNA, followed by futile repair cycles that compromise both mitochondrial and nuclear DNA integrity. In summary, this work highlights the importance of folate polyglutamylation in the maintenance of nucleotide homeostasis and genome stability.


Asunto(s)
Nucleótidos de Desoxiuracil/metabolismo , Ácido Fólico/metabolismo , Genoma Fúngico , Péptido Sintasas/genética , Saccharomyces cerevisiae/genética , Nucleótidos de Timina/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Daño del ADN , ADN de Hongos/genética , ADN de Hongos/metabolismo , Nucleótidos de Desoxiguanina/metabolismo , Eliminación de Gen , Regulación Fúngica de la Expresión Génica , Inestabilidad Genómica , Mitocondrias/genética , Mitocondrias/metabolismo , Mutación , Péptido Sintasas/deficiencia , Saccharomyces cerevisiae/metabolismo , Uracilo/metabolismo
14.
PLoS Genet ; 15(8): e1008136, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31381575

RESUMEN

The S-phase checkpoint plays an essential role in regulation of the ribonucleotide reductase (RNR) activity to maintain the dNTP pools. How eukaryotic cells respond appropriately to different levels of replication threats remains elusive. Here, we have identified that a conserved GSK-3 kinase Mck1 cooperates with Dun1 in regulating this process. Deleting MCK1 sensitizes dun1Δ to hydroxyurea (HU) reminiscent of mec1Δ or rad53Δ. While Mck1 is downstream of Rad53, it does not participate in the post-translational regulation of RNR as Dun1 does. Mck1 phosphorylates and releases the Crt1 repressor from the promoters of DNA damage-inducible genes as RNR2-4 and HUG1. Hug1, an Rnr2 inhibitor normally silenced, is induced as a counterweight to excessive RNR. When cells suffer a more severe threat, Mck1 inhibits HUG1 transcription. Consistently, only a combined deletion of HUG1 and CRT1, confers a dramatic boost of dNTP levels and the survival of mck1Δdun1Δ or mec1Δ cells assaulted by a lethal dose of HU. These findings reveal the division-of-labor between Mck1 and Dun1 at the S-phase checkpoint pathway to fine-tune dNTP homeostasis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Regulación Fúngica de la Expresión Génica/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/fisiología , Proteínas de Ciclo Celular/genética , Daño del ADN , Replicación del ADN/efectos de los fármacos , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Glucógeno Sintasa Quinasa 3/genética , Hidroxiurea/toxicidad , Nucleótidos/metabolismo , Fosforilación , Regiones Promotoras Genéticas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Ribonucleótido Reductasas/genética , Ribonucleótido Reductasas/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Saccharomyces cerevisiae/efectos de los fármacos , Proteínas de Saccharomyces cerevisiae/genética
15.
Psychol Health Med ; : 1-8, 2022 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-36120800

RESUMEN

The Covid-19 pandemic, which was declared a public health emergency on 30 January 2020, has made it crucial for humans to learn how to behave to control the pandemic's spread. Policymakers must assess human behaviour and their responses to pandemic breakouts to develop a strategy for limiting pandemics and their harm to society at large. The present study applying exploratory factor analysis assessed five aspects of human behaviour regarding Covid-19, namely compliance behaviour, avoidance behaviour, protective behaviour, informed behaviour, and risk perception. The study applying hierarchical regression discovered that by combining informed, protective, and avoidance behaviour, people can be convinced to embrace the compliance behaviour required by public authorities. Furthermore, higher risk perception also positively moderates the relationship between information and compliance behaviour.

16.
Nucleic Acids Res ; 47(8): 3986-3995, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30698744

RESUMEN

The four B-family DNA polymerases α, δ, ϵ and ζ cooperate to accurately replicate the eukaryotic nuclear genome. Here, we report that a Saccharomyces cerevisiae strain encoding the pol2-16 mutation that lacks Pol ϵ's polymerase and exonuclease activities has increased dNTP concentrations and an increased mutation rate at the CAN1 locus compared to wild type yeast. About half of this mutagenesis disappears upon deleting the REV3 gene encoding the catalytic subunit of Pol ζ. The remaining, still strong, mutator phenotype is synergistically elevated in an msh6Δ strain and has a mutation spectrum characteristic of mistakes made by Pol δ. The results support a model wherein slow-moving replication forks caused by the lack of Pol ϵ's catalytic domains result in greater involvement of mutagenic DNA synthesis by Pol ζ as well as diminished proofreading by Pol δ during replication.


Asunto(s)
ADN Polimerasa II/genética , ADN de Hongos/genética , ADN Polimerasa Dirigida por ADN/genética , Regulación Fúngica de la Expresión Génica , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Dominio Catalítico , ADN Polimerasa II/metabolismo , Replicación del ADN , ADN de Hongos/metabolismo , ADN Polimerasa Dirigida por ADN/deficiencia , Eliminación de Gen , Tasa de Mutación , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
17.
Nucleic Acids Res ; 47(1): 237-252, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30462295

RESUMEN

The balance and the overall concentration of intracellular deoxyribonucleoside triphosphates (dNTPs) are important determinants of faithful DNA replication. Despite the established fact that changes in dNTP pools negatively influence DNA replication fidelity, it is not clear why certain dNTP pool alterations are more mutagenic than others. As intracellular dNTP pools are mainly controlled by ribonucleotide reductase (RNR), and given the limited number of eukaryotic RNR mutations characterized so far, we screened for RNR1 mutations causing mutator phenotypes in Saccharomyces cerevisiae. We identified 24 rnr1 mutant alleles resulting in diverse mutator phenotypes linked in most cases to imbalanced dNTPs. Among the identified rnr1 alleles the strongest mutators presented a dNTP imbalance in which three out of the four dNTPs were elevated (dCTP, dTTP and dGTP), particularly if dGTP levels were highly increased. These rnr1 alleles caused growth defects/lethality in DNA replication fidelity-compromised backgrounds, and caused strong mutator phenotypes even in the presence of functional DNA polymerases and mismatch repair. In summary, this study pinpoints key residues that contribute to allosteric regulation of RNR's overall activity or substrate specificity. We propose a model that distinguishes between different dNTP pool alterations and provides a mechanistic explanation why certain dNTP imbalances are particularly detrimental.


Asunto(s)
Replicación del ADN/genética , Desoxirribonucleótidos/genética , Ribonucleótido Reductasas/genética , Proteínas de Saccharomyces cerevisiae/genética , Alelos , Reparación de la Incompatibilidad de ADN/genética , ADN Polimerasa Dirigida por ADN/genética , Homeostasis , Mutación/genética , Saccharomyces cerevisiae/genética
18.
J Biol Chem ; 294(44): 15889-15897, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31300555

RESUMEN

The building blocks of DNA, dNTPs, can be produced de novo or can be salvaged from deoxyribonucleosides. However, to what extent the absence of de novo dNTP production can be compensated for by the salvage pathway is unknown. Here, we eliminated de novo dNTP synthesis in the mouse heart and skeletal muscle by inactivating ribonucleotide reductase (RNR), a key enzyme for the de novo production of dNTPs, at embryonic day 13. All other tissues had normal de novo dNTP synthesis and theoretically could supply heart and skeletal muscle with deoxyribonucleosides needed for dNTP production by salvage. We observed that the dNTP and NTP pools in WT postnatal hearts are unexpectedly asymmetric, with unusually high dGTP and GTP levels compared with those in whole mouse embryos or murine cell cultures. We found that RNR inactivation in heart led to strongly decreased dGTP and increased dCTP, dTTP, and dATP pools; aberrant DNA replication; defective expression of muscle-specific proteins; progressive heart abnormalities; disturbance of the cardiac conduction system; and lethality between the second and fourth weeks after birth. We conclude that dNTP salvage cannot substitute for de novo dNTP synthesis in the heart and that cardiomyocytes and myocytes initiate DNA replication despite an inadequate dNTP supply. We discuss the possible reasons for the observed asymmetry in dNTP and NTP pools in WT hearts.


Asunto(s)
Desoxirribonucleótidos/biosíntesis , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/metabolismo , Animales , Replicación del ADN , Corazón/fisiología , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Ribonucleótido Reductasas/genética , Ribonucleótido Reductasas/metabolismo
19.
PLoS Genet ; 13(10): e1007082, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29069086

RESUMEN

Ribonucleotide reductase (RNR) provides the precursors for the generation of dNTPs, which are required for DNA synthesis and repair. Here, we investigated the function of the major RNR subunits Rnr1 and Rnr3 in telomere elongation in budding yeast. We show that Rnr1 is essential for the sustained elongation of short telomeres by telomerase. In the absence of Rnr1, cells harbor very short, but functional, telomeres, which cannot become elongated by increased telomerase activity or by tethering of telomerase to telomeres. Furthermore, we demonstrate that Rnr1 function is critical to prevent an early onset of replicative senescence and premature survivor formation in telomerase-negative cells but dispensable for telomere elongation by Homology-Directed-Repair. Our results suggest that telomerase has a "basal activity" mode that is sufficient to compensate for the "end-replication-problem" and does not require the presence of Rnr1 and a different "sustained activity" mode necessary for the elongation of short telomeres, which requires an upregulation of dNTP levels and dGTP ratios specifically through Rnr1 function. By analyzing telomere length and dNTP levels in different mutants showing changes in RNR complex composition and activity we provide evidence that the Mec1ATR checkpoint protein promotes telomere elongation by increasing both dNTP levels and dGTP ratios through Rnr1 upregulation in a mechanism that cannot be replaced by its homolog Rnr3.


Asunto(s)
Ribonucleótido Reductasas/genética , Saccharomycetales/genética , Telomerasa/metabolismo , Homeostasis del Telómero , Telómero , Senescencia Celular , Replicación del ADN , Saccharomycetales/citología , Saccharomycetales/crecimiento & desarrollo , Saccharomycetales/metabolismo , Telomerasa/genética
20.
Proc Natl Acad Sci U S A ; 114(22): E4442-E4451, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28416670

RESUMEN

Eukaryotic DNA replication fidelity relies on the concerted action of DNA polymerase nucleotide selectivity, proofreading activity, and DNA mismatch repair (MMR). Nucleotide selectivity and proofreading are affected by the balance and concentration of deoxyribonucleotide (dNTP) pools, which are strictly regulated by ribonucleotide reductase (RNR). Mutations preventing DNA polymerase proofreading activity or MMR function cause mutator phenotypes and consequently increased cancer susceptibility. To identify genes not previously linked to high-fidelity DNA replication, we conducted a genome-wide screen in Saccharomyces cerevisiae using DNA polymerase active-site mutants as a "sensitized mutator background." Among the genes identified in our screen, three metabolism-related genes (GLN3, URA7, and SHM2) have not been previously associated to the suppression of mutations. Loss of either the transcription factor Gln3 or inactivation of the CTP synthetase Ura7 both resulted in the activation of the DNA damage response and imbalanced dNTP pools. Importantly, these dNTP imbalances are strongly mutagenic in genetic backgrounds where DNA polymerase function or MMR activity is partially compromised. Previous reports have shown that dNTP pool imbalances can be caused by mutations altering the allosteric regulation of enzymes involved in dNTP biosynthesis (e.g., RNR or dCMP deaminase). Here, we provide evidence that mutations affecting genes involved in RNR substrate production can cause dNTP imbalances, which cannot be compensated by RNR or other enzymatic activities. Moreover, Gln3 inactivation links nutrient deprivation to increased mutagenesis. Our results suggest that similar genetic interactions could drive mutator phenotypes in cancer cells.


Asunto(s)
Reparación de la Incompatibilidad de ADN/genética , Replicación del ADN/genética , Mutagénesis/genética , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/genética , Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/metabolismo , Daño del ADN/genética , Fosfatos de Dinucleósidos/genética , Fosfatos de Dinucleósidos/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA