Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Mol Cell ; 65(1): 78-90, 2017 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-27916662

RESUMO

During DNA replication, proliferating cell nuclear antigen (PCNA) adopts a ring-shaped structure to promote processive DNA synthesis, acting as a sliding clamp for polymerases. Known posttranslational modifications function at the outer surface of the PCNA ring to favor DNA damage bypass. Here, we demonstrate that acetylation of lysine residues at the inner surface of PCNA is induced by DNA lesions. We show that cohesin acetyltransferase Eco1 targets lysine 20 at the sliding surface of the PCNA ring in vitro and in vivo in response to DNA damage. Mimicking constitutive acetylation stimulates homologous recombination and robustly suppresses the DNA damage sensitivity of mutations in damage tolerance pathways. In comparison to the unmodified trimer, structural differences are observed at the interface between protomers in the crystal structure of the PCNA-K20ac ring. Thus, acetylation regulates PCNA sliding on DNA in the presence of DNA damage, favoring homologous recombination linked to sister-chromatid cohesion.


Assuntos
Acetiltransferases/metabolismo , Cromátides , Cromossomos Fúngicos , Dano ao DNA , Instabilidade Genômica , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Processamento de Proteína Pós-Traducional , Reparo de DNA por Recombinação , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Acetilação , Acetiltransferases/química , Acetiltransferases/genética , DNA Polimerase III/genética , DNA Polimerase III/metabolismo , Genótipo , Humanos , Lisina , Modelos Moleculares , Mutação , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fenótipo , Antígeno Nuclear de Célula em Proliferação/química , Antígeno Nuclear de Célula em Proliferação/genética , Conformação Proteica , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Relação Estrutura-Atividade
2.
J Proteome Res ; 22(8): 2765-2773, 2023 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-37463329

RESUMO

Current protocols used to extract and purify histones are notoriously tedious, especially when using yeast cells. Here, we describe the use of a simple filter-aided sample preparation approach enabling histone extraction from yeast and mammalian cells using acidified ethanol, which not only improves extraction but also inactivates histone-modifying enzymes. We show that our improved method prevents N-terminal clipping of H3, an artifact frequently observed in yeast cells using standard histone extraction protocols. Our method is scalable and provides efficient recovery of histones when extracts are prepared from as few as two million yeast cells. We further demonstrate the application of this approach for the analysis of histone modifications in fungal clinical isolates available in a limited quantity. Compared with standard protocols, our method enables the study of histones and their modifications in a faster, simpler, and more robust manner.


Assuntos
Histonas , Saccharomyces cerevisiae , Animais , Histonas/metabolismo , Saccharomyces cerevisiae/metabolismo , Processamento de Proteína Pós-Traducional , Código das Histonas , Mamíferos/metabolismo
3.
Cell ; 134(2): 244-55, 2008 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-18662540

RESUMO

Chromatin assembly factor 1 (CAF-1) and Rtt106 participate in the deposition of newly synthesized histones onto replicating DNA to form nucleosomes. This process is critical for the maintenance of genome stability and inheritance of functionally specialized chromatin structures in proliferating cells. However, the molecular functions of the acetylation of newly synthesized histones in this DNA replication-coupled nucleosome assembly pathway remain enigmatic. Here we show that histone H3 acetylated at lysine 56 (H3K56Ac) is incorporated onto replicating DNA and, by increasing the binding affinity of CAF-1 and Rtt106 for histone H3, H3K56Ac enhances the ability of these histone chaperones to assemble DNA into nucleosomes. Genetic analysis indicates that H3K56Ac acts in a nonredundant manner with the acetylation of the N-terminal residues of H3 and H4 in nucleosome assembly. These results reveal a mechanism by which H3K56Ac regulates replication-coupled nucleosome assembly mediated by CAF-1 and Rtt106.


Assuntos
Replicação do DNA , Histonas/metabolismo , Nucleossomos/metabolismo , Saccharomyces cerevisiae/metabolismo , Acetilação , Dano ao DNA , DNA Fúngico/metabolismo , Histonas/química , Humanos , Lisina/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mutação , Estrutura Terciária de Proteína , Ribonucleases/metabolismo , Fase S , Saccharomyces cerevisiae/citologia , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
4.
Int J Mol Sci ; 23(19)2022 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-36232396

RESUMO

The eukaryotic DNA replication fork is a hub of enzymes that continuously act to synthesize DNA, propagate DNA methylation and other epigenetic marks, perform quality control, repair nascent DNA, and package this DNA into chromatin. Many of the enzymes involved in these spatiotemporally correlated processes perform their functions by binding to proliferating cell nuclear antigen (PCNA). A long-standing question has been how the plethora of PCNA-binding enzymes exert their activities without interfering with each other. As a first step towards deciphering this complex regulation, we studied how Chromatin Assembly Factor 1 (CAF-1) binds to PCNA. We demonstrate that CAF-1 binds to PCNA in a heretofore uncharacterized manner that depends upon a cation-pi (π) interaction. An arginine residue, conserved among CAF-1 homologs but absent from other PCNA-binding proteins, inserts into the hydrophobic pocket normally occupied by proteins that contain canonical PCNA interaction peptides (PIPs). Mutation of this arginine disrupts the ability of CAF-1 to bind PCNA and to assemble chromatin. The PIP of the CAF-1 p150 subunit resides at the extreme C-terminus of an apparent long α-helix (119 amino acids) that has been reported to bind DNA. The length of that helix and the presence of a PIP at the C-terminus are evolutionarily conserved among numerous species, ranging from yeast to humans. This arrangement of a very long DNA-binding coiled-coil that terminates in PIPs may serve to coordinate DNA and PCNA binding by CAF-1.


Assuntos
Cromatina , Replicação do DNA , Aminoácidos/metabolismo , Arginina/metabolismo , Cromatina/genética , Cromatina/metabolismo , Fator 1 de Modelagem da Cromatina/química , Fator 1 de Modelagem da Cromatina/genética , Fator 1 de Modelagem da Cromatina/metabolismo , DNA/metabolismo , Humanos , Peptídeos/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
5.
Proc Natl Acad Sci U S A ; 113(5): 1393-8, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26764384

RESUMO

Oncogenic transcription factors are commonly activated in acute leukemias and subvert normal gene expression networks to reprogram hematopoietic progenitors into preleukemic stem cells, as exemplified by LIM-only 2 (LMO2) in T-cell acute lymphoblastic leukemia (T-ALL). Whether or not these oncoproteins interfere with other DNA-dependent processes is largely unexplored. Here, we show that LMO2 is recruited to DNA replication origins by interaction with three essential replication enzymes: DNA polymerase delta (POLD1), DNA primase (PRIM1), and minichromosome 6 (MCM6). Furthermore, tethering LMO2 to synthetic DNA sequences is sufficient to transform these sequences into origins of replication. We next addressed the importance of LMO2 in erythroid and thymocyte development, two lineages in which cell cycle and differentiation are tightly coordinated. Lowering LMO2 levels in erythroid progenitors delays G1-S progression and arrests erythropoietin-dependent cell growth while favoring terminal differentiation. Conversely, ectopic expression in thymocytes induces DNA replication and drives these cells into cell cycle, causing differentiation blockade. Our results define a novel role for LMO2 in directly promoting DNA synthesis and G1-S progression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Replicação do DNA/genética , Células-Tronco Hematopoéticas/metabolismo , Proteínas com Domínio LIM/genética , Animais , Células-Tronco Hematopoéticas/citologia , Camundongos , Origem de Replicação , Fase S
6.
J Proteome Res ; 15(7): 2132-42, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27223649

RESUMO

Histone deacetylases (HDACs) catalyze the removal of acetylation marks from lysine residues on histone and nonhistone substrates. Their activity is generally associated with essential cellular processes such as transcriptional repression and heterochromatin formation. Interestingly, abnormal activity of HDACs has been reported in various types of cancers, which makes them a promising therapeutic target for cancer treatment. In the current study, we aim to understand the mechanisms underlying the function of HDACs using an in-depth quantitative analysis of changes in histone acetylation levels in Schizosaccharomyces pombe (S. pombe) lacking major HDAC activities. We employed a targeted quantitative mass spectrometry approach to profile changes of acetylation and methylation at multiple lysine residues on the N-terminal tail of histones H3 and H4. Our analyses identified a number of histone acetylation sites that are significantly affected by S. pombe HDAC mutations. We discovered that mutation of the Class I HDAC known as Clr6 causes a major increase in the abundance of triacetylated H4 molecules at K5, K8, and K12. A clr6-1 hypomorphic mutation also increased the abundance of multiple acetyl-lysines in histone H3. In addition, our study uncovered a few crosstalks between histone acetylation and methylation upon deletion of HDACs Hos2 and Clr3. We anticipate that the results from this study will greatly improve our current understanding of the mechanisms involved in HDAC-mediated gene regulation and heterochromatin assembly.


Assuntos
Código das Histonas , Histona Desacetilases/genética , Histonas/metabolismo , Mutação , Proteínas de Schizosaccharomyces pombe/metabolismo , Acetilação , Espectrometria de Massas/métodos , Metilação , Proteínas de Schizosaccharomyces pombe/análise
7.
EMBO J ; 31(1): 58-70, 2012 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-22020126

RESUMO

The MYST protein lysine acetyltransferases are evolutionarily conserved throughout eukaryotes and acetylate proteins to regulate diverse biological processes including gene regulation, DNA repair, cell-cycle regulation, stem cell homeostasis and development. Here, we demonstrate that MYST protein acetyltransferase activity requires active site lysine autoacetylation. The X-ray crystal structures of yeast Esa1 (yEsa1/KAT5) bound to a bisubstrate H4K16CoA inhibitor and human MOF (hMOF/KAT8/MYST1) reveal that they are autoacetylated at a strictly conserved lysine residue in MYST proteins (yEsa1-K262 and hMOF-K274) in the enzyme active site. The structure of hMOF also shows partial occupancy of K274 in the unacetylated form, revealing that the side chain reorients to a position that engages the catalytic glutamate residue and would block cognate protein substrate binding. Consistent with the structural findings, we present mass spectrometry data and biochemical experiments to demonstrate that this lysine autoacetylation on yEsa1, hMOF and its yeast orthologue, ySas2 (KAT8) occurs in solution and is required for acetylation and protein substrate binding in vitro. We also show that this autoacetylation occurs in vivo and is required for the cellular functions of these MYST proteins. These findings provide an avenue for the autoposttranslational regulation of MYST proteins that is distinct from other acetyltransferases but draws similarities to the phosphoregulation of protein kinases.


Assuntos
Histona Acetiltransferases/metabolismo , Lisina/genética , Acetilação , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Histona Acetiltransferases/genética , Histonas/metabolismo , Humanos , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
8.
J Biol Chem ; 289(19): 13186-96, 2014 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-24648511

RESUMO

In Saccharomyces cerevisiae, histone H3 lysine 56 acetylation (H3K56ac) is a modification of new H3 molecules deposited throughout the genome during S-phase. H3K56ac is removed by the sirtuins Hst3 and Hst4 at later stages of the cell cycle. Previous studies indicated that regulated degradation of Hst3 plays an important role in the genome-wide waves of H3K56 acetylation and deacetylation that occur during each cell cycle. However, little is known regarding the mechanism of cell cycle-regulated Hst3 degradation. Here, we demonstrate that Hst3 instability in vivo is dependent upon the ubiquitin ligase SCF(Cdc4) and that Hst3 is phosphorylated at two Cdk1 sites, threonine 380 and threonine 384. This creates a diphosphorylated degron that is necessary for Hst3 polyubiquitylation by SCF(Cdc4). Mutation of the Hst3 diphospho-degron does not completely stabilize Hst3 in vivo, but it nonetheless results in a significant fitness defect that is particularly severe in mutant cells treated with the alkylating agent methyl methanesulfonate. Unexpectedly, we show that Hst3 can be degraded between G2 and anaphase, a window of the cell cycle where Hst3 normally mediates genome-wide deacetylation of H3K56. Our results suggest an intricate coordination between Hst3 synthesis, genome-wide H3K56 deacetylation by Hst3, and cell cycle-regulated degradation of Hst3 by cyclin-dependent kinases and SCF(Cdc4).


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiologia , Proteínas F-Box/metabolismo , Genoma Fúngico/fisiologia , Histona Desacetilases/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/fisiologia , Acetilação , Proteínas de Ciclo Celular/genética , Estabilidade Enzimática/fisiologia , Proteínas F-Box/genética , Histona Desacetilases/genética , Histonas/genética , Histonas/metabolismo , Fosforilação/fisiologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Ubiquitina-Proteína Ligases/genética
9.
PLoS Genet ; 7(3): e1001354, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21483810

RESUMO

Methylation of histone H3 lysine 4 (H3K4me) is an evolutionarily conserved modification whose role in the regulation of gene expression has been extensively studied. In contrast, the function of H3K4 acetylation (H3K4ac) has received little attention because of a lack of tools to separate its function from that of H3K4me. Here we show that, in addition to being methylated, H3K4 is also acetylated in budding yeast. Genetic studies reveal that the histone acetyltransferases (HATs) Gcn5 and Rtt109 contribute to H3K4 acetylation in vivo. Whilst removal of H3K4ac from euchromatin mainly requires the histone deacetylase (HDAC) Hst1, Sir2 is needed for H3K4 deacetylation in heterochomatin. Using genome-wide chromatin immunoprecipitation (ChIP), we show that H3K4ac is enriched at promoters of actively transcribed genes and located just upstream of H3K4 tri-methylation (H3K4me3), a pattern that has been conserved in human cells. We find that the Set1-containing complex (COMPASS), which promotes H3K4me2 and -me3, also serves to limit the abundance of H3K4ac at gene promoters. In addition, we identify a group of genes that have high levels of H3K4ac in their promoters and are inadequately expressed in H3-K4R, but not in set1Δ mutant strains, suggesting that H3K4ac plays a positive role in transcription. Our results reveal a novel regulatory feature of promoter-proximal chromatin, involving mutually exclusive histone modifications of the same histone residue (H3K4ac and H3K4me).


Assuntos
Histonas/metabolismo , Lisina/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Acetilação , Eucromatina/genética , Eucromatina/metabolismo , Regulação Enzimológica da Expressão Gênica , Redes Reguladoras de Genes/genética , Heterocromatina/genética , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/genética , Lisina/genética , Metilação , Regiões Promotoras Genéticas , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/genética , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/metabolismo , Sirtuína 2/genética , Sirtuína 2/metabolismo
10.
Curr Opin Cell Biol ; 18(2): 137-44, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16487697

RESUMO

The packaging of DNA into chromatin creates a number of significant barriers to the detection of DNA lesions and their timely and accurate repair. Eukaryotic cells have evolved a number of enzymes that modulate chromatin structure and facilitate DNA repair. Recent research illustrates how nucleosome remodelling enzymes cooperate with both DNA-damage-inducible and constitutive histone modifications to promote many facets of the cellular response to DNA damage.


Assuntos
Dano ao DNA/fisiologia , Histonas/metabolismo , Processamento de Proteína Pós-Traducional , Acetilação , Animais , Proteínas de Ciclo Celular/fisiologia , Reparo do DNA/fisiologia , Humanos , Metilação , Modelos Biológicos , Nucleossomos/metabolismo , Fosforilação
11.
Biochim Biophys Acta Gene Regul Mech ; 1866(2): 194922, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36822575

RESUMO

Protein arginine methyltransferases (PRMTs) catalyze the transfer of methyl groups to specific arginine residues of histones and nonhistone proteins. There are nine members in the PRMT family (PRMT1 to PRMT9), and PRMT1 is a dominant member catalyzing majority of arginine methylation in the cell. However, none of the PRMTs is active with recombinant nucleosome as substrate in vitro. Here, we report the discovery of the first in class novel crosstalk between histone H4 lysine 20 (H4K20) monomethylation on nucleosome by SETD8 and histone H4 arginine 3 (H4R3) methylation by PRMT1 in vitro. Full kinetic characterization and mass spectrometry analysis indicated that PRMT1 is only active with recombinant nucleosomes monomethylated at H4K20 by SETD8. These data suggests that the level of activity of PRMT1 could potentially be regulated selectively by SETD8 in various pathways, providing a new approach for discovery of selective regulators of PRMT1 activity.


Assuntos
Histonas , Nucleossomos , Histonas/metabolismo , Metilação , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Arginina
12.
Nature ; 436(7048): 294-8, 2005 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-16015338

RESUMO

DNA breaks are extremely harmful lesions that need to be repaired efficiently throughout the genome. However, the packaging of DNA into nucleosomes is a significant barrier to DNA repair, and the mechanisms of repair in the context of chromatin are poorly understood. Here we show that lysine 56 (K56) acetylation is an abundant modification of newly synthesized histone H3 molecules that are incorporated into chromosomes during S phase. Defects in the acetylation of K56 in histone H3 result in sensitivity to genotoxic agents that cause DNA strand breaks during replication. In the absence of DNA damage, the acetylation of histone H3 K56 largely disappears in G2. In contrast, cells with DNA breaks maintain high levels of acetylation, and the persistence of the modification is dependent on DNA damage checkpoint proteins. We suggest that the acetylation of histone H3 K56 creates a favourable chromatin environment for DNA repair and that a key component of the DNA damage response is to preserve this acetylation.


Assuntos
Ciclo Celular/fisiologia , Dano ao DNA , Histonas/metabolismo , Lisina/metabolismo , Nucleossomos/metabolismo , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/genética , Acetilação , Bleomicina/farmacologia , Camptotecina/farmacologia , DNA/genética , DNA/metabolismo , Dano ao DNA/efeitos dos fármacos , Reparo do DNA , Histonas/química , Nucleossomos/química , Nucleossomos/genética , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo
13.
Mol Cell Proteomics ; 8(3): 506-18, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18936056

RESUMO

Sulfation and phosphorylation are post-translational modifications imparting an isobaric 80-Da addition on the side chain of serine, threonine, or tyrosine residues. These two post-translational modifications are often difficult to distinguish because of their similar MS fragmentation patterns. Targeted MS identification of these modifications in specific proteins commonly relies on their prior separation using gel electrophoresis and silver staining. In the present investigation, we report a potential pitfall in the interpretation of these modifications from silver-stained gels due to artifactual sulfation of serine, threonine, and tyrosine residues by sodium thiosulfate, a commonly used reagent that catalyzes the formation of metallic silver deposits onto proteins. Detailed MS analyses of gel-separated protein standards and Escherichia coli cell extracts indicated that several serine, threonine, and tyrosine residues were sulfated using silver staining protocols but not following Coomassie Blue staining. Sodium thiosulfate was identified as the reagent leading to this unexpected side reaction, and the degree of sulfation was correlated with increasing concentrations of thiosulfate up to 0.02%, which is typically used for silver staining. The significance of this artifact is discussed in the broader context of sulfation and phosphorylation site identification from in vivo and in vitro experiments.


Assuntos
Artefatos , Proteínas/metabolismo , Coloração pela Prata/métodos , Sulfatos/metabolismo , Sequência de Aminoácidos , Aminoácidos/química , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Cricetinae , Escherichia coli/metabolismo , Humanos , Hidroxilação/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/química , Dados de Sequência Molecular , Peptídeos/química , Fosfopiruvato Hidratase/química , Fosforilação/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Tiossulfatos/farmacologia
14.
DNA Repair (Amst) ; 104: 103140, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34087728

RESUMO

A broad spectrum of spontaneous and genotoxin-induced DNA lesions impede replication fork progression. The DNA damage response that acts to promote completion of DNA replication is associated with dynamic changes in chromatin structure that include two distinct processes which operate genome-wide during S-phase. The first, often referred to as histone recycling or parental histone segregation, is characterized by the transfer of parental histones located ahead of replication forks onto nascent DNA. The second, known as de novo chromatin assembly, consists of the deposition of new histone molecules onto nascent DNA. Because these two processes occur at all replication forks, their potential to influence a multitude of DNA repair and DNA damage tolerance mechanisms is considerable. The purpose of this review is to provide a description of parental histone segregation and de novo chromatin assembly, and to illustrate how these processes influence cellular responses to DNA replication roadblocks.


Assuntos
Cromatina/metabolismo , Dano ao DNA , Reparo do DNA , Replicação do DNA , Montagem e Desmontagem da Cromatina , DNA/metabolismo , Eucariotos/genética , Eucariotos/metabolismo , Código das Histonas , Humanos
15.
FEBS J ; 288(11): 3478-3506, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33155404

RESUMO

Candida albicans is a pathogenic fungus that causes systemic infections and mortality in immunosuppressed individuals. We previously showed that deacetylation of histone H3 lysine 56 by Hst3 is essential for C. albicans viability. Hst3 is a fungal-specific NAD+ -dependent protein deacetylase of the sirtuin family. In vivo, supraphysiological concentrations of nicotinamide (NAM) are required for Hst3 inhibition and cytotoxicity. This underscores the importance of identifying mechanisms by which C. albicans can modulate intracellular NAM concentrations. For the first time in a pathogenic fungus, we combine genetics, heavy isotope labeling, and targeted quantitative metabolomics to identify genes, pathways, and mechanisms by which C. albicans can reduce the cytotoxicity of high NAM concentrations. We discovered three distinct fates for supraphysiological NAM concentrations. First, upon transient exposure to NAM, high intracellular NAM concentrations rapidly return near the physiological levels observed in cells that are not exposed to NAM. Second, during the first step of a fungal-specific NAM salvage pathway, NAM is converted into nicotinic acid, a metabolite that cannot inhibit the sirtuin Hst3. Third, we provide evidence that NAM enters the NAD+ metabolome through a NAM exchange reaction that contributes to NAM-mediated inhibition of sirtuins. However, in contrast to the other fates of NAM, the NAM exchange reaction cannot cause a net decrease in the intracellular concentration of NAM. Therefore, this reaction cannot enhance resistance to NAM. In summary, we demonstrate that C. albicans possesses at least two mechanisms to attenuate the cytotoxicity of pharmacological NAM concentrations. It seems likely that those two mechanisms of resistance to cytotoxic NAM concentrations are conserved in many other pathogenic fungi.


Assuntos
Candida albicans/genética , Candidíase/genética , Proteínas Fúngicas/genética , Niacinamida/toxicidade , Candida albicans/patogenicidade , Candidíase/microbiologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Histona Desacetilases/genética , Histonas/genética , Humanos , Lisina/genética , NAD/metabolismo , Niacinamida/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Sirtuínas/genética
16.
Curr Opin Genet Dev ; 16(2): 112-8, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16510276

RESUMO

Proliferating cells need to synthesize large amounts of histones to rapidly package nascent DNA into nucleosomes. This is a challenging task for cells because changes in rates of DNA synthesis lead to an accumulation of excess histones, which interfere with many aspects of DNA metabolism. In addition, cells need to ensure that histone variants are incorporated at the correct chromosomal location. Recent discoveries have highlighted the importance of regulated histone proteolysis in preventing both the accumulation of excess histones and the mis-incorporation of histone variants at inappropriate loci.


Assuntos
Regulação da Expressão Gênica , Histonas/genética , Histonas/metabolismo , Animais , DNA/genética , DNA/metabolismo , Variação Genética , Histonas/biossíntese , Humanos , Hidrólise , Modelos Biológicos
17.
Structure ; 16(7): 1077-85, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18571423

RESUMO

RbAp46 and RbAp48 (pRB-associated proteins p46 and p48, also known as RBBP7 and RBBP4, respectively) are highly homologous histone chaperones that play key roles in establishing and maintaining chromatin structure. We report here the crystal structure of human RbAp46 bound to histone H4. RbAp46 folds into a seven-bladed beta propeller structure and binds histone H4 in a groove formed between an N-terminal alpha helix and an extended loop inserted into blade six. Surprisingly, histone H4 adopts a different conformation when interacting with RbAp46 than it does in either the nucleosome or in the complex with ASF1, another histone chaperone. Our structural and biochemical results suggest that when a histone H3/H4 dimer (or tetramer) binds to RbAp46 or RbAp48, helix 1 of histone H4 unfolds to interact with the histone chaperone. We discuss the implications of our findings for the assembly and function of RbAp46 and RbAp48 complexes.


Assuntos
Proteínas de Transporte/química , Histonas/química , Chaperonas Moleculares/química , Proteínas Nucleares/química , Sequência de Aminoácidos , Sítios de Ligação , Proteínas de Transporte/metabolismo , Histonas/metabolismo , Humanos , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Ligação Proteica , Proteína 7 de Ligação ao Retinoblastoma , Homologia de Sequência de Aminoácidos
18.
Curr Biol ; 16(13): 1280-9, 2006 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-16815704

RESUMO

BACKGROUND: Acetylation of histone H3 lysine 56 (K56Ac) occurs transiently in newly synthesized H3 during passage through S phase and is removed in G2. However, the physiologic roles and effectors of K56Ac turnover are unknown. RESULTS: The sirtuins Hst3p and, to a lesser extent, Hst4p maintain low levels of K56Ac outside of S phase. In hst3 hst4 mutants, K56 hyperacetylation nears 100%. Residues corresponding to the nicotinamide binding pocket of Sir2p are essential for Hst3p function, and H3 K56 deacetylation is inhibited by nicotinamide in vivo. Rapid inactivation of Hst3/Hst4p prior to S phase elevates K56Ac to 50% in G2, suggesting that K56-acetylated nucleosomes are assembled genome-wide during replication. Inducible expression of Hst3p in G1 or G2 triggers deacetylation of mature chromatin. Cells lacking Hst3/Hst4p exhibit many phenotypes: spontaneous DNA damage, chromosome loss, thermosensitivity, and acute sensitivity to genotoxic agents. These phenotypes are suppressed by mutation of histone H3 K56 into a nonacetylatable residue or by loss of K56Ac in cells lacking the histone chaperone Asf1. CONCLUSIONS: Our results underscore the critical importance of Hst3/Hst4p in controlling histone H3 K56Ac and thereby maintaining chromosome integrity.


Assuntos
Histona Desacetilases/fisiologia , Histonas/metabolismo , Lisina/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/enzimologia , Acetilação , Sequência de Aminoácidos , Sítios de Ligação , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/fisiologia , Cromatina/metabolismo , Dano ao DNA , Replicação do DNA , Genoma Fúngico , Instabilidade Genômica , Histona Desacetilases/química , Histona Desacetilases/genética , Chaperonas Moleculares , Dados de Sequência Molecular , Mutação , Niacinamida/metabolismo , Processamento de Proteína Pós-Traducional , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Alinhamento de Sequência , Sirtuínas/química , Sirtuínas/genética , Sirtuínas/fisiologia
19.
Anal Chem ; 81(15): 6300-9, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20337398

RESUMO

A novel method to improve the detection of protein ions using a linear ion trap mass spectrometer is presented. A scan function combining charge separation with segmented transmission of multiply charged ions was developed to enhance the sensitivity and resolution of the linear ion trap for the nanoLC-MS analysis of intact proteins. The analytical benefits of the present method are particularly apparent in protein analyses, where the increased proportion of multiply charged ions can exacerbate space-charge effects and compromise the dynamic range of the linear ion trap instrument. The enhanced ion storage and charge separation capabilities of our targeted and enhanced multiply charged scan mode provided a 4-fold increase in signal-to-noise and 5-fold increase in resolution, thus enabling the detection of closely related protein isoforms. The application of this method is demonstrated for low femtomole detection of protein standards and nuclear extracts enriched in histone proteins. The enhanced resolution of this scan mode also enabled us to monitor subtle changes in the methylation of a subpopulation of histone H3 that occurs in chicken DT40 cells lacking specific methyltransferase activity. The extent of the fold change and PTM site localization was performed using predictive software tools and targeted multiple reaction monitoring analysis of histone peptides. Monomethylation of Lys 79 in histone H3 (H3K79me1) was down regulated by 240-fold in methyltransferase deficient cells.


Assuntos
Histona-Lisina N-Metiltransferase/análise , Histonas/análise , Lisina/análise , Espectrometria de Massas , Animais , Células Cultivadas , Galinhas , Cromatografia Líquida de Alta Pressão , Histona Metiltransferases , Metilação , Nanotecnologia , Processamento de Proteína Pós-Traducional
20.
Genetics ; 179(4): 1769-84, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18579506

RESUMO

Deacetylation of histone H3 K56, regulated by the sirtuins Hst3p and Hst4p, is critical for maintenance of genomic stability. However, the physiological consequences of a lack of H3 K56 deacetylation are poorly understood. Here we show that cells lacking Hst3p and Hst4p, in which H3 K56 is constitutively hyperacetylated, exhibit hallmarks of spontaneous DNA damage, such as activation of the checkpoint kinase Rad53p and upregulation of DNA-damage inducible genes. Consistently, hst3 hst4 cells display synthetic lethality interactions with mutations that cripple genes involved in DNA replication and DNA double-strand break (DSB) repair. In most cases, synthetic lethality depends upon hyperacetylation of H3 K56 because it can be suppressed by mutation of K56 to arginine, which mimics the nonacetylated state. We also show that hst3 hst4 phenotypes can be suppressed by overexpression of the PCNA clamp loader large subunit, Rfc1p, and by inactivation of the alternative clamp loaders CTF18, RAD24, and ELG1. Loss of CTF4, encoding a replisome component involved in sister chromatid cohesion, also suppresses hst3 hst4 phenotypes. Genetic analysis suggests that CTF4 is a part of the K56 acetylation pathway that converges on and modulates replisome function. This pathway represents an important mechanism for maintenance of genomic stability and depends upon proper regulation of H3 K56 acetylation by Hst3p and Hst4p. Our data also suggest the existence of a precarious balance between Rfc1p and the other RFC complexes and that the nonreplicative forms of RFC are strongly deleterious to cells that have genomewide and constitutive H3 K56 hyperacetylation.


Assuntos
Dano ao DNA/fisiologia , Histona Desacetilases/metabolismo , Histonas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Sirtuínas/metabolismo , Acetilação , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Dano ao DNA/genética , Reparo do DNA , Replicação do DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Histona Acetiltransferases/metabolismo , Histona Desacetilases/genética , Peptídeos e Proteínas de Sinalização Intracelular , Complexos Multienzimáticos/metabolismo , Mutação , Fenótipo , Proteínas Serina-Treonina Quinases , Proteína de Replicação C/genética , Proteína de Replicação C/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Sirtuínas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA