Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Life (Basel) ; 12(1)2022 Jan 04.
Article in English | MEDLINE | ID: mdl-35054463

ABSTRACT

Severe acute respiratory syndrome coronavirus (SARS-CoV-2) is highly contagious and causes coronavirus disease 2019 (COVID-19). Reverse transcription quantitative polymerase chain reaction (RT-qPCR) is the most accurate and reliable molecular assay to detect active SARS-CoV-2 infection. However, a rapid increase in test subjects has created a global bottleneck in testing capacity. Given that efficient nucleic acid extraction greatly affects reliable and accurate testing results, we compared three extraction platforms: MagNA Pure 96 DNA and Viral NA Small Volume kit on MagNA Pure 96 (Roche, Basel, Switzerland), careGENETM Viral/Pathogen HiFi Nucleic Acid Isolation kit (WELLS BIO Inc., Seoul, Korea) on KingFisher Flex (Thermo Fisher Scientific, Rocklin, CA, USA), and SGRespiTM Pure kit (Seegene Inc., Seoul, Korea) on Maelstrom 9600 (Taiwan Advanced Nanotech Inc., Taoyuan, Taiwan). RNA was extracted from 245 residual respiratory specimens from the different types of samples (i.e., NPS, sputum, and saliva) using three different kits. The 95% limits of detection of median tissue culture infectious dose per milliliter (TCID50/mL) for the MagNA Pure 96, KingFisher Flex, and Maelstrom 9600 were 0.37-3.15 × 101, 0.41-3.62 × 101, and 0.33-1.98 × 101, respectively. The KingFisher Flex platform exhibited 99.2% sensitivity and 100% specificity, whereas Maelstrom 9600 exhibited 98.3-100% sensitivity and 100% specificity. Bland-Altman analysis revealed a 95.2% concordance between MagNA Pure 96 and KingFisher Flex and 95.4% concordance between MagNA Pure 96 and Maelstrom 9600, indicating that all three platforms provided statistically reliable results. This suggests that two modifying platforms, KingFisher Flex and Maelstrom 9600, are accurate and scalable extraction platforms for large-scale SARS-CoV-2 clinical detection and could help the management of COVID-19 patients.

2.
Genes (Basel) ; 10(4)2019 04 08.
Article in English | MEDLINE | ID: mdl-30965655

ABSTRACT

Microhomology-mediated end joining (MMEJ) anneals short, imperfect microhomologies flanking DNA breaks, producing repair products with deletions in a Ku- and RAD52-independent fashion. Puzzlingly, MMEJ preferentially selects certain microhomologies over others, even when multiple microhomologies are available. To define rules and parameters for microhomology selection, we altered the length, the position, and the level of mismatches to the microhomologies flanking homothallic switching (HO) endonuclease-induced breaks and assessed their effect on MMEJ frequency and the types of repair product formation. We found that microhomology of eight to 20 base pairs carrying no more than 20% mismatches efficiently induced MMEJ. Deletion of MSH6 did not impact MMEJ frequency. MMEJ preferentially chose a microhomology pair that was more proximal from the break. Interestingly, MMEJ events preferentially retained the centromere proximal side of the HO break, while the sequences proximal to the telomere were frequently deleted. The asymmetry in the deletional profile among MMEJ products was reduced when HO was induced on the circular chromosome. The results provide insight into how cells search and select microhomologies for MMEJ in budding yeast.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair/genetics , DNA Repair/genetics , Saccharomyces cerevisiae/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Saccharomyces cerevisiae Proteins/genetics , Sequence Deletion/genetics
3.
Nat Struct Mol Biol ; 26(3): 155-163, 2019 03.
Article in English | MEDLINE | ID: mdl-30778235

ABSTRACT

Ribonucleoside monophosphates (rNMPs) mis-incorporated during DNA replication are removed by RNase H2-dependent excision repair or by topoisomerase I (Top1)-catalyzed cleavage. The cleavage of rNMPs by Top1 produces 3' ends harboring terminal adducts, such as 2',3'-cyclic phosphate or Top1 cleavage complex (Top1cc), and leads to frequent mutagenesis and DNA damage checkpoint induction. We surveyed a range of candidate enzymes from Saccharomyces cerevisiae for potential roles in Top1-dependent genomic rNMP removal. Genetic and biochemical analyses reveal that Apn2 resolves phosphotyrosine-DNA conjugates, terminal 2',3'-cyclic phosphates, and their hydrolyzed products. APN2 also suppresses 2-base pair (bp) slippage mutagenesis in RNH201-deficient cells. Our results define additional activities of Apn2 in resolving a wide range of 3' end blocks and identify a role for Apn2 in maintaining genome integrity during rNMP repair.


Subject(s)
Base Pairing/genetics , DNA Repair/genetics , DNA Replication/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Ribonucleotides/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , 3' Untranslated Regions/genetics , DNA Topoisomerases, Type I/metabolism , DNA, Fungal/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Genome, Fungal/genetics , Mutagenesis/genetics , Ribonucleases/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
4.
Elife ; 72018 08 17.
Article in English | MEDLINE | ID: mdl-30117416

ABSTRACT

Translational efficiency correlates with longevity, yet its role in lifespan determination remains unclear. Using ribosome profiling, translation efficiency is globally reduced during replicative aging in budding yeast by at least two mechanisms: Firstly, Ssd1 is induced during aging, sequestering mRNAs to P-bodies. Furthermore, Ssd1 overexpression in young cells reduced translation and extended lifespan, while loss of Ssd1 reduced the translational deficit of old cells and shortened lifespan. Secondly, phosphorylation of eIF2α, mediated by the stress kinase Gcn2, was elevated in old cells, contributing to the global reduction in translation without detectable induction of the downstream Gcn4 transcriptional activator. tRNA overexpression activated Gcn2 in young cells and extended lifespan in a manner dependent on Gcn4. Moreover, overexpression of Gcn4 sufficed to extend lifespan in an autophagy-dependent manner in the absence of changes in global translation, indicating that Gcn4-mediated autophagy induction is the ultimate downstream target of activated Gcn2, to extend lifespan.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Longevity/genetics , Protein Biosynthesis , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae Proteins/genetics , Eukaryotic Initiation Factor-2/genetics , Gene Expression Regulation, Fungal , Phosphorylation , Ribosomes/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development
5.
Nat Commun ; 9(1): 2025, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29795289

ABSTRACT

Yeast Rad1-Rad10 (XPF-ERCC1 in mammals) incises UV, oxidation, and cross-linking agent-induced DNA lesions, and contributes to multiple DNA repair pathways. To determine how Rad1-Rad10 catalyzes inter-strand crosslink repair (ICLR), we examined sensitivity to ICLs from yeast deleted for SAW1 and SLX4, which encode proteins that interact physically with Rad1-Rad10 and bind stalled replication forks. Saw1, Slx1, and Slx4 are critical for replication-coupled ICLR in mus81 deficient cells. Two rad1 mutations that disrupt interactions between Rpa1 and Rad1-Rad10 selectively disable non-nucleotide excision repair (NER) function, but retain UV lesion repair. Mutations in the analogous region of XPF also compromised XPF interactions with Rpa1 and Slx4, and are proficient in NER but deficient in ICLR and direct repeat recombination. We propose that Rad1-Rad10 makes distinct contributions to ICLR depending on cell cycle phase: in G1, Rad1-Rad10 removes ICL via NER, whereas in S/G2, Rad1-Rad10 facilitates NER-independent replication-coupled ICLR.


Subject(s)
DNA Damage/genetics , DNA Repair Enzymes/metabolism , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Single-Strand Specific DNA and RNA Endonucleases/metabolism , Animals , CHO Cells , Cell Cycle/genetics , Cricetulus , Cross-Linking Reagents/toxicity , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Intravital Microscopy , Mutagenesis, Site-Directed , Mutation , Saccharomyces cerevisiae Proteins/genetics , Single-Strand Specific DNA and RNA Endonucleases/genetics , Ultraviolet Rays/adverse effects
6.
Nucleic Acids Res ; 46(10): 5075-5096, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29660012

ABSTRACT

Double strand DNA break repair (DSBR) comprises multiple pathways. A subset of DSBR pathways, including single strand annealing, involve intermediates with 3' non-homologous tails that must be removed to complete repair. In Saccharomyces cerevisiae, Rad1-Rad10 is the structure-specific endonuclease that cleaves the tails in 3' non-homologous tail removal (3' NHTR). Rad1-Rad10 is also an essential component of the nucleotide excision repair (NER) pathway. In both cases, Rad1-Rad10 requires protein partners for recruitment to the relevant DNA intermediate. Msh2-Msh3 and Saw1 recruit Rad1-Rad10 in 3' NHTR; Rad14 recruits Rad1-Rad10 in NER. We created two rad1 separation-of-function alleles, rad1R203A,K205A and rad1R218A; both are defective in 3' NHTR but functional in NER. In vitro, rad1R203A,K205A was impaired at multiple steps in 3' NHTR. The rad1R218A in vivo phenotype resembles that of msh2- or msh3-deleted cells; recruitment of rad1R218A-Rad10 to recombination intermediates is defective. Interactions among rad1R218A-Rad10 and Msh2-Msh3 and Saw1 are altered and rad1R218A-Rad10 interactions with RPA are compromised. We propose a model in which Rad1-Rad10 is recruited and positioned at the recombination intermediate through interactions, between Saw1 and DNA, Rad1-Rad10 and Msh2-Msh3, Saw1 and Msh2-Msh3 and Rad1-Rad10 and RPA. When any of these interactions is altered, 3' NHTR is impaired.


Subject(s)
DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Replication Protein A/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Single-Strand Specific DNA and RNA Endonucleases/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , MutS Homolog 2 Protein/metabolism , MutS Homolog 3 Protein/genetics , MutS Homolog 3 Protein/metabolism , Mutation , Protein Interaction Mapping , Replication Protein A/genetics , Saccharomyces cerevisiae/radiation effects , Saccharomyces cerevisiae Proteins/genetics , Single-Strand Specific DNA and RNA Endonucleases/genetics , Ultraviolet Rays
7.
Mutat Res ; 809: 81-87, 2018 05.
Article in English | MEDLINE | ID: mdl-28754468

ABSTRACT

DNA double-strand breaks (DSBs) are induced by a variety of genotoxic agents, including ionizing radiation and chemotherapy drugs for treating cancers. The elimination of DSBs proceeds via distinctive error-free and error-prone pathways. Repair by homologous recombination (HR) is largely error-free and mediated by RAD51/BRCA2 gene products. Classical non-homologous end joining (C-NHEJ) requires the Ku heterodimer and can efficiently rejoin breaks, with occasional loss or gain of DNA information. Recently, evidence has unveiled another DNA end-joining mechanism that is independent of recombination factors and Ku proteins, termed alternative non-homologous end joining (A-NHEJ). While A-NHEJ-mediated repair does not require homology, in a subtype of A-NHEJ, DSB breaks are sealed by microhomology (MH)-mediated base-pairing of DNA single strands, followed by nucleolytic trimming of DNA flaps, DNA gap filling, and DNA ligation, yielding products that are always associated with DNA deletion. This highly error-prone DSB repair pathway is termed microhomology-mediated end joining (MMEJ). Dissecting the mechanisms of MMEJ is of great interest because of its potential to destabilize the genome through gene deletions and chromosomal rearrangements in cells deficient in canonical repair pathways, including HR and C-NHEJ. In addition, evidence now suggests that MMEJ plays a physiological role in normal cells.


Subject(s)
BRCA2 Protein/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Ku Autoantigen/metabolism , Rad51 Recombinase/metabolism , Recombinational DNA Repair , Animals , BRCA2 Protein/genetics , Chromosome Aberrations , Gene Deletion , Humans , Ku Autoantigen/genetics , Rad51 Recombinase/genetics
8.
BMB Rep ; 48(12): 685-90, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26058396

ABSTRACT

The eukaryotic genome is packed into chromatin, which is important for the genomic integrity and gene regulation. Chromatin structures are maintained through assembly and disassembly of nucleosomes catalyzed by histone chaperones. Asf1 (anti-silencing function 1) is a highly conserved histone chaperone that mediates histone transfer on/off DNA and promotes histone H3 lysine 56 acetylation at globular core domain of histone H3. To elucidate the role of Asf1 in the modulation of chromatin structure, we screened and identified small molecules that inhibit Asf1 and H3K56 acetylation without affecting other histone modification. These pyrimidine-2,4,6-trione derivative molecules inhibited the nucleosome assembly mediated by Asf1 in vitro, and reduced the H3K56 acetylation in HeLa cells. Furthermore, production of HSV viral particles was reduced by these compounds. As Asf1 is implicated in genome integrity, cell proliferation, and cancer, current Asf1 inhibitor molecules may offer an opportunity for the therapeutic development for treatment of diseases.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Chromatin/drug effects , Small Molecule Libraries/pharmacology , Acetylation , Chromatin/metabolism , Chromatin Assembly and Disassembly/drug effects , Histones/metabolism , Humans , Molecular Chaperones , Nucleosomes/drug effects , Nucleosomes/metabolism
9.
Genes Dev ; 28(4): 396-408, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24532716

ABSTRACT

All eukaryotic cells divide a finite number of times, although the mechanistic basis of this replicative aging remains unclear. Replicative aging is accompanied by a reduction in histone protein levels, and this is a cause of aging in budding yeast. Here we show that nucleosome occupancy decreased by 50% across the whole genome during replicative aging using spike-in controlled micrococcal nuclease digestion followed by sequencing. Furthermore, nucleosomes became less well positioned or moved to sequences predicted to better accommodate histone octamers. The loss of histones during aging led to transcriptional induction of all yeast genes. Genes that are normally repressed by promoter nucleosomes were most induced, accompanied by preferential nucleosome loss from their promoters. We also found elevated levels of DNA strand breaks, mitochondrial DNA transfer to the nuclear genome, large-scale chromosomal alterations, translocations, and retrotransposition during aging.


Subject(s)
Aging/genetics , Genome, Fungal/genetics , Genomic Instability/genetics , Nucleosomes/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Up-Regulation , Chromosome Aberrations , DNA Breaks , DNA, Mitochondrial/genetics , Gene Expression Regulation, Fungal , Histones/metabolism , Promoter Regions, Genetic/genetics , TATA Box/genetics
10.
Front Genet ; 4: 136, 2013.
Article in English | MEDLINE | ID: mdl-23967007

ABSTRACT

It is well accepted that non-coding RNAs play a critical role in regulating gene expression. Recent paradigm-setting studies are now revealing that non-coding RNAs, other than microRNAs, also play intriguing roles in the maintenance of chromatin structure, in the DNA damage response, and in adult human stem cell aging. In this review, we will discuss the complex inter-dependent relationships among non-coding RNA transcription, maintenance of genomic stability, chromatin structure, and adult stem cell senescence. DNA damage-induced non-coding RNAs transcribed in the vicinity of the DNA break regulate recruitment of the DNA damage machinery and DNA repair efficiency. We will discuss the correlation between non-coding RNAs and DNA damage repair efficiency and the potential role of changing chromatin structures around double-strand break sites. On the other hand, induction of non-coding RNA transcription from the repetitive Alu elements occurs during human stem cell aging and hinders efficient DNA repair causing entry into senescence. We will discuss how this fine balance between transcription and genomic instability may be regulated by the dramatic changes to chromatin structure that accompany cellular senescence.

11.
Nucleic Acids Res ; 41(10): 5199-209, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23563152

ABSTRACT

The mammalian genome encodes multiple variants of histone H3 including H3.1/H3.2 and H3.3. In contrast to H3.1/H3.2, H3.3 is enriched in the actively transcribed euchromatin and the telomeric heterochromatins. However, the mechanism for H3.3 to incorporate into the different domains of chromatin is not known. Here, taking the advantage of well-defined transcription analysis system of yeast, we attempted to understand the molecular mechanism of selective deposition of human H3.3 into actively transcribed genes. We show that there are systemic H3 substrate-selection mechanisms operating even in yeasts, which encode a single type of H3. Yeast HIR complex mediated H3-specific recognition specificity for deposition of H3.3 in the transcribed genes. A critical component of this process was the H3 A-IG code composed of amino acids 87, 89 and 90. The preference toward H3.3 was completely lost when HIR subunits were absent and partially suppressed by human HIRA. Asf1 allows the influx of H3, regardless of H3 type. We propose that H3.3 is introduced into the active euchromatin by targeting the recycling pathway that is mediated by HIRA (or HIR), and this H3-selection mechanism is highly conserved through the evolution. These results also uncover an unexpected role of RI chaperones in evolution of variant H3s.


Subject(s)
Biological Evolution , Histone Chaperones/physiology , Histones/metabolism , Transcription, Genetic , Amino Acid Sequence , Cell Cycle Proteins/genetics , Conserved Sequence , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Fungal Proteins/physiology , Histone Chaperones/genetics , Histone Chaperones/metabolism , Histones/chemistry , Humans , Mutation , Protein Structure, Tertiary , Transcription Factors/genetics , Yeasts/genetics , Yeasts/metabolism
12.
Mol Cells ; 33(2): 195-201, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22286234

ABSTRACT

The biological functions of Myc are to regulate cell growth,apoptosis, cell differentiation and stem-cell self-renewal. Abnormal accumulation of c-Myc is able to induce excessive proliferation of normal cells. von Hippel-Lindau protein(pVHL) is a key regulator of hypoxia-inducible factor 1α(HIF1α), thus accumulation and hyperactivation of HIF1α is the most prominent feature of VHL-mutated renal cell carcinoma. Interestingly, the Myc pathway is reported to be activated in renal cell carcinoma even though the precise molecular mechanism still remains to be established. Here, we demonstrated that pVHL locates at the c-Myc promoter region through physical interaction with Myc. Furthermore, pVHL reinforces HDAC1/2 recruitment to the Myc promoter, which leads to the auto-suppression of Myc. Therefore, one possible mechanism of Myc auto-suppression by pVHL entails removing histone acetylation. Our study identifies a novel mechanism for pVHL-mediated negative regulation of c-Myc transcription.


Subject(s)
Carcinoma, Renal Cell/genetics , Gene Expression Regulation, Neoplastic , Histone Deacetylases/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Acetylation , Carcinoma, Renal Cell/enzymology , Cell Proliferation , HEK293 Cells , Homeostasis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney Neoplasms/enzymology , Promoter Regions, Genetic/genetics , Protein Binding , Response Elements/genetics , Sequence Deletion/genetics , Transcriptional Activation/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics
13.
Proc Natl Acad Sci U S A ; 108(1): 85-90, 2011 Jan 04.
Article in English | MEDLINE | ID: mdl-21173268

ABSTRACT

In mammals, the canonical histone H3 and the variant H3.3 are assembled into chromatin through replication-coupled and replication-independent (RI) histone deposition pathways, respectively, to play distinct roles in chromatin function. H3.3 is largely associated with transcriptionally active regions via the activity of RI histone chaperone, HIRA. However, the precise role of the RI pathway and HIRA in active transcription and the mechanisms by which H3.3 affects gene activity are not known. In this study, we show that HIRA is an essential factor for muscle development by establishing MyoD activation in myotubes. HIRA and Asf1a, but not CHD1 or Asf1b, mediate H3.3 incorporation in the promoter and the critical upstream regulatory regions of the MyoD gene. HIRA and H3.3 are required for epigenetic transition into the more permissive chromatin structure for polymerase II recruitment to the promoter, regardless of transcription-associated covalent modification of histones. Our results suggest distinct epigenetic management of the master regulator with RI pathway components for cellular differentiation.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly/physiology , Histone Chaperones/metabolism , Histones/metabolism , Muscle Development/physiology , MyoD Protein/metabolism , Transcription Factors/metabolism , Transcriptional Activation/physiology , Animals , Cell Line , Chromatin Immunoprecipitation , DNA Primers/genetics , Fluorescent Antibody Technique , Immunoblotting , Immunoprecipitation , Mice , Microarray Analysis , RNA Interference , RNA, Small Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Activation/genetics , Transfection
14.
BMB Rep ; 42(4): 227-31, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19403047

ABSTRACT

The eukaryotic genome forms a chromatin structure that contains repeating nucleosome structures. Nucleosome packaging is regulated by chromatin remodeling factors such as histone chaperones. The Saccharomyces cerevisiae H3/H4 histone chaperones, CAF-1 and Asf1, regulate DNA replication and chromatin assembly. CAF-1 function is largely restricted to non-transcriptional processes in heterochromatin, whereas Asf1 regulates transcription together with another H3/H4 chaperone, HIR. This study examined the role of the yeast H3/H4 histone chaperones, Asf1, HIR, and CAF-1 in chromatin dynamics during transcription. Unexpectedly, CAF-1 was recruited to the actively transcribed region in a similar way to HIR and Asf1. In addition, the three histone chaperones genetically interacted with Set2-dependent H3 K36 methylation. Similar to histone chaperones, Set2 was required for tolerance to excess histone H3 but not to excess H2A, suggesting that CAF-1, Asf1, HIR, and Set2 function in a related pathway and target chromatin during transcription.


Subject(s)
Molecular Chaperones/physiology , Ribonucleases/physiology , Saccharomyces cerevisiae Proteins/physiology , Transcription, Genetic/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Histones/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Methyltransferases/physiology , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Kinases/physiology , Ribonucleases/genetics , Ribonucleases/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , TATA-Box Binding Protein/physiology
15.
Biochem Biophys Res Commun ; 374(3): 543-8, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18657516

ABSTRACT

To understand the role of histone H3 sub-domains in chromatin function, 35 histone H3 tandem alanine mutants were generated and tested for their viability and sensitivity to DNA damaging agents. Among 13 non-viable H3 mutants, 6 were mapped around the alphaN helix and preceding tail region. Mutants with individual alanine substitutions in this region were viable but developed multiple sensitivities to DNA damaging agents. The only viable triple mutant, REI49-50A, in the alphaN helix region could not grow when combined with histone chaperone mutations, such as asf1Delta, cac1Delta, or hir1Delta, suggesting that this particular region is important when the histone assembly/disassembly pathway is compromised. In addition, further analysis showed that T45, E50, or F54 of the alphaN helix genetically interacted with a histone chaperone (Asf1) and transcription elongation factors (Paf1 and Hpr1). These results suggest a specific role of the H3 alphaN helix in histone dynamics mediated by histone chaperones, which might be important during transcription elongation.


Subject(s)
Histones/metabolism , Nucleosomes/metabolism , Saccharomyces cerevisiae/metabolism , Alanine/genetics , Alanine/metabolism , Alkylating Agents/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Cell Cycle Proteins/metabolism , DNA Mutational Analysis , Histones/chemistry , Histones/genetics , Hydroxyurea/pharmacology , Methyl Methanesulfonate/pharmacology , Molecular Chaperones/metabolism , Molecular Sequence Data , Mutation , Nuclear Proteins/metabolism , Nucleosomes/drug effects , Protein Structure, Secondary , Ribonucleotide Reductases/antagonists & inhibitors , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Tandem Repeat Sequences
16.
EMBO J ; 26(21): 4467-74, 2007 Oct 31.
Article in English | MEDLINE | ID: mdl-17914459

ABSTRACT

Transcription by RNA polymerase II is accompanied by dynamic changes in chromatin, including the eviction/deposition of nucleosomes or the covalent modification of histone subunits. This study examined the role of the histone H3/H4 chaperones, Asf1 and HIR, in histone mobility during transcription, with particular focus on the histone exchange pathway, using a dual histone expression system. The results showed that the exchange of H3/H4 normally occurs during transcription by the histone chaperones. Both Asf1 and HIR are important for histone deposition but have a different effect on histone exchange. While Asf1 mediated incorporation of external H3/H4 and renewal of pre-existing histones, HIR opposed it. The balance of two opposing activities might be an important mechanism for determining current chromatin states.


Subject(s)
Histones/metabolism , Molecular Chaperones/metabolism , Cell Cycle Proteins/chemistry , Fungal Proteins/metabolism , Gene Deletion , Gene Silencing , Histones/chemistry , Mutation , Nuclear Proteins/metabolism , Nucleosomes/metabolism , RNA Polymerase II/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic
17.
Biochem Biophys Res Commun ; 349(2): 463-70, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16959218

ABSTRACT

Histone H3 methyltransferases are involved in the epigenetic control of transcription and heterochromatin maintenance. In Saccharomyces cerevisiae, deletion of a histone H3 methyltransferase SET1 leads to the induction of a subset of stress responsive genes in a Rad53 dependent manner. This type of induction was observed only in the absence of SET1 and not in the absence of other histone methyltransferases, SET2 or DOT1. We show that the increased expression of the stress responsive genes results from a lack of histone H3 lysine (K) 4 methylation. The loss of mono-methylation on H3 K4 is necessary to increase the expression of the stress responsive genes, while the loss of di- or tri-methylation induced by deletion of either RRM domain of Set1 or the upstream effector molecules hardly affected their expression. These results suggest that mono- and multiple methylation of H3 K4 have different roles. The mono-methylation of H3 K4 might be required for the global integrity of chromatin structure, which is normally monitored by the Rad53 dependent chromatin surveillance system.


Subject(s)
Chromatin/chemistry , Histones/chemistry , Lysine/chemistry , Saccharomyces cerevisiae/metabolism , Cell Cycle Proteins/metabolism , Checkpoint Kinase 2 , DNA-Binding Proteins/physiology , Epigenesis, Genetic , Fungal Proteins/chemistry , Heterochromatin/chemistry , Histone-Lysine N-Methyltransferase , Methyltransferases/physiology , Mutagenesis , Nuclear Proteins/physiology , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Transcription Factors/physiology
18.
J Microbiol ; 43(6): 516-22, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16410768

ABSTRACT

The phosphorylation of C-terminal domain (CTD) of Rpb1p, the largest subunit of RNA polymerase II plays an important role in transcription and the coupling of various cellular events to transcription. In this study, its role in DNA damage response is closely examined in Saccharomyces cerevisiae, focusing specifically on several transcription factors that mediate or respond to the phosphorylation of the CTD. CTDK-1, the pol II CTD kinase, FCP1, the CTD phosphatase, ESS1, the CTD phosphorylation dependent cis-trans isomerase, and RSP5, the phosphorylation dependent pol II ubiquitinating enzyme, were chosen for the study. We determined that the CTD phosphorylation of CTD, which occurred predominantly at serine 2 within a heptapeptide repeat, was enhanced in response to a variety of sources of DNA damage. This modification was shown to be mediated by CTDK-1. Although mutations in ESS1 or FCP1 caused cells to become quite sensitive to DNA damage, the characteristic pattern of CTD phosphorylation remained unaltered, thereby implying that ESS1 and FCP1 play roles downstream of CTD phosphorylation in response to DNA damage. Our data suggest that the location or extent of CTD phosphorylation might be altered in response to DNA damage, and that the modified CTD, ESS1, and FCP1 all contribute to cellular survival in such conditions.


Subject(s)
DNA Repair , Gene Expression Regulation, Fungal , RNA Polymerase II/metabolism , RNA Polymerase II/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , DNA Damage , Mutation , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/physiology , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/physiology , Phosphorylation , Protein Kinases/physiology , Protein Structure, Tertiary , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/physiology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...