Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 560(7718): 331-335, 2018 08.
Article in English | MEDLINE | ID: mdl-30069045

ABSTRACT

Eukaryotic genomes are generally organized in multiple chromosomes. Here we have created a functional single-chromosome yeast from a Saccharomyces cerevisiae haploid cell containing sixteen linear chromosomes, by successive end-to-end chromosome fusions and centromere deletions. The fusion of sixteen native linear chromosomes into a single chromosome results in marked changes to the global three-dimensional structure of the chromosome due to the loss of all centromere-associated inter-chromosomal interactions, most telomere-associated inter-chromosomal interactions and 67.4% of intra-chromosomal interactions. However, the single-chromosome and wild-type yeast cells have nearly identical transcriptome and similar phenome profiles. The giant single chromosome can support cell life, although this strain shows reduced growth across environments, competitiveness, gamete production and viability. This synthetic biology study demonstrates an approach to exploration of eukaryote evolution with respect to chromosome structure and function.


Subject(s)
Chromosomes, Artificial, Yeast/genetics , Genetic Engineering/methods , Genetic Fitness/genetics , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Synthetic Biology/methods , Artificial Gene Fusion/methods , Centromere/genetics , Evolution, Molecular , Meiosis , Microbial Viability/genetics , Saccharomyces cerevisiae/growth & development , Spores, Fungal/genetics , Telomere/genetics , Transcriptome
2.
Biochem J ; 479(23): 2433-2447, 2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36416748

ABSTRACT

The KEOPS complex is an evolutionarily conserved protein complex in all three domains of life (Bacteria, Archaea, and Eukarya). In budding yeast Saccharomyces cerevisiae, the KEOPS complex (ScKEOPS) consists of five subunits, which are Kae1, Bud32, Cgi121, Pcc1, and Gon7. The KEOPS complex is an ATPase and is required for tRNA N6-threonylcarbamoyladenosine modification, telomere length maintenance, and efficient DNA repair. Here, recombinant ScKEOPS full complex and Kae1-Pcc1-Gon7 and Bud32-Cgi121 subcomplexes were purified and their biochemical activities were examined. KEOPS was observed to have ATPase and GTPase activities, which are predominantly attributed to the Bud32 subunit, as catalytically dead Bud32, but not catalytically dead Kae1, largely eliminated the ATPase/GTPase activity of KEOPS. In addition, KEOPS could hydrolyze ADP to adenosine or GDP to guanosine, and produce PPi, indicating that KEOPS is an ADP/GDP nucleotidase. Further mutagenesis characterization of Bud32 and Kae1 subunits revealed that Kae1, but not Bud32, is responsible for the ADP/GDP nucleotidase activity. In addition, the Kae1V309D mutant exhibited decreased ADP/GDP nucleotidase activity in vitro and shortened telomeres in vivo, but showed only a limited defect in t6A modification, suggesting that the ADP/GDP nucleotidase activity of KEOPS contributes to telomere length regulation.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Diphosphate/metabolism , GTP Phosphohydrolases/metabolism
3.
Int J Qual Health Care ; 35(3)2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37417461

ABSTRACT

Since the public long-term care insurance (LTCI) system was piloted in Chengdu, China, in October 2017, there has been considerable growth of LTC institutions in China. This study aimed to evaluate the health value effect of LTCI in older patients with severe disabilities in an LTC institution. This prospective study was based on data from 985 severe disability patients with or without LTCI from October 2017 to May 2021 in the Eighth People's Hospital, Chengdu, China. The Cox proportional hazard model estimated LTCI's health value, including survival probability and risk of pneumonia/pressure ulcers. Subgroup analysis was performed for sex, age, Charlson Comorbidity Index (CCI), and the number of drugs. In the analysis, 519 and 466 patients in LTCI and non-LTCI groups were included, respectively. In adjusted Cox analyses, the LTCI group had a significantly elevated survival rate compared with the non-LTCI groups at 12 months (P < .001, hazard ratio (HR) = 1.758, 95% confidence interval (CI) 1.300-2.376). At 40 months, the adjusted survival rate was 62.6% in the LTCI group, which was significantly higher (53.7%; P = .003, HR = 1.438, 95% CI 1.131-1.831). The subgroups of patients aged 60 to 79 years (interaction P = .007) and with CCI ≥ 3 (interaction P = .026) were more significantly associated with survival improvement than those aged >80 years and with CCI< 3. The LTCI group was also at lower risk for hospital-acquired pneumonia (P = .016, HR 0.622, 95% CI 0.422-0.917) and pressure ulcers (P = .008, HR 0.695, 95% CI 0.376-0.862). The improved survival of LTCI remained stable in sensitivity analyses. For older patients with severe disabilities, in a LTC institution, LTCI significantly improved their health profile and longevity after a year, suggesting the large role and development potentiality of institution care in the LTCI system of China.


Subject(s)
Insurance, Long-Term Care , Pressure Ulcer , Humans , Aged , Prospective Studies , China , Outcome Assessment, Health Care , Long-Term Care
4.
J Med Virol ; 94(6): 2317-2330, 2022 06.
Article in English | MEDLINE | ID: mdl-35174515

ABSTRACT

Pooled data from 2352 hospitalized coronavirus disease 2019 (COVID-19) patients with viral RNA in feces across 46 studies were analyzed and the pooled prevalence of fecal RNA was 46.8% (95% confidence interval [CI]: 0.383-0.554). The pooled analysis showed that the occurrence of total gastrointestinal (GI) symptoms was 28.5% (95% CI: 0.125-0.44) in COVID-19 patients with fecal RNA, that of both respiratory and GI symptoms was 21.9% (95% CI: 0.09-0.346), that of only GI symptoms was 19.8% (95% CI: 0.107-0.288), and that of only respiratory symptoms was 50.5%(95% CI: 0.267-0.744). The pooled data showed no significant difference in positive fecal RNA between severe and nonsevere cases (odds ratio = 2.009, p = 0.079, 95% CI: 0.922-4.378). During hospital admission, after samples from the respiratory system tested negative for viral RNA, 55.4% (95% CI: 0.418-0.669) of the patients with positive fecal RNA had persistent shedding of fecal RNA and pooled results from the other 4 studies including 848 discharged patients with nucleic acid-negative stool samples indicated that the occurrence of repositive stool swabs was 18.1% (95% CI: 0.028-0.335), that of repositive respiratory swabs was 22.8% (95% CI: 0.003-0.452), that of both repositive stool and respiratory swabs was 19.1% (95% CI: 0.019-0.363), and that of only repositive stool swabs was 9.6% (95% CI: 0.010-0.203). The digestive tract may be an important organ involved in COVID-19 infection and in the excretion of the virus. Because of the potential risk of fecal-oral transmission, giving emphasis on stool swab tests can help increase the detection rate of asymptomatic carriers and reduce missed diagnoses.


Subject(s)
COVID-19 , Gastrointestinal Diseases , COVID-19/diagnosis , Feces , Humans , RNA, Viral/genetics , SARS-CoV-2/genetics
5.
Nucleic Acids Res ; 48(22): 12792-12803, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33270890

ABSTRACT

Telomeres at the ends of eukaryotic chromosomes are essential for genome integrality and stability. In order to identify genes that sustain telomere maintenance independently of telomerase recruitment, we have exploited the phenotype of over-long telomeres in the cells that express Cdc13-Est2 fusion protein, and examined 195 strains, in which individual non-essential gene deletion causes telomere shortening. We have identified 24 genes whose deletion results in dramatic failure of Cdc13-Est2 function, including those encoding components of telomerase, Yku, KEOPS and NMD complexes, as well as quite a few whose functions are not obvious in telomerase activity regulation. We have characterized Swc4, a shared subunit of histone acetyltransferase NuA4 and chromatin remodeling SWR1 (SWR1-C) complexes, in telomere length regulation. Deletion of SWC4, but not other non-essential subunits of either NuA4 or SWR1-C, causes significant telomere shortening. Consistently, simultaneous disassembly of NuA4 and SWR1-C does not affect telomere length. Interestingly, inactivation of Swc4 in telomerase null cells accelerates both telomere shortening and senescence rates. Swc4 associates with telomeric DNA in vivo, suggesting a direct role of Swc4 at telomeres. Taken together, our work reveals a distinct role of Swc4 in telomere length regulation, separable from its canonical roles in both NuA4 and SWR1-C.


Subject(s)
Adenosine Triphosphatases/genetics , Histone Acetyltransferases/genetics , Saccharomyces cerevisiae Proteins/genetics , Telomere Homeostasis/genetics , Chromatin/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Fungal/genetics , Histones/genetics , Humans , Multiprotein Complexes/genetics , Saccharomyces cerevisiae/genetics , Telomerase/genetics , Telomere/genetics , Telomere-Binding Proteins/genetics
6.
Nucleic Acids Res ; 47(11): 5684-5697, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30937455

ABSTRACT

KEOPS complex is one of the most conserved protein complexes in eukaryotes. It plays important roles in both telomere uncapping and tRNA N6-threonylcarbamoyladenosine (t6A) modification in budding yeast. But whether KEOPS complex plays any roles in DNA repair remains unknown. Here, we show that KEOPS complex plays positive roles in both DNA damage response and homologous recombination-mediated DNA repair independently of its t6A synthesis function. Additionally, KEOPS displays DNA binding activity in vitro, and is recruited to the chromatin at DNA breaks in vivo, suggesting a direct role of KEOPS in DSB repair. Mechanistically, KEOPS complex appears to promote DNA end resection through facilitating the association of Exo1 and Dna2 with DNA breaks. Interestingly, inactivation of both KEOPS and Mre11/Rad50/Xrs2 (MRX) complexes results in synergistic defect in DNA resection, revealing that KEOPS and MRX have some redundant functions in DNA resection. Thus we uncover a t6A-independent role of KEOPS complex in DNA resection, and propose that KEOPS might be a DSB sensor to assist cells in maintaining chromosome stability.


Subject(s)
DNA Helicases/physiology , DNA, Fungal , Exodeoxyribonucleases/physiology , Homologous Recombination , Recombinational DNA Repair , Saccharomyces cerevisiae Proteins/physiology , Binding, Competitive , Chromatin/chemistry , DNA/chemistry , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/metabolism , Exodeoxyribonucleases/metabolism , Metalloendopeptidases/metabolism , Mutation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Telomere/metabolism , Transcription Factors/metabolism
7.
Nucleic Acids Res ; 45(6): 3308-3322, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28180293

ABSTRACT

Rad6 and Bre1, ubiquitin-conjugating E2 and E3 enzymes respectively, are responsible for histone H2B lysine 123 mono-ubiquitination (H2Bub1) in Saccharomyces cerevisiae. Previous studies have shown that Rad6 and Bre1 regulate telomere length and recombination. However, the underlying molecular mechanism remains largely unknown. Here we report that H2BK123 mutation results in telomere shortening, while inactivation of Ubp8 and/or Ubp10, deubiquitinases of H2Bub1, leads to telomere lengthening in Rad6-Bre1-dependent manner. In telomerase-deficient cells, inactivation of Rad6-Bre1 pathway retards telomere shortening rate and the onset of senescence, while deletion of UBP8 and/or UBP10 accelerates senescence. Thus, Rad6-Bre1 pathway regulates both telomere length and recombination through its role in H2Bub1. Additionally, inactivation of both Rad6-Bre1-H2Bub1 and Mre11-Rad50-Xrs2 (MRX) pathways causes synthetic growth defects and telomere shortening in telomerase-proficient cells, and significantly accelerates senescence and eliminates type II telomere recombination in telomerase-deficient cells. Furthermore, RAD6 or BRE1 deletion, or H2BK123R mutation decreases the accumulation of ssDNA at telomere ends. These results support the model that Rad6-Bre1-H2Bub1 cooperates with MRX to promote telomere-end resection and thus positively regulates both telomerase- and recombination-dependent telomere replication. This study provides a mechanistic link between histone H2B ubiquitination and telomere replication.


Subject(s)
Histones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Telomere Homeostasis , Telomere/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination , DNA, Single-Stranded/metabolism , Endopeptidases/metabolism , Gene Deletion , Microbial Viability , Nuclear Proteins/metabolism , Recombination, Genetic , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Telomere Shortening , Telomere-Binding Proteins/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/genetics
8.
PLoS Genet ; 11(3): e1005071, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25822194

ABSTRACT

DNA double strand break (DSB) is one of the major damages that cause genome instability and cellular aging. The homologous recombination (HR)-mediated repair of DSBs plays an essential role in assurance of genome stability and cell longevity. Telomeres resemble DSBs and are competent for HR. Here we show that in budding yeast Saccharomyces cerevisiae telomere recombination elicits genome instability and accelerates cellular aging. Inactivation of KEOPS subunit Cgi121 specifically inhibits telomere recombination, and significantly extends cell longevity in both telomerase-positive and pre-senescing telomerase-negative cells. Deletion of CGI121 in the short-lived yku80(tel) mutant restores lifespan to cgi121Δ level, supporting the function of Cgi121 in telomeric single-stranded DNA generation and thus in promotion of telomere recombination. Strikingly, inhibition of telomere recombination is able to further slow down the aging process in long-lived fob1Δ cells, in which rDNA recombination is restrained. Our study indicates that HR activity at telomeres interferes with telomerase to pose a negative impact on cellular longevity.


Subject(s)
Longevity/genetics , Recombination, Genetic , Saccharomyces cerevisiae Proteins/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , Genomic Instability , Saccharomyces cerevisiae/genetics , Telomerase/genetics , Telomere/genetics
9.
Biochem J ; 467(3): 507-15, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25715670

ABSTRACT

Protein arginine methyltransferases (PRMTs) are a family of enzymes that can methylate protein arginine residues. PRMTs' substrates include histones and a variety of non-histone proteins. Previous studies have shown that yeast Hmt1 is a type I PRMT and methylates histone H4 arginine 3 and several mRNA-binding proteins. Hmt1 forms dimers or oligomers, but how dimerization or oligomerization affects its activity remains largely unknown. We now report that Hmt1 can methylate histone H3 arginine 2 (H3R2) in vitro. The dimerization but not hexamerization is essential for Hmt1's activity. Interestingly, the methyltransferase activity of Hmt1 on histone H3R2 requires reciprocal contributions from two Hmt1 molecules. Our results suggest an intermolecular trans-complementary mechanism by which Hmt1 dimer methylates its substrates.


Subject(s)
Histones/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Amino Acid Sequence , Arginine/chemistry , Catalytic Domain , Gene Deletion , Genes, Fungal , Histones/chemistry , Histones/genetics , Methylation , Models, Molecular , Molecular Sequence Data , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Quaternary , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid , Substrate Specificity
10.
PLoS Genet ; 9(1): e1003208, 2013.
Article in English | MEDLINE | ID: mdl-23390378

ABSTRACT

Telomeres are protein-DNA structures found at the ends of linear chromosomes and are crucial for genome integrity. Telomeric DNA length is primarily maintained by the enzyme telomerase. Cells lacking telomerase will undergo senescence when telomeres become critically short. In Saccharomyces cerevisiae, a very small percentage of cells lacking telomerase can remain viable by lengthening telomeres via two distinct homologous recombination pathways. These "survivor" cells are classified as either Type I or Type II, with each class of survivor possessing distinct telomeric DNA structures and genetic requirements. To elucidate the regulatory pathways contributing to survivor generation, we knocked out the telomerase RNA gene TLC1 in 280 telomere-length-maintenance (TLM) gene mutants and examined telomere structures in post-senescent survivors. We uncovered new functional roles for 10 genes that affect the emerging ratio of Type I versus Type II survivors and 22 genes that are required for Type II survivor generation. We further verified that Pif1 helicase was required for Type I recombination and that the INO80 chromatin remodeling complex greatly affected the emerging frequency of Type I survivors. Finally, we found the Rad6-mediated ubiquitination pathway and the KEOPS complex were required for Type II recombination. Our data provide an independent line of evidence supporting the idea that these genes play important roles in telomere dynamics.


Subject(s)
DNA Helicases , Homologous Recombination , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Telomerase/genetics , Chromosomes/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Genomic Instability , RNA/genetics , RNA/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Telomerase/metabolism , Telomere/genetics , Telomere Homeostasis/genetics , Ubiquitination
11.
EMBO J ; 30(14): 2829-42, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21685874

ABSTRACT

The SAGA (Spt-Ada-Gcn5 acetyltransferase) complex is an important chromatin modifying complex that can both acetylate and deubiquitinate histones. Sgf29 is a novel component of the SAGA complex. Here, we report the crystal structures of the tandem Tudor domains of Saccharomyces cerevisiae and human Sgf29 and their complexes with H3K4me2 and H3K4me3 peptides, respectively, and show that Sgf29 selectively binds H3K4me2/3 marks. Our crystal structures reveal that Sgf29 harbours unique tandem Tudor domains in its C-terminus. The tandem Tudor domains in Sgf29 tightly pack against each other face-to-face with each Tudor domain harbouring a negatively charged pocket accommodating the first residue alanine and methylated K4 residue of histone H3, respectively. The H3A1 and K4me3 binding pockets and the limited binding cleft length between these two binding pockets are the structural determinants in conferring the ability of Sgf29 to selectively recognize H3K4me2/3. Our in vitro and in vivo functional assays show that Sgf29 recognizes methylated H3K4 to recruit the SAGA complex to its targets sites and mediates histone H3 acetylation, underscoring the importance of Sgf29 in gene regulation.


Subject(s)
Acetyltransferases/chemistry , Acetyltransferases/metabolism , Gene Expression Regulation , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/metabolism , Histones/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Trans-Activators/metabolism , Acetylation , Acetyltransferases/genetics , Amino Acid Sequence , Blotting, Western , Chromatin Immunoprecipitation , Histone Acetyltransferases/genetics , Humans , Molecular Sequence Data , Peptide Fragments , Protein Processing, Post-Translational , Protein Structure, Tertiary , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid , Trans-Activators/genetics
12.
Nucleic Acids Res ; 40(2): 581-93, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21930512

ABSTRACT

Eukaryotic chromosome ends have a DNA-protein complex structure termed telomere. Integrity of telomeres is essential for cell proliferation. Genome-wide screenings for telomere length maintenance genes identified several components of the transcriptional regulator, the Mediator complex. Our work provides evidence that Mediator is involved in telomere length regulation and telomere heterochromatin maintenance. Tail module of Mediator is required for telomere silencing by promoting or stabilizing Sir protein binding and spreading on telomeres. Mediator binds on telomere and may be a component of telomeric chromatin. Our study reveals a specific role of Mediator complex at the heterochromatic telomere and this function is specific to telomeres as it has no effect on the HMR locus.


Subject(s)
Heterochromatin/metabolism , Mediator Complex/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Telomere Homeostasis , Telomere/metabolism , Acetylation , Gene Silencing , Heterochromatin/chemistry , Histones/metabolism , Mediator Complex/genetics , Mutation , Protein Subunits/genetics , Protein Subunits/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Shelterin Complex , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Sirtuin 2/metabolism , Telomere/enzymology , Telomere-Binding Proteins/metabolism , Transcription Factors/metabolism
13.
PLoS Genet ; 7(1): e1001272, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21249184

ABSTRACT

Recent studies have established that the highly condensed and transcriptionally silent heterochromatic domains in budding yeast are virtually dynamic structures. The underlying mechanisms for heterochromatin dynamics, however, remain obscure. In this study, we show that histones are dynamically acetylated on H4K12 at telomeric heterochromatin, and this acetylation regulates several of the dynamic telomere properties. Using a de novo heterochromatin formation assay, we surprisingly found that acetylated H4K12 survived the formation of telomeric heterochromatin. Consistently, the histone acetyltransferase complex NuA4 bound to silenced telomeric regions and acetylated H4K12. H4K12 acetylation prevented the over-accumulation of Sir proteins at telomeric heterochromatin and elimination of this acetylation caused defects in multiple telomere-related processes, including transcription, telomere replication, and recombination. Together, these data shed light on a potential histone acetylation mark within telomeric heterochromatin that contributes to telomere plasticity.


Subject(s)
Heterochromatin/genetics , Histones/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Telomere/genetics , Acetylation , DNA Replication , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Lysine/metabolism , Recombination, Genetic , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Telomerase/metabolism , Transcription, Genetic
14.
Elife ; 122024 Apr 24.
Article in English | MEDLINE | ID: mdl-38656297

ABSTRACT

Telomeres, which are chromosomal end structures, play a crucial role in maintaining genome stability and integrity in eukaryotes. In the baker's yeast Saccharomyces cerevisiae, the X- and Y'-elements are subtelomeric repetitive sequences found in all 32 and 17 telomeres, respectively. While the Y'-elements serve as a backup for telomere functions in cells lacking telomerase, the function of the X-elements remains unclear. This study utilized the S. cerevisiae strain SY12, which has three chromosomes and six telomeres, to investigate the role of X-elements (as well as Y'-elements) in telomere maintenance. Deletion of Y'-elements (SY12YΔ), X-elements (SY12XYΔ+Y), or both X- and Y'-elements (SY12XYΔ) did not impact the length of the terminal TG1-3 tracks or telomere silencing. However, inactivation of telomerase in SY12YΔ, SY12XYΔ+Y, and SY12XYΔ cells resulted in cellular senescence and the generation of survivors. These survivors either maintained their telomeres through homologous recombination-dependent TG1-3 track elongation or underwent microhomology-mediated intra-chromosomal end-to-end joining. Our findings indicate the non-essential role of subtelomeric X- and Y'-elements in telomere regulation in both telomerase-proficient and telomerase-null cells and suggest that these elements may represent remnants of S. cerevisiae genome evolution. Furthermore, strains with fewer or no subtelomeric elements exhibit more concise telomere structures and offer potential models for future studies in telomere biology.


Subject(s)
Repetitive Sequences, Nucleic Acid , Saccharomyces cerevisiae , Telomerase , Telomere , Saccharomyces cerevisiae/genetics , Telomere/metabolism , Telomere/genetics , Repetitive Sequences, Nucleic Acid/genetics , Telomerase/genetics , Telomerase/metabolism , Telomere Homeostasis , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Deletion
15.
EMBO J ; 28(10): 1466-78, 2009 May 20.
Article in English | MEDLINE | ID: mdl-19369944

ABSTRACT

In budding yeast Saccharomyces cerevisiae, telomere length maintenance involves a complicated network as more than 280 telomere maintenance genes have been identified in the nonessential gene deletion mutant set. As a supplement, we identified additional 29 telomere maintenance genes, which were previously taken as essential genes. In this study, we report a novel function of Sua5p in telomere replication. Epistasis analysis and telomere sequencing show that sua5Delta cells display progressively shortened telomeres at early passages, and Sua5 functions downstream telomerase recruitment. Further, biochemical, structural and genetic studies show that Sua5p specifically binds single-stranded telomeric (ssTG) DNA in vitro through a distinct DNA-binding region on its surface, and the DNA-binding ability is essential for its telomere function. Thus, Sua5p represents a novel ssTG DNA-binding protein and positively regulates the telomere length in vivo.


Subject(s)
DNA Replication , DNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Telomere/metabolism , DNA, Fungal/metabolism , DNA-Binding Proteins/genetics , Gene Deletion , Saccharomyces cerevisiae Proteins/genetics
16.
Cell Discov ; 9(1): 11, 2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36693846

ABSTRACT

Chromosomes occupy discrete spaces in the interphase cell nucleus, called chromosome territory. The structural and functional relevance of chromosome territory remains elusive. We fused chromosome 15 and 17 in mouse haploid embryonic stem cells (haESCs), resulting in distinct changes of territories in the cognate chromosomes, but with little effect on gene expression, pluripotency and gamete functions of haESCs. The karyotype-engineered haESCs were successfully implemented in generating heterozygous (2n = 39) and homozygous (2n = 38) mouse models. Mice containing the fusion chromosome are fertile, and their representative tissues and organs display no phenotypic abnormalities, suggesting unscathed development. These results indicate that the mammalian chromosome architectures are highly resilient, and reorganization of chromosome territories can be readily tolerated during cell differentiation and mouse development.

17.
DNA Repair (Amst) ; 127: 103512, 2023 07.
Article in English | MEDLINE | ID: mdl-37230009

ABSTRACT

In the baker's yeast Saccharomyces cerevisiae, NuA4 and SWR1-C, two multisubunit complexes, are involved in histone acetylation and chromatin remodeling, respectively. Eaf1 is the assembly platform subunit of NuA4, Swr1 is the assembly platform and catalytic subunit of SWR1-C, while Swc4, Yaf9, Arp4 and Act1 form a functional module, and is present in both NuA4 and SWR1 complexes. ACT1 and ARP4 are essential for cell survival. Deletion of SWC4, but not YAF9, EAF1 or SWR1 results in a severe growth defect, but the underlying mechanism remains largely unknown. Here, we show that swc4Δ, but not yaf9Δ, eaf1Δ, or swr1Δ cells display defects in DNA ploidy and chromosome segregation, suggesting that the defects observed in swc4Δ cells are independent of NuA4 or SWR1-C integrity. Swc4 is enriched in the nucleosome-free regions (NFRs) of the genome, including characteristic regions of RDN5s, tDNAs and telomeres, independently of Yaf9, Eaf1 or Swr1. In particular, rDNA, tDNA and telomere loci are more unstable and prone to recombination in the swc4Δ cells than in wild-type cells. Taken together, we conclude that the chromatin associated Swc4 protects nucleosome-free chromatin of rDNA, tDNA and telomere loci to ensure genome integrity.


Subject(s)
Nucleosomes , Saccharomyces cerevisiae Proteins , Humans , Histones/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , DNA, Ribosomal , Chromatin , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Telomere/genetics , Telomere/metabolism , Genomic Instability , Chromatin Assembly and Disassembly , Histone Acetyltransferases/genetics , Transcription Factors/genetics
18.
J Biol Chem ; 286(40): 34770-6, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21849496

ABSTRACT

Recent transcription of GAL genes transiently leaves an H3K4 methylation mark at their promoters, providing an epigenetic memory for the recent transcriptional activity. However, the physiological significance of this mark is enigmatic. In our study, we show that the transient H3K4 di- and trimethylation at recently transcribed GAL1 inhibited the reinduction of GAL1. The H3K4 methylation functioned by recruiting the Isw1 ATPase onto GAL1 and thereby limiting the action of RNA polymerase II during GAL1 reactivation. Strikingly, the H3K4 methylation was also observed at the promoters of inositol- and fatty acid-responsive genes after recent transcription and played a negative role in their reinduction. Taken together, our data present a new mechanism by which H3K4 methylation regulates gene transcription.


Subject(s)
Galactokinase/metabolism , Gene Expression Regulation, Fungal , Histones/chemistry , Lysine/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphatases/chemistry , Epigenesis, Genetic , Fungal Proteins/metabolism , Galactose/metabolism , Gene Deletion , Kinetics , Methylation , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/metabolism , Transcription, Genetic
19.
Biochem J ; 433(2): 295-302, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21067515

ABSTRACT

Saccharomyces cerevesiae Rph1 is a histone demethylase orthologous to human JMJD2A (Jumonji-domain-containing protein 2A) that can specifically demethylate tri- and di-methylated Lys³6 of histone H3. c-Rph1, the catalytic core of Rph1, is responsible for the demethylase activity, which is essential for the transcription elongation of some actively transcribed genes. In the present work, we report the crystal structures of c-Rph1 in apo form and in complex with Ni²(+) and α-KG [2-oxoglutarate (α-ketoglutarate)]. The structure of c-Rph1 is composed of a JmjN (Jumonji N) domain, a long ß-hairpin, a mixed structural motif and a JmjC domain. The α-KG cofactor forms hydrogen-bonding interactions with the side chains of conserved residues, and the Ni²(+) ion at the active site is chelated by conserved residues and the cofactor. Structural comparison of Rph1 with JMJD2A indicates that the substrate-binding cleft of Rph1 is formed with several structural elements of the JmjC domain, the long ß-hairpin and the mixed structural motif; and the methylated Lys³6 of H3 is recognized by several conserved residues of the JmjC domain. In vitro biochemical results show that mutations of the key residues at the catalytic centre and in the substrate-binding cleft abolish the demethylase activity. In vivo growth phenotype analyses also demonstrate that these residues are essential for its functional roles in transcription elongation. Taken together, our structural and biological data provide insights into the molecular basis of the histone demethylase activity and the substrate specificity of Rph1.


Subject(s)
Biocatalysis , Catalytic Domain , Histone Demethylases/chemistry , Repressor Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Amino Acid Sequence , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
20.
PLoS Genet ; 5(6): e1000535, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19557187

ABSTRACT

Telomeres are nucleoprotein structures located at the linear ends of eukaryotic chromosomes. Telomere integrity is required for cell proliferation and survival. Although the vast majority of eukaryotic species use telomerase as a primary means for telomere maintenance, a few species can use recombination or retrotransposon-mediated maintenance pathways. Since Saccharomyces cerevisiae can use both telomerase and recombination to replicate telomeres, budding yeast provides a useful system with which to examine the evolutionary advantages of telomerase and recombination in preserving an organism or cell under natural selection. In this study, we examined the life span in telomerase-null, post-senescent type II survivors that have employed homologous recombination to replicate their telomeres. Type II recombination survivors stably maintained chromosomal integrity but exhibited a significantly reduced replicative life span. Normal patterns of cell morphology at the end of a replicative life span and aging-dependent sterility were observed in telomerase-null type II survivors, suggesting the type II survivors aged prematurely in a manner that is phenotypically consistent with that of wild-type senescent cells. The shortened life span of type II survivors was extended by calorie restriction or TOR1 deletion, but not by Fob1p inactivation or Sir2p over-expression. Intriguingly, rDNA recombination was decreased in type II survivors, indicating that the premature aging of type II survivors was not caused by an increase in extra-chromosomal rDNA circle accumulation. Reintroduction of telomerase activity immediately restored the replicative life span of type II survivors despite their heterogeneous telomeres. These results suggest that telomere recombination accelerates cellular aging in telomerase-null type II survivors and that telomerase is likely a superior telomere maintenance pathway in sustaining yeast replicative life span.


Subject(s)
Recombination, Genetic , Saccharomyces cerevisiae/physiology , Telomere/genetics , Cellular Senescence , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Telomere/physiology
SELECTION OF CITATIONS
SEARCH DETAIL