Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Methods Protoc ; 5(5)2022 Oct 04.
Article in English | MEDLINE | ID: mdl-36287051

ABSTRACT

Until recently, the favored method for making directed modifications to the budding yeast genome involved the introduction of a DNA template carrying the desired genetic changes along with a selectable marker, flanked by homology arms. This approach both limited the ability to make changes within genes due to disruption by the introduced selectable marker and prevented the use of that selectable marker for subsequent genomic manipulations. Following the discovery of CRISPR-Cas9-mediated genome editing, protocols were developed for modifying any DNA region of interest in a similar single transformation step without the need for a permanent selectable marker. This approach involves the generation of a DNA double-strand break (DSB) at the desired genomic location by the Cas9 nuclease, expressed on a plasmid which also expresses the guide RNA (gRNA) sequence directing the location of the DSB. The DSB is subsequently repaired via homologous recombination using a PCR-derived DNA repair template. Here, we describe in detail an improved method for incorporation of the gRNA-encoding DNA sequences into the Cas9 expression plasmid. Using Golden Gate cloning, annealed oligonucleotides bearing unique single-strand DNA overhangs are ligated into directional restriction enzyme sites. We describe the use of this CRISPR-Cas9 genome editing protocol to introduce multiple types of directed genetic changes into the yeast genome.

2.
DNA Repair (Amst) ; 108: 103240, 2021 12.
Article in English | MEDLINE | ID: mdl-34687987

ABSTRACT

Unlike all other biological molecules that are degraded and replaced if damaged, DNA must be repaired as chromosomes cannot be replaced. Indeed, DNA endures a wide variety of structural damage that need to be repaired accurately to maintain genomic stability and proper functioning of cells and to prevent mutation leading to disease. Given that the genome is packaged into chromatin within eukaryotic cells, it has become increasingly evident that the chromatin context of DNA both facilitates and regulates DNA repair processes. In this review, we discuss mechanisms involved in removal of histones (chromatin disassembly) from around DNA lesions, by histone chaperones and chromatin remodelers, that promotes accessibility of the DNA repair machinery. We also elaborate on how the deposition of core histones and specific histone variants onto DNA (chromatin assembly) during DNA repair promotes repair processes, the role of histone post translational modifications in these processes and how chromatin structure is reestablished after DNA repair is complete.


Subject(s)
DNA Repair , Histones , Animals , Chromatin , Chromatin Assembly and Disassembly , Histones/metabolism , Humans
3.
DNA Repair (Amst) ; 108: 103217, 2021 12.
Article in English | MEDLINE | ID: mdl-34481157

ABSTRACT

The RING-type E3 ubiquitin ligases RNF8 and RNF168 recruit DNA damage response (DDR) factors to chromatin flanking DNA double strand breaks (DSBs) including 53BP1, which protects DNA ends from resection during DNA DSB repair by non-homologous end joining (NHEJ). Deficiency of RNF8 or RNF168 does not lead to demonstrable NHEJ defects, but like deficiency of 53BP1, the combined deficiency of XLF and RNF8 or RNF168 leads to diminished NHEJ in lymphocytes arrested in G0/G1 phase. The function of RNF8 in NHEJ depends on its E3 ubiquitin ligase activity. Loss of RNF8 or RNF168 in G0/G1-phase lymphocytes leads to the resection of broken DNA ends, demonstrating that RNF8 and RNF168 function to protect DNA ends from nucleases, pos sibly through the recruitment of 53BP1. However, the loss of 53BP1 leads to more severe resection than the loss of RNF8 or RNF168. Moreover, in 53BP1-deficient cells, the loss of RNF8 or RNF168 leads to diminished DNA end resection. We conclude that RNF8 and RNF168 regulate pathways that both prevent and promote DNA end resection in cells arrested in G0/G1 phase.


Subject(s)
DNA-Binding Proteins , Ubiquitin , DNA/metabolism , DNA End-Joining Repair , DNA Repair , DNA-Binding Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
5.
Cancer Cell ; 36(4): 402-417.e13, 2019 10 14.
Article in English | MEDLINE | ID: mdl-31564638

ABSTRACT

Metastasis is the leading cause of cancer mortality. Chromatin remodeling provides the foundation for the cellular reprogramming necessary to drive metastasis. However, little is known about the nature of this remodeling and its regulation. Here, we show that metastasis-inducing pathways regulate histone chaperones to reduce canonical histone incorporation into chromatin, triggering deposition of H3.3 variant at the promoters of poor-prognosis genes and metastasis-inducing transcription factors. This specific incorporation of H3.3 into chromatin is both necessary and sufficient for the induction of aggressive traits that allow for metastasis formation. Together, our data clearly show incorporation of histone variant H3.3 into chromatin as a major regulator of cell fate during tumorigenesis, and histone chaperones as valuable therapeutic targets for invasive carcinomas.


Subject(s)
Carcinoma/pathology , Chromatin/metabolism , Gene Expression Regulation, Neoplastic , Histones/metabolism , Neoplasm Metastasis/genetics , Animals , Carcinogenesis/genetics , Carcinoma/genetics , Cell Line, Tumor , Chromatin/genetics , Chromatin Assembly Factor-1/genetics , Chromatin Assembly Factor-1/metabolism , Disease Progression , Epigenesis, Genetic , Epithelial-Mesenchymal Transition/genetics , Female , Histones/genetics , Humans , Male , Mice , Promoter Regions, Genetic/genetics , RNA-Seq , Transcription Factors/genetics , Xenograft Model Antitumor Assays
6.
Curr Genet ; 65(3): 717-720, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30673825

ABSTRACT

The number of times a cell divides before irreversibly arresting is termed replicative lifespan. Despite discovery of many chemical, dietary and genetic interventions that extend replicative lifespan, usually first discovered in budding yeast and subsequently shown to apply to metazoans, there is still little understanding of the underlying molecular mechanisms involved. One unifying theme is that most, if not all, interventions that extend replicative lifespan induce "hormesis", where a little inflicted damage makes cells more able to resist similar challenges in the future. One of the many cellular changes that occur during hormesis is a global reduction in protein synthesis, which has been linked to enhanced longevity in many organisms. Our recent study in budding yeast found that it was not the reduction in protein synthesis per se, but rather the subsequent induction of the conserved Gcn4 transcriptional regulator and its ability to induce autophagy that was responsible for extending replicative lifespan. We propose that Gcn4-dependent induction of autophagy occurring downstream of reduced global protein synthesis may be a unifying molecular mechanism for many interventions that extend replicative lifespan.


Subject(s)
Autophagy , Basic-Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation, Fungal , Hormesis , Longevity , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
7.
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
8.
Cell Cycle ; 17(12): 1413-1424, 2018.
Article in English | MEDLINE | ID: mdl-29954236

ABSTRACT

The Ataxia-telangiectasia mutated (ATM) kinase and the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) are activated by DNA double-strand breaks (DSBs). These DSBs occur in the context of chromatin but how chromatin influences the activation of these kinases is not known. Here we show that loss of the replication-dependent chromatin assembly factors ASF1A/B or CAF-1 compromises ATM activation, while augmenting DNA-PKcs activation, in response to DNA DSBs. Cells deficient in ASF1A/B or CAF-1 exhibit reduced histone H4 lysine 16 acetylation (H4K16ac), a histone mark known to promote ATM activation. ASF1A interacts with the histone acetyl transferase, hMOF that mediates H4K16ac. ASF1A depletion leads to increased recruitment of DNA-PKcs to DSBs. We propose normal chromatin assembly and H4K16ac during DNA replication is required to regulate ATM and DNA-PKcs activity in response to the subsequent induction of DNA DSBs.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Cell Cycle Proteins/genetics , DNA-Activated Protein Kinase/genetics , Histone Chaperones/genetics , Nuclear Proteins/genetics , Acetylation , Cell Line, Tumor , Chromatin/genetics , DNA/genetics , DNA Breaks, Double-Stranded , DNA Replication/genetics , HCT116 Cells , HeLa Cells , Histones/genetics , Humans , Molecular Chaperones , Signal Transduction/genetics
9.
Mol Cell ; 69(5): 879-892.e5, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29478807

ABSTRACT

The access-repair-restore model for the role of chromatin in DNA repair infers that chromatin is a mere obstacle to DNA repair. However, here we show that blocking chromatin assembly, via knockdown of the histone chaperones ASF1 or CAF-1 or a mutation that prevents ASF1A binding to histones, hinders Rad51 loading onto ssDNA during homologous recombination. This is a consequence of reduced recruitment of the Rad51 loader MMS22L-TONSL to ssDNA, resulting in persistent RPA foci, extensive DNA end resection, persistent activation of the ATR-Chk1 pathway, and cell cycle arrest. In agreement, histones occupy ssDNA during DNA repair in yeast. We also uncovered DNA-PKcs-dependent DNA damage-induced ASF1A phosphorylation, which enhances chromatin assembly, promoting MMS22L-TONSL recruitment and, hence, Rad51 loading. We propose that transient assembly of newly synthesized histones onto ssDNA serves to recruit MMS22L-TONSL to efficiently form the Rad51 nucleofilament for strand invasion, suggesting an active role of chromatin assembly in homologous recombination.


Subject(s)
Cell Cycle Proteins/metabolism , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Homologous Recombination , Molecular Chaperones/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Rad51 Recombinase/metabolism , Cell Cycle Checkpoints/physiology , Cell Cycle Proteins/genetics , Chromatin Assembly Factor-1/genetics , Chromatin Assembly Factor-1/metabolism , DNA Damage/physiology , DNA, Single-Stranded/genetics , DNA-Binding Proteins/genetics , HeLa Cells , Humans , K562 Cells , Molecular Chaperones/genetics , NF-kappa B/genetics , Nuclear Proteins/genetics , Rad51 Recombinase/genetics
10.
Nat Commun ; 8(1): 56, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28676626

ABSTRACT

Upon environmental changes, proliferating cells delay cell cycle to prevent further damage accumulation. Yeast Cip1 is a Cdk1 and Cln2-associated protein. However, the function and regulation of Cip1 are still poorly understood. Here we report that Cip1 expression is co-regulated by the cell-cycle-mediated factor Mcm1 and the stress-mediated factors Msn2/4. Overexpression of Cip1 arrests cell cycle through inhibition of Cdk1-G1 cyclin complexes at G1 stage and the stress-activated protein kinase-dependent Cip1 T65, T69, and T73 phosphorylation may strengthen the Cip1and Cdk1-G1 cyclin interaction. Cip1 accumulation mainly targets Cdk1-Cln3 complex to prevent Whi5 phosphorylation and inhibit early G1 progression. Under osmotic stress, Cip1 expression triggers transient G1 delay which plays a functionally redundant role with another hyperosmolar activated CKI, Sic1. These findings indicate that Cip1 functions similarly to mammalian p21 as a stress-induced CDK inhibitor to decelerate cell cycle through G1 cyclins to cope with environmental stresses.A G1 cell cycle regulatory kinase Cip1 has been identified in budding yeast but how this is regulated is unclear. Here the authors identify cell cycle (Mcm1) and stress-mediated (Msn 2/4) transcription factors as regulating Cip1, causing stress induced CDK inhibition and delay in cell cycle progression.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclins/metabolism , Gene Expression Regulation, Fungal , Saccharomyces cerevisiae Proteins/metabolism , DNA-Binding Proteins/metabolism , Minichromosome Maintenance 1 Protein/metabolism , Mitogen-Activated Protein Kinases/metabolism , Osmotic Pressure , Saccharomyces cerevisiae , Stress, Physiological , Transcription Factors/metabolism
11.
Elife ; 52016 Sep 30.
Article in English | MEDLINE | ID: mdl-27690308

ABSTRACT

The histone chaperone Chromatin Assembly Factor 1 (CAF-1) deposits tetrameric (H3/H4)2 histones onto newly-synthesized DNA during DNA replication. To understand the mechanism of the tri-subunit CAF-1 complex in this process, we investigated the protein-protein interactions within the CAF-1-H3/H4 architecture using biophysical and biochemical approaches. Hydrogen/deuterium exchange and chemical cross-linking coupled to mass spectrometry reveal interactions that are essential for CAF-1 function in budding yeast, and importantly indicate that the Cac1 subunit functions as a scaffold within the CAF-1-H3/H4 complex. Cac1 alone not only binds H3/H4 with high affinity, but also promotes histone tetramerization independent of the other subunits. Moreover, we identify a minimal region in the C-terminus of Cac1, including the structured winged helix domain and glutamate/aspartate-rich domain, which is sufficient to induce (H3/H4)2 tetramerization. These findings reveal a key role of Cac1 in histone tetramerization, providing a new model for CAF-1-H3/H4 architecture and function during eukaryotic replication.

13.
Genome Biol ; 15(12): 513, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25517638

ABSTRACT

BACKGROUND: Hypoxia induces the epithelial-mesenchymal transition, EMT, to promote cancer metastasis. In addition to transcriptional regulation mediated by hypoxia-inducible factors, HIFs, other epigenetic mechanisms of gene regulation, such as histone modifications and DNA methylation, are utilized under hypoxia. However, whether DNA demethylation mediated by TET1, a DNA dioxygenase converting 5-methylcytosine, 5mC, into 5-hydroxymethylcytosine, 5hmC, plays a role in hypoxia-induced EMT is largely unknown. RESULTS: We show that TET1 regulates hypoxia-responsive gene expression. Hypoxia/HIF-2α regulates the expression of TET1. Knockdown of TET1 mitigates hypoxia-induced EMT. RNA sequencing and 5hmC sequencing identified the set of TET1-regulated genes. Cholesterol metabolic process genes are among the genes that showed high prevalence and statistical significance. We characterize one of the genes, INSIG1 (insulin induced gene 1), to confirm its expression and the 5hmC levels in its promoter. Knockdown of INSIG1 also mitigates hypoxia-induced EMT. Finally, TET1 is shown to be a transcriptional co-activator that interacts with HIF-1α and HIF-2α to enhance their transactivation activity independent of its enzymatic activity. TET1 acts as a co-activator to further enhance the expression of INSIG1 together with HIF-2α. We define the domain in HIF-1α that interacts with TET1 and map the domain in TET1 that confers transactivation to a 200 amino acid region that contains a CXXC domain. The TET1 catalytically inactive mutant is capable of rescuing hypoxia-induced EMT in TET1 knockdown cells. CONCLUSIONS: These findings demonstrate that TET1 serves as a transcription co-activator to regulate hypoxia-responsive gene expression and EMT, in addition to its role in demethylating 5mC.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Epithelial-Mesenchymal Transition , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/metabolism , Catalytic Domain , Cell Hypoxia , Cell Line, Tumor , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mixed Function Oxygenases , Neoplasms/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics
14.
Nat Commun ; 5: 5312, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25387524

ABSTRACT

In yeast, the initiation of telomere replication at the late S phase involves in combined actions of kinases on Cdc13, the telomere binding protein. Cdc13 recruits telomerase to telomeres through its interaction with Est1, a component of telomerase. However, how cells terminate the function of telomerase at G2/M is still elusive. Here we show that the protein phosphatase 2A (PP2A) subunit Pph22 and the yeast Aurora kinase homologue Ipl1 coordinately inhibit telomerase at G2/M by dephosphorylating and phosphorylating the telomerase recruitment domain of Cdc13, respectively. While Pph22 removes Tel1/Mec1-mediated Cdc13 phosphorylation to reduce Cdc13-Est1 interaction, Ipl1-dependent Cdc13 phosphorylation elicits dissociation of Est1-TLC1, the template RNA component of telomerase. Failure of these regulations prevents telomerase from departing telomeres, causing perturbed telomere lengthening and prolonged M phase. Together our results demonstrate that differential and additive actions of PP2A and Aurora on Cdc13 limit telomerase action by removing active telomerase from telomeres at G2/M phase.


Subject(s)
Aurora Kinases/physiology , Cell Division/physiology , G2 Phase/physiology , Protein Phosphatase 2/physiology , Saccharomyces cerevisiae Proteins/physiology , Telomerase/physiology , Telomere-Binding Proteins/physiology , Telomere/physiology , Aurora Kinases/metabolism , Protein Phosphatase 2/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Telomerase/metabolism , Telomere/metabolism , Telomere-Binding Proteins/metabolism
15.
Nucleic Acids Res ; 37(11): 3602-11, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19359360

ABSTRACT

Budding yeast telomerase is mainly activated by Tel1/Mec1 (yeast ATM/ATR) on Cdc13 from late S to G2 phase of the cell cycle. Here, we demonstrated that the telomerase-recruitment domain of Cdc13 is also phosphorylated by Cdk1 at the same cell cycle stage as the Tel1/Mec1-dependent regulation. Phosphor-specific gel analysis demonstrated that Cdk1 phosphorylates residues 308 and 336 of Cdc13. The residue T308 of Cdc13 is critical for efficient Mec1-mediated S306 phosphorylation in vitro. Phenotypic analysis in vivo revealed that the mutations in the Cdc13 S/TP motifs phosphorylated by Cdk1 caused cell cycle delay and telomere shortening and these phenotypes could be partially restored by the replacement with a negative charge residue. In the absence of Ku or Tel1, Cdk1-mediated phosphorylation of Cdc13 showed no effect on telomere length maintenance. Moreover, this Cdk1-mediated phosphorylation was required to promote the regular turnover of Cdc13. Together these results demonstrate that Cdk1 phosphorylates the telomerase recruitment domain of Cdc13, thereby preserves optimal function and expression level of Cdc13 for precise telomere replication and cell cycle progression.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle/genetics , Saccharomyces cerevisiae Proteins/metabolism , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Homeostasis , Mutation , Phosphorylation , Protein Stability , Protein Structure, Tertiary , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Telomere/chemistry , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...