Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Mol Cell ; 77(6): 1206-1221.e7, 2020 03 19.
Article in English | MEDLINE | ID: mdl-31980388

ABSTRACT

Alternative polyadenylation (APA) contributes to transcriptome complexity by generating mRNA isoforms with varying 3' UTR lengths. APA leading to 3' UTR shortening (3' US) is a common feature of most cancer cells; however, the molecular mechanisms are not understood. Here, we describe a widespread mechanism promoting 3' US in cancer through ubiquitination of the mRNA 3' end processing complex protein, PCF11, by the cancer-specific MAGE-A11-HUWE1 ubiquitin ligase. MAGE-A11 is normally expressed only in the male germline but is frequently re-activated in cancers. MAGE-A11 is necessary for cancer cell viability and is sufficient to drive tumorigenesis. Screening for targets of MAGE-A11 revealed that it ubiquitinates PCF11, resulting in loss of CFIm25 from the mRNA 3' end processing complex. This leads to APA of many transcripts affecting core oncogenic and tumor suppressors, including cyclin D2 and PTEN. These findings provide insights into the molecular mechanisms driving APA in cancer and suggest therapeutic strategies.


Subject(s)
3' Untranslated Regions/genetics , Antigens, Neoplasm/metabolism , Lung Neoplasms/pathology , Neoplasm Proteins/metabolism , Ovarian Neoplasms/pathology , RNA, Messenger/metabolism , Ubiquitin/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , Antigens, Neoplasm/genetics , Apoptosis , Biomarkers, Tumor , Carcinogenesis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Cleavage And Polyadenylation Specificity Factor/genetics , Cleavage And Polyadenylation Specificity Factor/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Polyadenylation , RNA Splicing , RNA, Messenger/genetics , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Xenograft Model Antitumor Assays , mRNA Cleavage and Polyadenylation Factors/genetics
2.
Proc Natl Acad Sci U S A ; 121(20): e2400610121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38713623

ABSTRACT

Chromatin replication is intricately intertwined with the recycling of parental histones to the newly duplicated DNA strands for faithful genetic and epigenetic inheritance. The transfer of parental histones occurs through two distinct pathways: leading strand deposition, mediated by the DNA polymerase ε subunits Dpb3/Dpb4, and lagging strand deposition, facilitated by the MCM helicase subunit Mcm2. However, the mechanism of the facilitation of Mcm2 transferring parental histones to the lagging strand while moving along the leading strand remains unclear. Here, we show that the deletion of Pol32, a nonessential subunit of major lagging-strand DNA polymerase δ, results in a predominant transfer of parental histone H3-H4 to the leading strand during replication. Biochemical analyses further demonstrate that Pol32 can bind histone H3-H4 both in vivo and in vitro. The interaction of Pol32 with parental histone H3-H4 is disrupted through the mutation of the histone H3-H4 binding domain within Mcm2. Our findings identify the DNA polymerase δ subunit Pol32 as a critical histone chaperone downstream of Mcm2, mediating the transfer of parental histones to the lagging strand during DNA replication.


Subject(s)
DNA Replication , DNA-Directed DNA Polymerase , Saccharomyces cerevisiae Proteins , DNA Polymerase III/metabolism , DNA Polymerase III/genetics , Histones/metabolism , Minichromosome Maintenance Complex Component 2/metabolism , Minichromosome Maintenance Complex Component 2/genetics , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , DNA-Directed DNA Polymerase/metabolism
3.
Mol Cell ; 72(1): 140-151.e3, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30244834

ABSTRACT

Although essential for epigenetic inheritance, the transfer of parental histone (H3-H4)2 tetramers that contain epigenetic modifications to replicating DNA strands is poorly understood. Here, we show that the Mcm2-Ctf4-Polα axis facilitates the transfer of parental (H3-H4)2 tetramers to lagging-strand DNA at replication forks. Mutating the conserved histone-binding domain of the Mcm2 subunit of the CMG (Cdc45-MCM-GINS) DNA helicase, which translocates along the leading-strand template, results in a marked enrichment of parental (H3-H4)2 on leading strand, due to the impairment of the transfer of parental (H3-H4)2 to lagging strands. Similar effects are observed in Ctf4 and Polα primase mutants that disrupt the connection of the CMG helicase to Polα that resides on lagging-strand template. Our results support a model whereby parental (H3-H4)2 complexes displaced from nucleosomes by DNA unwinding at replication forks are transferred by the CMG-Ctf4-Polα complex to lagging-strand DNA for nucleosome assembly at the original location.


Subject(s)
DNA Polymerase III/genetics , DNA Replication/genetics , DNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Chromatin Assembly and Disassembly/genetics , DNA Helicases/genetics , Epigenesis, Genetic , Histones/genetics , Multiprotein Complexes/genetics , Nucleosomes/genetics , Protein Binding , Saccharomyces cerevisiae/genetics
4.
Nucleic Acids Res ; 52(9): 5138-5151, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38554108

ABSTRACT

Recycling of parental histones is an important step in epigenetic inheritance. During DNA replication, DNA polymerase epsilon subunit DPB3/DPB4 and DNA replication helicase subunit MCM2 are involved in the transfer of parental histones to the leading and lagging strands, respectively. Single Dpb3 deletion (dpb3Δ) or Mcm2 mutation (mcm2-3A), which each disrupts one parental histone transfer pathway, leads to the other's predominance. However, the biological impact of the two histone transfer pathways on chromatin structure and DNA repair remains elusive. In this study, we used budding yeast Saccharomyces cerevisiae to determine the genetic and epigenetic outcomes from disruption of parental histone H3-H4 tetramer transfer. We found that a dpb3Δ mcm2-3A double mutant did not exhibit the asymmetric parental histone patterns caused by a single dpb3Δ or mcm2-3A mutation, suggesting that the processes by which parental histones are transferred to the leading and lagging strands are independent. Surprisingly, the frequency of homologous recombination was significantly lower in dpb3Δ, mcm2-3A and dpb3Δ mcm2-3A mutants, likely due to the elevated levels of free histones detected in the mutant cells. Together, these findings indicate that proper transfer of parental histones during DNA replication is essential for maintaining chromatin structure and that lower homologous recombination activity due to parental histone transfer defects is detrimental to cells.


Subject(s)
DNA Replication , Histones , Homologous Recombination , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Histones/metabolism , Histones/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Homologous Recombination/genetics , DNA Replication/genetics , Mutation , Chromatin/metabolism , Chromatin/genetics , DNA Polymerase II/metabolism , DNA Polymerase II/genetics , Epigenesis, Genetic , DNA Repair
5.
Mol Cell ; 68(2): 446-455.e3, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29033319

ABSTRACT

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


Subject(s)
Cell Cycle Proteins/metabolism , Checkpoint Kinase 2/metabolism , DNA Polymerase III/metabolism , DNA Polymerase II/metabolism , DNA Replication/physiology , DNA, Fungal/biosynthesis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cell Cycle Proteins/genetics , Checkpoint Kinase 2/genetics , DNA Polymerase II/genetics , DNA Polymerase III/genetics , DNA, Fungal/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
6.
Mol Cell ; 65(2): 272-284, 2017 Jan 19.
Article in English | MEDLINE | ID: mdl-28107649

ABSTRACT

The histone chaperone HIRA is involved in depositing histone variant H3.3 into distinct genic regions, including promoters, enhancers, and gene bodies. However, how HIRA deposits H3.3 to these regions remains elusive. Through a short hairpin RNA (shRNA) screening, we identified single-stranded DNA binding protein replication protein A (RPA) as a regulator of the deposition of newly synthesized H3.3 into chromatin. We show that RPA physically interacts with HIRA to form RPA-HIRA-H3.3 complexes, and it co-localizes with HIRA and H3.3 at gene promoters and enhancers. Depletion of RPA1, the largest subunit of the RPA complex, dramatically reduces both HIRA association with chromatin and the deposition of newly synthesized H3.3 at promoters and enhancers and leads to altered transcription at gene promoters. These results support a model whereby RPA, best known for its role in DNA replication and repair, recruits HIRA to promoters and enhancers and regulates deposition of newly synthesized H3.3 to these regulatory elements for gene regulation.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Enhancer Elements, Genetic , Histone Chaperones/metabolism , Histones/metabolism , Promoter Regions, Genetic , Replication Protein A/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Binding Sites , Cell Cycle Proteins/genetics , Chromatin/genetics , DNA/genetics , DNA-Binding Proteins/genetics , G1 Phase , HEK293 Cells , HeLa Cells , Histone Chaperones/genetics , Humans , Protein Binding , Protein Interaction Domains and Motifs , RNA Interference , Replication Protein A/genetics , Transcription Factors/genetics , Transfection
7.
PLoS Biol ; 19(6): e3001281, 2021 06.
Article in English | MEDLINE | ID: mdl-34077419

ABSTRACT

Nutrient-responsive protein kinases control the balance between anabolic growth and catabolic processes such as autophagy. Aberrant regulation of these kinases is a major cause of human disease. We report here that the vertebrate nonreceptor tyrosine kinase Src-related kinase lacking C-terminal regulatory tyrosine and N-terminal myristylation sites (SRMS) inhibits autophagy and promotes growth in a nutrient-responsive manner. Under nutrient-replete conditions, SRMS phosphorylates the PHLPP scaffold FK506-binding protein 51 (FKBP51), disrupts the FKBP51-PHLPP complex, and promotes FKBP51 degradation through the ubiquitin-proteasome pathway. This prevents PHLPP-mediated dephosphorylation of AKT, causing sustained AKT activation that promotes growth and inhibits autophagy. SRMS is amplified and overexpressed in human cancers where it drives unrestrained AKT signaling in a kinase-dependent manner. SRMS kinase inhibition activates autophagy, inhibits cancer growth, and can be accomplished using the FDA-approved tyrosine kinase inhibitor ibrutinib. This illuminates SRMS as a targetable vulnerability in human cancers and as a new target for pharmacological induction of autophagy in vertebrates.


Subject(s)
Autophagy , Neoplasms/metabolism , Neoplasms/pathology , Tacrolimus Binding Proteins/metabolism , src-Family Kinases/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagy/drug effects , Beclin-1/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Enzyme Activation/drug effects , Mice , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Piperidines/pharmacology , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors
8.
Proc Natl Acad Sci U S A ; 118(38)2021 09 21.
Article in English | MEDLINE | ID: mdl-34531325

ABSTRACT

In response to DNA replication stress, DNA replication checkpoint kinase Mec1 phosphorylates Mrc1, which in turn activates Rad53 to prevent the generation of deleterious single-stranded DNA, a process that remains poorly understood. We previously reported that lagging-strand DNA synthesis proceeds farther than leading strand in rad53-1 mutant cells defective in replication checkpoint under replication stress, resulting in the exposure of long stretches of the leading-strand templates. Here, we show that asymmetric DNA synthesis is also observed in mec1-100 and mrc1-AQ cells defective in replication checkpoint but, surprisingly, not in mrc1∆ cells in which both DNA replication and checkpoint functions of Mrc1 are missing. Furthermore, depletion of either Mrc1 or its partner, Tof1, suppresses the asymmetric DNA synthesis in rad53-1 mutant cells. Thus, the DNA replication checkpoint pathway couples leading- and lagging-strand DNA synthesis by attenuating the replication function of Mrc1-Tof1 under replication stress.


Subject(s)
Cell Cycle Proteins/metabolism , Checkpoint Kinase 2/metabolism , DNA Replication/physiology , Saccharomyces cerevisiae Proteins/metabolism , Cell Cycle Proteins/genetics , Checkpoint Kinase 2/genetics , DNA Replication/genetics , DNA, Fungal/genetics , Intracellular Signaling Peptides and Proteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomycetales/genetics , Saccharomycetales/metabolism
9.
J Biol Chem ; 296: 100374, 2021.
Article in English | MEDLINE | ID: mdl-33548228

ABSTRACT

The recent discovery of the cancer-associated E76K mutation in histone H2B (H2BE76-to-K) in several types of cancers revealed a new class of oncohistone. H2BE76K weakens the stability of histone octamers, alters gene expression, and promotes colony formation. However, the mechanism linking the H2BE76K mutation to cancer development remains largely unknown. In this study, we knock in the H2BE76K mutation in MDA-MB-231 breast cancer cells using CRISPR/Cas9 and show that the E76K mutant histone H2B preferentially localizes to genic regions. Interestingly, genes upregulated in the H2BE76K mutant cells are enriched for the E76K mutant H2B and are involved in cell adhesion and proliferation pathways. We focused on one H2BE76K target gene, ADAM19 (a disintegrin and metalloproteinase-domain-containing protein 19), a gene highly expressed in various human cancers including breast invasive carcinoma, and demonstrate that H2BE76K directly promotes ADAM19 transcription by facilitating efficient transcription along the gene body. ADAM19 depletion reduced the colony formation ability of the H2BE76K mutant cells, whereas wild-type MDA-MB-231 cells overexpressing ADAM19 mimics the colony formation phenotype of the H2BE76K mutant cells. Collectively, our data demonstrate the mechanism by which H2BE76K deregulates the expression of genes that control oncogenic properties through a combined effect of its specific genomic localization and nucleosome destabilization effect.


Subject(s)
ADAM Proteins/genetics , Breast Neoplasms/genetics , Histones/genetics , ADAM Proteins/metabolism , Breast Neoplasms/metabolism , Carcinogenesis/genetics , Cell Line, Tumor , Female , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Histones/metabolism , Humans , Mutation/genetics , Nucleosomes , Oncogenes/genetics , Polymorphism, Single Nucleotide/genetics
10.
EMBO J ; 37(17)2018 09 03.
Article in English | MEDLINE | ID: mdl-30065069

ABSTRACT

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


Subject(s)
Genome, Fungal , Genomic Instability , Models, Biological , Molecular Chaperones/metabolism , Replication Protein A/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , DNA, Fungal/genetics , DNA, Fungal/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Karyopherins/genetics , Karyopherins/metabolism , Molecular Chaperones/genetics , Replication Protein A/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
11.
Mol Cell ; 56(4): 551-63, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25449133

ABSTRACT

In eukaryotic cells, DNA replication proceeds with continuous synthesis of leading-strand DNA and discontinuous synthesis of lagging-strand DNA. Here we describe a method, eSPAN (enrichment and sequencing of protein-associated nascent DNA), which reveals the genome-wide association of proteins with leading and lagging strands of DNA replication forks. Using this approach in budding yeast, we confirm the strand specificities of DNA polymerases delta and epsilon and show that the PCNA clamp is enriched at lagging strands compared with leading-strand replication. Surprisingly, at stalled forks, PCNA is unloaded specifically from lagging strands. PCNA unloading depends on the Elg1-containing alternative RFC complex, ubiquitination of PCNA, and the checkpoint kinases Mec1 and Rad53. Cells deficient in PCNA unloading exhibit increased chromosome breaks. Our studies provide a tool for studying replication-related processes and reveal a mechanism whereby checkpoint kinases regulate strand-specific unloading of PCNA from stalled replication forks to maintain genome stability.


Subject(s)
DNA Replication , DNA, Fungal/biosynthesis , Proliferating Cell Nuclear Antigen/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Chromatin Immunoprecipitation , Chromosomes, Fungal/genetics , DNA Damage , DNA Polymerase II/metabolism , DNA Polymerase III/metabolism , DNA, Fungal/genetics , Genomic Instability , Protein Binding , Sequence Analysis, DNA , Ubiquitination
12.
Proc Natl Acad Sci U S A ; 116(27): 13311-13319, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31209047

ABSTRACT

Cellular senescence defines an irreversible cell growth arrest state linked to loss of tissue function and aging in mammals. This transition from proliferation to senescence is typically characterized by increased expression of the cell-cycle inhibitor p16INK4a and formation of senescence-associated heterochromatin foci (SAHF). SAHF formation depends on HIRA-mediated nucleosome assembly of histone H3.3, which is regulated by the serine/threonine protein kinase Pak2. However, it is unknown if Pak2 contributes to cellular senescence. Here, we show that depletion of Pak2 delayed oncogene-induced senescence in IMR90 human fibroblasts and oxidative stress-induced senescence of mouse embryonic fibroblasts (MEFs), whereas overexpression of Pak2 accelerated senescence of IMR90 cells. Importantly, depletion of Pak2 in BubR1 progeroid mice attenuated the onset of aging-associated phenotypes and extended life span. Pak2 is required for expression of genes involved in cellular senescence and regulated the deposition of newly synthesized H3.3 onto chromatin in senescent cells. Together, our results demonstrate that Pak2 is an important regulator of cellular senescence and organismal aging, in part through the regulation of gene expression and H3.3 nucleosome assembly.


Subject(s)
Aging , Cellular Senescence , p21-Activated Kinases/physiology , Aging/metabolism , Animals , Cell Line , Chromatin Immunoprecipitation , Gene Expression Regulation , Histones/metabolism , Longevity , Mice, Knockout , Oxidative Stress , Real-Time Polymerase Chain Reaction , p21-Activated Kinases/metabolism
13.
Nucleic Acids Res ; 47(21): 11114-11131, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31586391

ABSTRACT

Establishment and subsequent maintenance of distinct chromatin domains during embryonic stem cell (ESC) differentiation are crucial for lineage specification and cell fate determination. Here we show that the histone chaperone Chromatin Assembly Factor 1 (CAF-1), which is recruited to DNA replication forks through its interaction with proliferating cell nuclear antigen (PCNA) for nucleosome assembly, participates in the establishment of H3K27me3-mediated silencing during differentiation. Deletion of CAF-1 p150 subunit impairs the silencing of many genes including Oct4, Sox2 and Nanog as well as the establishment of H3K27me3 at these gene promoters during ESC differentiation. Mutations of PCNA residues involved in recruiting CAF-1 to the chromatin also result in defects in differentiation in vitro and impair early embryonic development as p150 deletion. Together, these results reveal that the CAF-1-PCNA nucleosome assembly pathway plays an important role in the establishment of H3K27me3-mediated silencing during cell fate determination.


Subject(s)
Cell Differentiation/genetics , Chromatin Assembly Factor-1/physiology , Embryonic Stem Cells/physiology , Heterochromatin/metabolism , Pluripotent Stem Cells/physiology , Animals , Cells, Cultured , Chromatin Assembly Factor-1/genetics , Chromatin Assembly Factor-1/metabolism , Chromatin Assembly and Disassembly/genetics , DNA Replication/genetics , Gene Silencing/physiology , Histone Methyltransferases/metabolism , Histones/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Proliferating Cell Nuclear Antigen/metabolism
14.
Proc Natl Acad Sci U S A ; 115(27): E6162-E6171, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29915027

ABSTRACT

Bivalent chromatin domains containing repressive H3K27me3 and active H3K4me3 modifications are barriers for the expression of lineage-specific genes in ES cells and must be resolved for the transcription induction of these genes during differentiation, a process that remains largely unknown. Here, we show that Asf1a, a histone chaperone involved in nucleosome assembly and disassembly, regulates the resolution of bivalent domains and activation of lineage-specific genes during mouse ES cell differentiation. Deletion of Asf1a does not affect the silencing of pluripotent genes, but compromises the expression of lineage-specific genes during ES cell differentiation. Mechanistically, the Asf1a-histone interaction, but not the role of Asf1a in nucleosome assembly, is required for gene transcription. Asf1a is recruited to several bivalent promoters, partially through association with transcription factors, and mediates nucleosome disassembly during differentiation. We suggest that Asf1a-mediated nucleosome disassembly provides a means for resolution of bivalent domain barriers for induction of lineage-specific genes during differentiation.


Subject(s)
Cell Differentiation , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , Histones/metabolism , Mouse Embryonic Stem Cells/metabolism , Nucleosomes/metabolism , Animals , Cell Cycle Proteins , Chromosomal Proteins, Non-Histone/genetics , Gene Deletion , Histones/genetics , Mice , Molecular Chaperones , Mouse Embryonic Stem Cells/cytology , Nucleosomes/genetics
15.
Genes Dev ; 27(9): 985-90, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23603901

ABSTRACT

Recent studies have identified a Lys 27-to-methionine (K27M) mutation at one allele of H3F3A, one of the two genes encoding histone H3 variant H3.3, in 60% of high-grade pediatric glioma cases. The median survival of this group of patients after diagnosis is ∼1 yr. Here we show that the levels of H3K27 di- and trimethylation (H3K27me2 and H3K27me3) are reduced globally in H3.3K27M patient samples due to the expression of the H3.3K27M mutant allele. Remarkably, we also observed that H3K27me3 and Ezh2 (the catalytic subunit of H3K27 methyltransferase) at chromatin are dramatically increased locally at hundreds of gene loci in H3.3K27M patient cells. Moreover, the gain of H3K27me3 and Ezh2 at gene promoters alters the expression of genes that are associated with various cancer pathways. These results indicate that H3.3K27M mutation reprograms epigenetic landscape and gene expression, which may drive tumorigenesis.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , Histones/genetics , Histones/metabolism , Mutation , Cell Line, Tumor , Genome, Human/genetics , Glioma/physiopathology , Humans , Methylation , Tumor Cells, Cultured
16.
Nucleic Acids Res ; 45(1): 169-180, 2017 01 09.
Article in English | MEDLINE | ID: mdl-27679476

ABSTRACT

DNA replication is tightly regulated to occur once and only once per cell cycle. How chromatin, the physiological substrate of DNA replication machinery, regulates DNA replication remains largely unknown. Here we show that histone H3 lysine 9 demethylase Kdm4d regulates DNA replication in eukaryotic cells. Depletion of Kdm4d results in defects in DNA replication, which can be rescued by the expression of H3K9M, a histone H3 mutant transgene that reverses the effect of Kdm4d on H3K9 methylation. Kdm4d interacts with replication proteins, and its recruitment to DNA replication origins depends on the two pre-replicative complex components (origin recognition complex [ORC] and minichromosome maintenance [MCM] complex). Depletion of Kdm4d impairs the recruitment of Cdc45, proliferating cell nuclear antigen (PCNA), and polymerase δ, but not ORC and MCM proteins. These results demonstrate a novel mechanism by which Kdm4d regulates DNA replication by reducing the H3K9me3 level to facilitate formation of pre-initiative complex.


Subject(s)
Chromatin/metabolism , DNA Replication , Histones/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Animals , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/chemistry , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , HCT116 Cells , HeLa Cells , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/metabolism , Lamin Type B/genetics , Lamin Type B/metabolism , Methylation , Mice , Minichromosome Maintenance Complex Component 4/genetics , Minichromosome Maintenance Complex Component 4/metabolism , NIH 3T3 Cells , Osteoblasts/cytology , Osteoblasts/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
17.
Nucleic Acids Res ; 45(7): 4142-4157, 2017 04 20.
Article in English | MEDLINE | ID: mdl-27998933

ABSTRACT

miRNAs play important roles during mammalian spermatogenesis. However, the function of most miRNAs in spermatogenesis and the underlying mechanisms remain unknown. Here, we report that miR-202 is highly expressed in mouse spermatogonial stem cells (SSCs), and is oppositely regulated by Glial cell-Derived Neurotrophic Factor (GDNF) and retinoic acid (RA), two key factors for SSC self-renewal and differentiation. We used inducible CRISPR-Cas9 to knockout miR-202 in cultured SSCs, and found that the knockout SSCs initiated premature differentiation accompanied by reduced stem cell activity and increased mitosis and apoptosis. Target genes were identified with iTRAQ-based proteomic analysis and RNA sequencing, and are enriched with cell cycle regulators and RNA-binding proteins. Rbfox2 and Cpeb1 were found to be direct targets of miR-202 and Rbfox2 but not Cpeb1, is essential for the differentiation of SSCs into meiotic cells. Accordingly, an SSC fate-regulatory network composed of signaling molecules of GDNF and RA, miR-202 and diverse downstream effectors has been identified.


Subject(s)
Adult Germline Stem Cells/metabolism , Cell Cycle/genetics , MicroRNAs/metabolism , RNA Splicing Factors/biosynthesis , Adult Germline Stem Cells/cytology , Animals , Gene Knockout Techniques , Male , Meiosis/genetics , Mice, Inbred C57BL , Mice, Inbred DBA , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Proteomics , Sequence Analysis, RNA , Spermatogenesis/genetics , Transcription Factors/biosynthesis , mRNA Cleavage and Polyadenylation Factors/biosynthesis
18.
Mol Cell Proteomics ; 12(5): 1144-57, 2013 May.
Article in English | MEDLINE | ID: mdl-23325766

ABSTRACT

Mammalian spermatogenesis consists of many cell types and biological processes and serves as an excellent model for studying gene regulation at transcriptional and post-transcriptional levels. Many key proteins, miRNAs, and perhaps piRNAs have been shown to be involved in post-transcriptional regulation of spermatogenesis. However, a systematic method for assessing the relationship between protein and mRNA expression has not been available for studying mechanisms of post-transcriptional regulation. In the present study, we used the iTRAQ-based quantitative proteomic approach to identify 2008 proteins in mouse type A spermatogonia, pachytene spermatocytes, round spermatids, and elongative spermatids with high confidence. Of these proteins, 1194 made up four dynamically changing clusters, which reflect the mitotic amplification, meiosis, and post-meiotic development of germ cells. We identified five major regulatory mechanisms termed "transcript only," "transcript degradation," "translation repression," "translation de-repression," and "protein degradation" based on changes in protein level relative to changes in mRNA level at the mitosis/meiosis transition and the meiosis/post-meiotic development transition. We found that post-transcriptional regulatory mechanisms are related to the generation of piRNAs and antisense transcripts. Our results provide a valuable inventory of proteins produced during mouse spermatogenesis and contribute to elucidating the mechanisms of the post-transcriptional regulation of gene expression in mammalian spermatogenesis.


Subject(s)
Proteome/metabolism , RNA Interference , Spermatogenesis/genetics , Transcriptome , Animals , Gene Expression Profiling , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Sequence Annotation , Oligonucleotide Array Sequence Analysis , Proteome/genetics , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spermatids/metabolism , Testis/cytology , Testis/metabolism
19.
Res Sq ; 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38352584

ABSTRACT

Background . Human hexokinase 2 ( HK2 ) plays an important role in regulating Warburg effect, which metabolizes glucose to lactate acid even in the presence of ample oxygen and provides intermediate metabolites to support cancer cell proliferation and tumor growth. HK2 overexpression has been observed in various types of cancers and targeting HK2 -driven Warburg effect has been suggested as a potential cancer therapeutic strategy. Given that epigenetic enzymes utilize metabolic intermediates as substrates or co-factors to carry out post-translational modification of DNA and histones in cells, we hypothesized that altering HK2 expression-mediated cellular glycolysis rates could impact the epigenome and, consequently, genome stability in yeast. To test this hypothesis, we established genetic models with different yeast hexokinase 2 ( HXK2) expression in Saccharomyces cerevisiae yeast cells and investigated the effect of HXK2 -dependent metabolism on parental nucleosome transfer, a key DNA replication-coupled epigenetic inheritance process, and chromatin stability. Results . By comparing the growth of mutant yeast cells carrying single deletion of hxk1Δ , hxk2Δ , or double-loss of hxk1Δ hxk2Δ to wild-type cells, we demonstrated that HXK2 is the dominant HXK in yeast cell growth. Surprisingly, manipulating HXK2 expression in yeast, whether through overexpression or deletion, had only a marginal impact on parental nucleosome assembly, but a noticeable trend with decrease chromatin instability. However, targeting yeast cells with 2-deoxy-D-glucose (2-DG), a HK2 inhibitor that has been proposed as an anti-cancer treatment, significantly increased chromatin instability. Conclusion . Our findings suggest that in yeast cells lacking HXK2 , alternative HXK s such as HXK1 or glucokinase 1 ( GLK1 ) play a role in supporting glycolysis at a level that adequately maintain epigenomic stability. While our study demonstrated an increase in epigenetic instability with 2-DG treatment, the observed effect seemed to occur independently of Hxk2-mediated glycolysis inhibition. Thus, additional research is needed to identify the molecular mechanism through which 2-DG influences chromatin stability.

20.
RNA ; 17(7): 1191-203, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21602304

ABSTRACT

PIWI-interacting RNAs (piRNAs) are a class of small RNAs abundantly expressed in animal gonads. piRNAs that map to retrotransposons are generated by a "ping-pong" amplification loop to suppress the activity of retrotransposons. However, the biogenesis and function of other categories of piRNAs have yet to be investigated. In this study, we first profiled the expression of small RNAs in type A spermatogonia, pachytene spermatocytes, and round spermatids by deep sequencing. We then focused on the computational analysis of the potential piRNAs generated in the present study as well as other published sets. piRNAs mapping to retrotransposons, mRNAs, and intergenic regions had different length distributions and were differentially regulated in spermatogenesis. piRNA-generating mRNAs (PRMRs), whose expression positively correlated with their piRNA products, constituted one-third of the protein-coding genes and were evolutionarily conserved and enriched with splicing isoforms and antisense transcripts. PRMRs with piRNAs preferentially mapped to CDSs and 3' UTRs partitioned into three clusters differentially expressed during spermatogenesis and enriched with unique sets of functional annotation terms related to housekeeping activities as well as spermatogenesis-specific processes. Intergenic piRNAs were divided into 2992 clusters probably representing novel transcriptional units that have not been reported. The transcripts of a large number of genes involved in spermatogenesis are the precursors of piRNAs, and these genes are intricately regulated by alternative splicing and antisense transcripts. piRNAs, whose regulatory role in gene expression awaits to be identified, are clearly products of a novel regulatory process that needs to be defined.


Subject(s)
RNA, Small Interfering/genetics , Spermatogenesis/genetics , Animals , Animals, Newborn , Base Sequence , Cells, Cultured , Cluster Analysis , Gene Expression Profiling , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Microarray Analysis , RNA, Small Interfering/metabolism , Spermatids/metabolism , Spermatocytes/metabolism , Spermatogenesis/physiology , Spermatogonia/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL