Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
FASEB J ; 38(9): e23633, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38690712

ABSTRACT

Recent reports suggest that the Hippo signaling pathway regulates testis development, though its exact roles in Sertoli cell differentiation remain unknown. Here, we examined the functions of the main Hippo pathway kinases, large tumor suppressor homolog kinases 1 and 2 (Lats1 and Lats2) in developing mouse Sertoli cells. Conditional inactivation of Lats1/2 in Sertoli cells resulted in the disorganization and overgrowth of the testis cords, the induction of a testicular inflammatory response and germ cell apoptosis. Stimulated by retinoic acid 8 (STRA8) expression in germ cells additionally suggested that germ cells may have been preparing to enter meiosis prior to their loss. Gene expression analyses of the developing testes of conditional knockout animals further suggested impaired Sertoli cell differentiation, epithelial-to-mesenchymal transition, and the induction of a specific set of genes associated with Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ)-mediated integrin signaling. Finally, the involvement of YAP/TAZ in Sertoli cell differentiation was confirmed by concomitantly inactivating Yap/Taz in Lats1/2 conditional knockout model, which resulted in a partial rescue of the testicular phenotypic changes. Taken together, these results identify Hippo signaling as a crucial pathway for Sertoli cell development and provide novel insight into Sertoli cell fate maintenance.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Differentiation , Protein Serine-Threonine Kinases , Sertoli Cells , Tumor Suppressor Proteins , YAP-Signaling Proteins , Animals , Sertoli Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Male , Mice , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Cell Differentiation/physiology , Mice, Knockout , Signal Transduction , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Testis/metabolism , Epithelial-Mesenchymal Transition/physiology , Transcription Factors/metabolism , Transcription Factors/genetics , Acyltransferases/genetics , Acyltransferases/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics
2.
Sci Adv ; 9(36): eadi2050, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37672574

ABSTRACT

The insulator model explains the workings of the H19 and Igf2 imprinted domain in the soma, where insulation of the Igf2 promoter from its enhancers occurs by CTCF in the maternally inherited unmethylated chromosome but not the paternally inherited methylated allele. The molecular mechanism that targets paternal methylation imprint establishment to the imprinting control region (ICR) in the male germline is unknown. We tested the function of prospermatogonia-specific broad low-level transcription in this process using mouse genetics. Paternal imprint establishment was abnormal when transcription was stopped at the entry point to the ICR. The germline epimutation persisted into the paternal allele of the soma, resulting in reduced Igf2 in fetal organs and reduced fetal growth, consistent with the insulator model and insulin-like growth factor 2 (IGF2)'s role as fetal growth factor. These results collectively support the role of broad low-level transcription through the H19/Igf2 ICR in the establishment of its paternal methylation imprint in the male germ line, with implications for Silver-Russell syndrome.


Subject(s)
Fetal Development , Protein Processing, Post-Translational , Animals , Mice , Methylation , Alleles , Phosphorylation
3.
Methods Mol Biol ; 2529: 229-251, 2022.
Article in English | MEDLINE | ID: mdl-35733018

ABSTRACT

Chromatin immunoprecipitation (ChIP) enables the study of DNA-protein interactions. When coupled with high-throughput sequencing (ChIP-seq), this method allows the generation of genome-wide profiles of the distribution of specific proteins in a given cellular context. Typical ChIP-seq experiments require millions of cells as input material and thus are not ideal to study many in vivo cell populations. Here, we describe an ultra-low-input native ChIP-seq method, ULI-NChIP-seq, to profile histone modification patterns in as low as 150 cells.


Subject(s)
High-Throughput Nucleotide Sequencing , Histones , Chromatin Immunoprecipitation/methods , High-Throughput Nucleotide Sequencing/methods , Histones/genetics , Histones/metabolism , Methylation , Protein Processing, Post-Translational , Sequence Analysis, DNA/methods
4.
Dev Cell ; 56(5): 671-686.e6, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33596408

ABSTRACT

A father's lifestyle impacts offspring health; yet, the underlying molecular mechanisms remain elusive. We hypothesized that a diet that changes methyl donor availability will alter the sperm and embryo epigenomes to impact embryonic gene expression and development. Here, we demonstrate that a folate-deficient (FD) diet alters histone H3 lysine 4 trimethylation (H3K4me3) in sperm at developmental genes and putative enhancers. A subset of H3K4me3 alterations in sperm are retained in the pre-implantation embryo and associated with deregulated embryonic gene expression. Using a genetic mouse model in which sires have pre-existing altered H3K4me2/3 in sperm, we show that a FD diet exacerbates alterations in sperm H3K4me3 and embryonic gene expression, leading to an increase in developmental defect severity. These findings imply that paternal H3K4me3 is transmitted to the embryo and influences gene expression and development. It further suggests that epigenetic errors can accumulate in sperm to worsen offspring developmental outcomes.


Subject(s)
Congenital Abnormalities/pathology , DNA Methylation , Diet , Embryo, Mammalian/cytology , Gene Expression Regulation, Developmental , Histones/chemistry , Spermatozoa/metabolism , Animals , Animals, Newborn , Chromatin/chemistry , Chromatin/genetics , Congenital Abnormalities/etiology , Congenital Abnormalities/metabolism , Embryo, Mammalian/metabolism , Epigenesis, Genetic , Female , Histones/genetics , Male , Mice , Mice, Inbred C57BL , Phenotype
5.
Nat Commun ; 12(1): 210, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431884

ABSTRACT

Histone acetylation is a ubiquitous hallmark of transcription, but whether the link between histone acetylation and transcription is causal or consequential has not been addressed. Using immunoblot and chromatin immunoprecipitation-sequencing in S. cerevisiae, here we show that the majority of histone acetylation is dependent on transcription. This dependency is partially explained by the requirement of RNA polymerase II (RNAPII) for the interaction of H4 histone acetyltransferases (HATs) with gene bodies. Our data also confirms the targeting of HATs by transcription activators, but interestingly, promoter-bound HATs are unable to acetylate histones in the absence of transcription. Indeed, HAT occupancy alone poorly predicts histone acetylation genome-wide, suggesting that HAT activity is regulated post-recruitment. Consistent with this, we show that histone acetylation increases at nucleosomes predicted to stall RNAPII, supporting the hypothesis that this modification is dependent on nucleosome disruption during transcription. Collectively, these data show that histone acetylation is a consequence of RNAPII promoting both the recruitment and activity of histone acetyltransferases.


Subject(s)
Genome, Fungal , Histones/metabolism , Saccharomyces cerevisiae/genetics , Transcription, Genetic , Acetylation , Animals , Chromatin/metabolism , Histone Acetyltransferases/metabolism , Mice , Trans-Activators/metabolism
6.
Nat Commun ; 11(1): 5417, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33110091

ABSTRACT

De novo DNA methylation (DNAme) during mammalian spermatogenesis yields a densely methylated genome, with the exception of CpG islands (CGIs), which are hypomethylated in sperm. While the paternal genome undergoes widespread DNAme loss before the first S-phase following fertilization, recent mass spectrometry analysis revealed that the zygotic paternal genome is paradoxically also subject to a low level of de novo DNAme. However, the loci involved, and impact on transcription were not addressed. Here, we employ allele-specific analysis of whole-genome bisulphite sequencing data and show that a number of genomic regions, including several dozen CGI promoters, are de novo methylated on the paternal genome by the 2-cell stage. A subset of these promoters maintains DNAme through development to the blastocyst stage. Consistent with paternal DNAme acquisition, many of these loci are hypermethylated in androgenetic blastocysts but hypomethylated in parthenogenetic blastocysts. Paternal DNAme acquisition is lost following maternal deletion of Dnmt3a, with a subset of promoters, which are normally transcribed from the paternal allele in blastocysts, being prematurely transcribed at the 4-cell stage in maternal Dnmt3a knockout embryos. These observations uncover a role for maternal DNMT3A activity in post-fertilization epigenetic reprogramming and transcriptional silencing of the paternal genome.


Subject(s)
Blastocyst/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Genome , Maternal Inheritance , Paternal Inheritance , Alleles , Animals , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , DNA Methyltransferase 3A , Epigenomics , Female , Gene Expression Regulation, Developmental , Male , Mice, Inbred DBA , Oocytes/metabolism , Spermatozoa/metabolism
7.
Nat Cell Biol ; 22(4): 355-357, 2020 04.
Article in English | MEDLINE | ID: mdl-32231308

Subject(s)
Chromatin , Oocytes , Histones , Zygote
8.
Science ; 367(6480): 870-874, 2020 02 21.
Article in English | MEDLINE | ID: mdl-32079766

ABSTRACT

Diapause is a state of suspended development that helps organisms survive extreme environments. How diapause protects living organisms is largely unknown. Using the African turquoise killifish (Nothobranchius furzeri), we show that diapause preserves complex organisms for extremely long periods of time without trade-offs for subsequent adult growth, fertility, and life span. Transcriptome analyses indicate that diapause is an active state, with dynamic regulation of metabolism and organ development genes. The most up-regulated genes in diapause include Polycomb complex members. The chromatin mark regulated by Polycomb, H3K27me3, is maintained at key developmental genes in diapause, and the Polycomb member CBX7 mediates repression of metabolism and muscle genes in diapause. CBX7 is functionally required for muscle preservation and diapause maintenance. Thus, vertebrate diapause is a state of suspended life that is actively maintained by specific chromatin regulators, and this has implications for long-term organism preservation.


Subject(s)
Diapause/physiology , Killifishes/growth & development , Muscle, Skeletal/growth & development , Polycomb Repressive Complex 1/metabolism , Animals , Diapause/genetics , Gene Expression Regulation, Developmental , Histones/metabolism , Mutation , Polycomb Repressive Complex 1/genetics
9.
Nat Commun ; 10(1): 5674, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31831741

ABSTRACT

Imprinted genes are expressed from a single parental allele, with the other allele often silenced by DNA methylation (DNAme) established in the germline. While species-specific imprinted orthologues have been documented, the molecular mechanisms underlying the evolutionary switch from biallelic to imprinted expression are unknown. During mouse oogenesis, gametic differentially methylated regions (gDMRs) acquire DNAme in a transcription-guided manner. Here we show that oocyte transcription initiating in lineage-specific endogenous retroviruses (ERVs) is likely responsible for DNAme establishment at 4/6 mouse-specific and 17/110 human-specific imprinted gDMRs. The latter are divided into Catarrhini- or Hominoidea-specific gDMRs embedded within transcripts initiating in ERVs specific to these primate lineages. Strikingly, imprinting of the maternally methylated genes Impact and Slc38a4 was lost in the offspring of female mice harboring deletions of the relevant murine-specific ERVs upstream of these genes. Our work reveals an evolutionary mechanism whereby maternally silenced genes arise from biallelically expressed progenitors.


Subject(s)
DNA Methylation , Evolution, Molecular , Genomic Imprinting , Promoter Regions, Genetic/genetics , Retroviridae/genetics , Animals , Epigenomics , Female , Germ Cells , Haplorhini , Humans , Macaca , Male , Mice , Oocytes/metabolism , Pan troglodytes , Primates , Species Specificity , Terminal Repeat Sequences
10.
Elife ; 82019 05 31.
Article in English | MEDLINE | ID: mdl-31148541

ABSTRACT

Hundreds of retrovirus-like sequences have features that suggest they might be gene enhancers, but only a small fraction displays gene-regulating activity in experiments on mouse stem cells.


Subject(s)
DNA Transposable Elements/genetics , Enhancer Elements, Genetic , Animals , Endogenous Retroviruses/genetics , Humans , Mice , Models, Genetic , Reproducibility of Results
11.
Nat Genet ; 51(5): 844-856, 2019 05.
Article in English | MEDLINE | ID: mdl-31040401

ABSTRACT

The oocyte epigenome plays critical roles in mammalian gametogenesis and embryogenesis. Yet, how it is established remains elusive. Here, we report that histone-lysine N-methyltransferase SETD2, an H3K36me3 methyltransferase, is a crucial regulator of the mouse oocyte epigenome. Deficiency in Setd2 leads to extensive alterations of the oocyte epigenome, including the loss of H3K36me3, failure in establishing the correct DNA methylome, invasion of H3K4me3 and H3K27me3 into former H3K36me3 territories and aberrant acquisition of H3K4me3 at imprinting control regions instead of DNA methylation. Importantly, maternal depletion of SETD2 results in oocyte maturation defects and subsequent one-cell arrest after fertilization. The preimplantation arrest is mainly due to a maternal cytosolic defect, since it can be largely rescued by normal oocyte cytosol. However, chromatin defects, including aberrant imprinting, persist in these embryos, leading to embryonic lethality after implantation. Thus, these data identify SETD2 as a crucial player in establishing the maternal epigenome that in turn controls embryonic development.


Subject(s)
Embryonic Development/genetics , Epigenesis, Genetic , Genomic Imprinting , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Animals , DNA (Cytosine-5-)-Methyltransferases/deficiency , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Female , Histone Code/genetics , Histone-Lysine N-Methyltransferase/deficiency , Histones/metabolism , Male , Mice , Mice, Knockout , Models, Genetic , Oocytes/metabolism , Oogenesis/genetics , Pregnancy
12.
Cell Rep ; 27(1): 282-293.e4, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30943408

ABSTRACT

Mammalian histone methyltransferase G9a (also called EHMT2) deposits H3K9me2 on chromatin and is essential for postimplantation development. However, its role in oogenesis and preimplantation development remains poorly understood. We show that H3K9me2-enriched chromatin domains in mouse oocytes are generally depleted of CG methylation, contrasting with their association in embryonic stem and somatic cells. Oocyte-specific disruption of G9a results in reduced H3K9me2 enrichment and impaired reorganization of heterochromatin in oocytes, but only a modest reduction in CG methylation is detected. Furthermore, in both oocytes and 2-cell embryos, G9a depletion has limited impact on the expression of genes and retrotransposons. Although their CG methylation is minimally affected, preimplantation embryos derived from such oocytes show abnormal chromosome segregation and frequent developmental arrest. Our findings illuminate the functional importance of G9a independent of CG methylation in preimplantation development and call into question the proposed role for H3K9me2 in CG methylation protection in zygotes.


Subject(s)
Blastocyst/metabolism , DNA Methylation , Histone Code , Histone-Lysine N-Methyltransferase/metabolism , Oocytes/metabolism , Animals , Chromosome Segregation , Female , Histone-Lysine N-Methyltransferase/genetics , Histones/chemistry , Histones/metabolism , Mice , Mice, Inbred C57BL
13.
Nat Commun ; 9(1): 3331, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30127397

ABSTRACT

De novo DNA methylation (DNAme) during mouse oogenesis occurs within transcribed regions enriched for H3K36me3. As many oocyte transcripts originate in long terminal repeats (LTRs), which are heterogeneous even between closely related mammals, we examined whether species-specific LTR-initiated transcription units (LITs) shape the oocyte methylome. Here we identify thousands of syntenic regions in mouse, rat, and human that show divergent DNAme associated with private LITs, many of which initiate in lineage-specific LTR retrotransposons. Furthermore, CpG island (CGI) promoters methylated in mouse and/or rat, but not human oocytes, are embedded within rodent-specific LITs and vice versa. Notably, at a subset of such CGI promoters, DNAme persists on the maternal genome in fertilized and parthenogenetic mouse blastocysts or in human placenta, indicative of species-specific epigenetic inheritance. Polymorphic LITs are also responsible for disparate DNAme at promoter CGIs in distantly related mouse strains, revealing that LITs also promote intra-species divergence in CGI DNAme.


Subject(s)
DNA Methylation/genetics , Inheritance Patterns/genetics , Oocytes/metabolism , Retroelements/genetics , Terminal Repeat Sequences/genetics , Transcription, Genetic , Animals , CpG Islands/genetics , DNA, Intergenic/genetics , Fertilization/genetics , Gene Expression Regulation , Humans , Mammals/metabolism , Mice, Inbred C57BL , Polymorphism, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Species Specificity , Synteny/genetics
14.
Genetics ; 205(3): 1113-1123, 2017 03.
Article in English | MEDLINE | ID: mdl-28108585

ABSTRACT

Histone post-translational modifications (PTMs) alter chromatin structure by promoting the interaction of chromatin-modifying complexes with nucleosomes. The majority of chromatin-modifying complexes contain multiple domains that preferentially interact with modified histones, leading to speculation that these domains function in concert to target nucleosomes with distinct combinations of histone PTMs. In Saccharomyces cerevisiae, the NuA3 histone acetyltransferase complex contains three domains, the PHD finger in Yng1, the PWWP domain in Pdp3, and the YEATS domain in Taf14; which in vitro bind to H3K4 methylation, H3K36 methylation, and acetylated and crotonylated H3K9, respectively. While the in vitro binding has been well characterized, the relative in vivo contributions of these histone PTMs in targeting NuA3 is unknown. Here, through genome-wide colocalization and by mutational interrogation, we demonstrate that the PHD finger of Yng1, and the PWWP domain of Pdp3 independently target NuA3 to H3K4 and H3K36 methylated chromatin, respectively. In contrast, we find no evidence to support the YEATS domain of Taf14 functioning in NuA3 recruitment. Collectively our results suggest that the presence of multiple histone PTM binding domains within NuA3, rather than restricting it to nucleosomes containing distinct combinations of histone PTMs, can serve to increase the range of nucleosomes bound by the complex. Interestingly, however, the simple presence of NuA3 is insufficient to ensure acetylation of the associated nucleosomes, suggesting a secondary level of acetylation regulation that does not involve control of HAT-nucleosome interactions.


Subject(s)
Histone Acetyltransferases/metabolism , Histones/metabolism , Protein Processing, Post-Translational , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Acetylation , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/genetics , Histones/genetics , Methylation , Nucleosomes/genetics , Nucleosomes/metabolism , Protein Domains , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
15.
Cell Stem Cell ; 19(1): 81-94, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27151458

ABSTRACT

Repression of endogenous retroviruses (ERVs) in mammals involves several epigenetic mechanisms. Acute loss of the maintenance methyltransferase Dnmt1 induces widespread DNA demethylation and transcriptional activation of ERVs, including CpG-rich IAP (intracisternal A particle) proviruses. Here, we show that this effect is not due simply to a loss of DNA methylation. Conditional deletions reveal that both Dnmt1 and Np95 are essential for maintenance DNA methylation. However, while IAPs are derepressed in Dnmt1-ablated embryos and embryonic stem cells (ESCs), these ERVs remain silenced when Np95 is deleted alone or in combination with Dnmt1. This paradoxical phenotype results from an ectopic interaction between NP95 and the H3K9 methyltransferase SETDB1. Normally, SETDB1 maintains silencing of IAPs, but in the absence of DNMT1, prolonged binding of NP95 to hemimethylated DNA transiently disrupts SETDB1-dependent H3K9me3 deposition. Thus, our observations reveal an unexpected antagonistic interplay between two repressive pathways involved in retroviral silencing in mammalian cells.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA/metabolism , Endogenous Retroviruses/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Mouse Embryonic Stem Cells/metabolism , Nuclear Proteins/metabolism , Virus Activation , Animals , CCAAT-Enhancer-Binding Proteins , Cell Differentiation , DNA (Cytosine-5-)-Methyltransferase 1 , Female , Gene Dosage , Gene Expression Regulation, Developmental , Gene Silencing , Genes, Intracisternal A-Particle , Genetic Loci , Histones/metabolism , Lysine/metabolism , Mice , Mice, Knockout , Models, Biological , Mutation/genetics , Nuclear Proteins/chemistry , Placenta/metabolism , Pregnancy , Protein Binding , Protein Domains , Trophoblasts/metabolism , Ubiquitin-Protein Ligases
16.
Nat Commun ; 6: 6033, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25607992

ABSTRACT

Combined chromatin immunoprecipitation and next-generation sequencing (ChIP-seq) has enabled genome-wide epigenetic profiling of numerous cell lines and tissue types. A major limitation of ChIP-seq, however, is the large number of cells required to generate high-quality data sets, precluding the study of rare cell populations. Here, we present an ultra-low-input micrococcal nuclease-based native ChIP (ULI-NChIP) and sequencing method to generate genome-wide histone mark profiles with high resolution from as few as 10(3) cells. We demonstrate that ULI-NChIP-seq generates high-quality maps of covalent histone marks from 10(3) to 10(6) embryonic stem cells. Subsequently, we show that ULI-NChIP-seq H3K27me3 profiles generated from E13.5 primordial germ cells isolated from single male and female embryos show high similarity to recent data sets generated using 50-180 × more material. Finally, we identify sexually dimorphic H3K27me3 enrichment at specific genic promoters, thereby illustrating the utility of this method for generating high-quality and -complexity libraries from rare cell populations.


Subject(s)
Chromatin Immunoprecipitation/methods , Animals , Artifacts , Cell Separation , Embryonic Stem Cells/metabolism , Female , Flow Cytometry , Genetic Association Studies , Genome , High-Throughput Nucleotide Sequencing , Histones/chemistry , Histones/metabolism , Male , Mice , Micrococcal Nuclease/metabolism , Oligonucleotide Array Sequence Analysis/methods , Polymerase Chain Reaction , Sequence Analysis, DNA/methods , Sex Factors
17.
Genes Dev ; 28(18): 2041-55, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25228647

ABSTRACT

Transcription of endogenous retroviruses (ERVs) is inhibited by de novo DNA methylation during gametogenesis, a process initiated after birth in oocytes and at approximately embryonic day 15.5 (E15.5) in prospermatogonia. Earlier in germline development, the genome, including most retrotransposons, is progressively demethylated. Young ERVK and ERV1 elements, however, retain intermediate methylation levels. As DNA methylation reaches a low point in E13.5 primordial germ cells (PGCs) of both sexes, we determined whether retrotransposons are marked by H3K9me3 and H3K27me3 using a recently developed low-input ChIP-seq (chromatin immunoprecipitation [ChIP] combined with deep sequencing) method. Although these repressive histone modifications are found predominantly on distinct genomic regions in E13.5 PGCs, they concurrently mark partially methylated long terminal repeats (LTRs) and LINE1 elements. Germline-specific conditional knockout of the H3K9 methyltransferase SETDB1 yields a decrease of both marks and DNA methylation at H3K9me3-enriched retrotransposon families. Strikingly, Setdb1 knockout E13.5 PGCs show concomitant derepression of many marked ERVs, including intracisternal A particle (IAP), ETn, and ERVK10C elements, and ERV-proximal genes, a subset in a sex-dependent manner. Furthermore, Setdb1 deficiency is associated with a reduced number of male E13.5 PGCs and postnatal hypogonadism in both sexes. Taken together, these observations reveal that SETDB1 is an essential guardian against proviral expression prior to the onset of de novo DNA methylation in the germline.


Subject(s)
DNA Methylation , Endogenous Retroviruses/metabolism , Germ Cells/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Animals , Chromatin Immunoprecipitation , Endogenous Retroviruses/genetics , Female , Gametogenesis/genetics , Gene Deletion , Gene Knockout Techniques , Gene Silencing , Germ Cells/virology , Histone-Lysine N-Methyltransferase/genetics , Male , Mice , Transcription, Genetic , Virus Activation/genetics
18.
Proc Natl Acad Sci U S A ; 110(5): 1658-63, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23319653

ABSTRACT

Among the earliest responses of mammalian cells to DNA damage is catalytic activation of a nuclear enzyme poly(ADP-ribose) polymerase-1 (PARP-1). Activated PARP-1 forms the polymers of ADP-ribose (pADPr or PAR) that posttranslationally modify its target proteins, such as PARP-1 and DNA repair-related proteins. Although this metabolism is known to be implicated in other repair pathways, here we show its role in the versatile nucleotide excision repair pathway (NER) that removes a variety of DNA damages including those induced by UV. We show that PARP inhibition or specific depletion of PARP-1 decreases the efficiency of removal of UV-induced DNA damage from human skin fibroblasts or mouse epidermis. Using NER-proficient and -deficient cells and in vitro PARP-1 assays, we show that damaged DNA-binding protein 2 (DDB2), a key lesion recognition protein of the global genomic subpathway of NER (GG-NER), associates with PARP-1 in the vicinity of UV-damaged chromatin, stimulates its catalytic activity, and is modified by pADPr. PARP inhibition abolishes UV-induced interaction of DDB2 with PARP-1 or xeroderma pigmentosum group C (XPC) and also decreases localization of XPC to UV-damaged DNA, which is a key step that leads to downstream events in GG-NER. Thus, PARP-1 collaborates with DDB2 to increase the efficiency of the lesion recognition step of GG-NER.


Subject(s)
DNA Damage , DNA Repair , DNA/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Animals , Cell Line, Transformed , DNA/genetics , DNA-Binding Proteins/metabolism , Epidermis/drug effects , Epidermis/metabolism , Epidermis/radiation effects , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Immunoblotting , Mice , Mice, Hairless , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/genetics , Protein Binding/drug effects , Protein Binding/radiation effects , RNA Interference , Time Factors , Ultraviolet Rays
19.
Proc Natl Acad Sci U S A ; 109(45): 18505-10, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23091032

ABSTRACT

Histone H3 lysine 4 trimethylation (H3K4me3) is a hallmark of transcription initiation, but how H3K4me3 is demethylated during gene repression is poorly understood. Jhd2, a JmjC domain protein, was recently identified as the major H3K4me3 histone demethylase (HDM) in Saccharomyces cerevisiae. Although JHD2 is required for removal of methylation upon gene repression, deletion of JHD2 does not result in increased levels of H3K4me3 in bulk histones, indicating that this HDM is unable to demethylate histones during steady-state conditions. In this study, we showed that this was due to the negative regulation of Jhd2 activity by histone H3 lysine 14 acetylation (H3K14ac), which colocalizes with H3K4me3 across the yeast genome. We demonstrated that loss of the histone H3-specific acetyltransferases (HATs) resulted in genome-wide depletion of H3K4me3, and this was not due to a transcription defect. Moreover, H3K4me3 levels were reestablished in HAT mutants following loss of JHD2, which suggested that H3-specific HATs and Jhd2 serve opposing functions in regulating H3K4me3 levels. We revealed the molecular basis for this suppression by demonstrating that H3K14ac negatively regulated Jhd2 demethylase activity on an acetylated peptide in vitro. These results revealed the existence of a general mechanism for removal of H3K4me3 following gene repression.


Subject(s)
Histones/metabolism , Lysine/metabolism , Saccharomyces cerevisiae/metabolism , Acetylation , Gene Expression Regulation, Fungal , Histone Acetyltransferases/metabolism , Jumonji Domain-Containing Histone Demethylases , Methylation , Models, Biological , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins , Substrate Specificity
20.
Ann N Y Acad Sci ; 1266: 1-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22901250

ABSTRACT

Semi-conservative replication ensures that the DNA sequence of sister chromatids is identical except for replication errors and variation in the length of telomere repeats resulting from replicative losses and variable end processing. What happens with the various epigenetic marks during DNA replication is less clear. Many chromatin marks are likely to be copied onto both sister chromatids in conjunction with DNA replication, whereas others could be distributed randomly between sister chromatids. Epigenetic differences between sister chromatids could also emerge in a more predictable manner, for example, following processes that are associated with lagging strand DNA replication. The resulting epigenetic differences between sister chromatids could result in different gene expression patterns in daughter cells. This possibility has been difficult to test because techniques to distinguish between parental sister chromatids require analysis of single cells and are not obvious. Here, we briefly review the topic of sister chromatid epigenetics and discuss how the identification of sister chromatids in cells could change the way we think about asymmetric cell divisions and stochastic variation in gene expression between cells in general and paired daughter cells in particular.


Subject(s)
Chromatids/genetics , Epigenesis, Genetic , Animals , Cell Division/genetics , DNA Methylation/genetics , DNA Replication/genetics , DNA, Satellite/genetics , Gene Expression , In Situ Hybridization, Fluorescence , Mice , Models, Genetic , Stochastic Processes
SELECTION OF CITATIONS
SEARCH DETAIL
...